Institute for Health and Consumer Protection
Physical and Chemical Exposure Unit
I-21020 Ispra (VA), Italy
Final Report
The INDEX project
Critical Appraisal of the Setting and Implementation of Indoor
Exposure Limits in the EU
Dimitrios Kotzias, Kimmo Koistinen, Stylianos Kephalopoulos,
Christian Schlitt, Paolo Carrer, Marco Maroni,
Matti Jantunen, Christian Cochet, Séverine Kirchner,
Thomas Lindvall, James McLaughlin, Lars Mølhave,
Eduardo de Oliveira Fernandes and Bernd Seifert
2005 EUR 21590 EN
European Commission
Directorate-General Joint Research Centre
http://www.jrc.cec.eu.int
Legal Notice
Neither the European Commission nor any person acting on behalf of the
Commission is responsible for the use which might be made of this publication.
A great deal of additional information on the European Union is available on the Internet.
It can be accessed through the Europa server
http://europa.eu
EUR 21590EN
ISSN 1018-5593
Luxembourg: Office for Official Publications of the European Communities
© European Communities, 2005
Reproduction is authorised provided the source is acknowledged
Printed in Italy
The INDEX project Final report
2
Legal notice
Neither the European Commission nor any person acting on behalf of the Commission is responsible for the use which
might be made of the following information.
Foreword
In the past decades a large number of studies have indicated the presence of different compounds belonging to a variety
of chemical classes in indoor environments (buildings, homes). The presence of these chemicals in indoor air is the
result of infiltration of polluted outdoor air and of emissions from various indoor sources, including building materials,
activities of the occupants, consumer products, smoking etc.
For many of these chemicals, the risk to human health and comfort is almost totally unknown and difficult to predict
because of lack of toxicological data and information on the dose-response characteristics in humans or animal models.
On the other hand, a full toxicological testing as requested by the “existing chemicals” legislation is difficult to
accomplish for these compounds, because it would involve investigation of acute and subacute toxicity, mutagenicity,
carcinogenicity and reproductive toxicity according to testing protocols that are complex, time-consuming and
expensive. Moreover, the EU policy on limitation of unnecessary animal testing further limits the possibility of
advocating a generalized animal testing of these chemicals.
The result of this situation is that there is an objective difficulty in regulating the presence of these substances in indoor
air principally because of the absence of adequate hazard and risk assessment. There is therefore an urgent need to
develop a strategy for the identification of priorities in testing, assessment and regulation.
In the frame of the INDEX project the existing knowledge worldwide has been assessed on
- type and levels of chemicals in indoor air and
- available toxicological information to allow the assessment of risk to health and comfort.
The collection and evaluation of the aforementioned information within the frame of the INDEX project shall contribute
to develop a strategy for prioritization in assessment and regulation of chemicals in indoor environments.
The Steering Committee
The INDEX project Final report
3
Organisation responsible of the project: European Commission, Joint Research Centre, Institute for Health and
Consumer Protection, Physical and Chemical Exposure Unit, Ispra, Italy (JRC/IHCP/PCE).
Project leader: Dr. Dimtrios Kotzias, JRC/IHCP/PCE
Sources of funding: DG Consumer Protection (SANCO) and JRC/IHCP/PCE
Sub-contractors: University of Milan, Department of Occupational Health, Unit Ospedale Luigi Sacco, Milan, Italy
National Public Health Institute (KTL), Kuopio, Finland
Preparation of documents: All working drafts and this final report were prepared by the Joint Research Centre
(Institute for Health and Consumer Protection, Physical and Chemical Exposure Unit, JRC), the University of Milan
(Department of Occupational Health, Unit Ospedale Luigi Sacco, UNIMI), the National Public Health Institute
(Department of Environmental Hygiene, Kuopio, Finland, KTL) and with the individual contributions and advice in the
course of the work from members of the Steering Committee.
Final drafting:
Hazard Identification / Exposure Assessment:
Dr. Kimmo Koistinen and Dr. Dimitrios Kotzias (JRC)
Dose/Response Assessment / Risk Characterization:
Christian Schlitt, Dr. Paolo Carrer and Prof. Marco Maroni (UNIMI)
Risk Management:
Prof. Matti J. Jantunen (KTL)
Date of last literature search: September 2004
Steering Committee Members:
Dr. Christian Cochet Centre Scientifique et Technique du Bâtiment, Division Santé–Bâtiment,
Marne la Vallée, France
Prof. Matti J. Jantunen KTL, Department of Environmental Hygiene, Kuopio, Finland
Dr. Stylianos Kephalopoulos European Commission, Joint Research Centre, Institute for Health and
Consumer Protection, Physical and Chemical Exposure Unit, Ispra, Italy
Dr. Séverine Kirchner Centre Scientifique et Technique du Bâtiment, Division Santé–Bâtiment,
Marne la Vallée, France
Prof. Thomas Lindvall Karolinska Institute, Institute of Environmental Medicine, Stockholm,
Sweden
Prof. Marco Maroni Università di Milano, Department of Occupational Health, Unit Ospedale
Luigi Sacco, Milan, Italy
Dr. James P. McLaughlin University College Dublin, Department of Physics, Dublin, Ireland
Prof. Lars Mølhave Aarhus Universitet, Institut for Miljø– og Arbejdsmedicin, Århus, Denmark
Prof. Eduardo de Oliveira Fernandes Universidade do Porto, Departamento de Engenharia Mecânica, Faculdade
de Engenharia, Porto, Portugal
Prof. Bernd Seifert Umweltbundesamt, Abteilung Umwelthygiene, Berlin, Germany
The INDEX project Final report
4
Contents
Foreword ........................................................................................................................................................................................................................... 2
Contents ........................................................................................................................................................................................................................... 4
Executive Summary ....................................................................................................................................................... 9
Introduction and objectives.......................................................................................................................................................................................... 9
Methodology................................................................................................................................................................................................................ 9
Risk assessment of the selected compounds ............................................................................................................................................................. 10
Risk management tools.............................................................................................................................................................................................. 14
Recommendations and management options ............................................................................................................................................................ 14
1. Introduction .............................................................................................................................................................. 17
2. Objectives ................................................................................................................................................................ 18
3. Methodology ............................................................................................................................................................. 19
3.1 Risk Assessment ....................................................................................................................................................................................................... 19
3.1.1 Selection of Indoor Air Chemicals for Consideration (Hazard Identification) ............................................................................................. 19
3.1.2 Exposure Assessment ..................................................................................................................................................................................... 22
3.1.3 Dose/Response Assessment............................................................................................................................................................................ 22
3.1.4 Risk Characterization and prioritisation of chemicals ................................................................................................................................... 26
3.2 Risk Management ..................................................................................................................................................................................................... 27
4. Risk Assessment of the selected chemicals ............................................................................................................. 29
Formaldehyde............................................................................................................................................................... 30
1. Compound Identification............................................................................................................................................................................................ 30
2. Physical and Chemical Properties .............................................................................................................................................................................. 30
3. Indoor Air Exposure Assessment ............................................................................................................................................................................... 30
Emission sources........................................................................................................................................................................................................ 30
Indoor air and exposure concentrations..................................................................................................................................................................... 31
4. Toxicokinetics............................................................................................................................................................................................................. 35
Absorption.................................................................................................................................................................................................................. 35
Distribution ................................................................................................................................................................................................................ 35
Metabolism and elimination ...................................................................................................................................................................................... 35
5. Health effects .............................................................................................................................................................................................................. 35
Effects of short-term exposure................................................................................................................................................................................... 35
Effects of long-term exposure (noncancer) ............................................................................................................................................................... 39
Carcinogenic effects .................................................................................................................................................................................................. 43
Interactions with other chemicals .............................................................................................................................................................................. 46
Odour perception ....................................................................................................................................................................................................... 47
Summary of Formaldehyde Dose Response Assessment ......................................................................................................................................... 47
6. Risk Characterization.................................................................................................................................................................................................. 49
Health hazard evaluation ........................................................................................................................................................................................... 49
Percentage of population exposed beyond given threshold levels............................................................................................................................ 49
Cancer risk evaluation ............................................................................................................................................................................................... 49
Comments .................................................................................................................................................................................................................. 49
Result ......................................................................................................................................................................................................................... 50
Carbon monoxide ......................................................................................................................................................... 51
1. Compound identification ............................................................................................................................................................................................ 51
2. Physical and Chemical properties............................................................................................................................................................................... 52
3. Indoor Air Exposure assessment ................................................................................................................................................................................ 52
Indoor air and exposure concentrations..................................................................................................................................................................... 52
Emission sources........................................................................................................................................................................................................ 54
4. Toxicokinetics............................................................................................................................................................................................................. 59
Endogenous sources of carbon monoxide:................................................................................................................................................................ 59
Absorption.................................................................................................................................................................................................................. 59
Elimination................................................................................................................................................................................................................. 61
5. Health effects .............................................................................................................................................................................................................. 61
Effects of short-term exposure................................................................................................................................................................................... 61
Effects of long-term exposure ................................................................................................................................................................................... 68
Carcinogenic and genotoxic effects........................................................................................................................................................................... 70
Summary of Carbon Monoxide Dose Response Assessment ................................................................................................................................... 70
6. Risk Characterization.................................................................................................................................................................................................. 71
Health hazard evaluation ........................................................................................................................................................................................... 71
Percentage of population exposed beyond given threshold CO levels ..................................................................................................................... 72
Comments .................................................................................................................................................................................................................. 72
Result ......................................................................................................................................................................................................................... 74
Nitrogen dioxide ........................................................................................................................................................... 75
1. Compound identification ............................................................................................................................................................................................ 75
2. Physical and Chemical properties............................................................................................................................................................................... 75
3. Indoor Air Exposure assessment ................................................................................................................................................................................ 75
Emission sources........................................................................................................................................................................................................ 75
Indoor air and exposure concentrations..................................................................................................................................................................... 76
4. Toxicokinetics............................................................................................................................................................................................................. 79
Absorption.................................................................................................................................................................................................................. 79
Distribution ................................................................................................................................................................................................................ 79
The INDEX project Final report
5
Elimination................................................................................................................................................................................................................. 79
5. Health effects .............................................................................................................................................................................................................. 79
Effects of short-term exposure................................................................................................................................................................................... 79
Effects of long-term exposure ................................................................................................................................................................................... 84
Carcinogenic and mutagenic effects.......................................................................................................................................................................... 87
Interaction with other chemicals................................................................................................................................................................................ 87
Odour perception ....................................................................................................................................................................................................... 88
Summary of Nitrogen dioxide Dose Response Assessment ..................................................................................................................................... 89
6. Risk characterization................................................................................................................................................................................................... 90
Health hazard evaluation of short- and long-term exposure ..................................................................................................................................... 90
Exposure data of relevance for the European population ......................................................................................................................................... 90
Result ......................................................................................................................................................................................................................... 92
Benzene ................................................................................................................................................................ 93
1. Compound identification ............................................................................................................................................................................................ 93
2. Physical and Chemical properties............................................................................................................................................................................... 93
3. Indoor Air Exposure assessment ................................................................................................................................................................................ 93
Emission sources........................................................................................................................................................................................................ 93
Indoor air and exposure concentrations..................................................................................................................................................................... 94
4. Toxicokinetics............................................................................................................................................................................................................. 97
Absorption.................................................................................................................................................................................................................. 97
Distribution ................................................................................................................................................................................................................ 97
Metabolism and elimination ...................................................................................................................................................................................... 97
5. Health effects .............................................................................................................................................................................................................. 98
Effects of short-term exposure................................................................................................................................................................................... 98
Effects of long-term exposure (noncancer) ............................................................................................................................................................. 103
Genotoxic effects ..................................................................................................................................................................................................... 107
Carcinogenic effects ................................................................................................................................................................................................ 108
Susceptible population............................................................................................................................................................................................. 112
Interactions with other chemicals ............................................................................................................................................................................ 113
Odour perception ..................................................................................................................................................................................................... 113
Summary of Benzene Dose Response Assessment................................................................................................................................................. 114
6. Risk Characterization................................................................................................................................................................................................ 116
Cancer risk evaluation ............................................................................................................................................................................................. 116
Population cancer risk estimates of benzene-induced leukemia ............................................................................................................................. 116
Health hazard evaluation (noncancer) ..................................................................................................................................................................... 118
Comments ................................................................................................................................................................................................................ 119
Result ....................................................................................................................................................................................................................... 120
Naphthalene .............................................................................................................................................................. 121
1. Compound identification .......................................................................................................................................................................................... 121
2. Physical and Chemical properties............................................................................................................................................................................. 121
3. Indoor Air Exposure assessment .............................................................................................................................................................................. 121
Indoor air and exposure concentrations................................................................................................................................................................... 121
4. Toxicokinetics........................................................................................................................................................................................................... 124
Absorption................................................................................................................................................................................................................ 124
Distribution .............................................................................................................................................................................................................. 124
Metabolism and elimination .................................................................................................................................................................................... 124
5. Health effects ............................................................................................................................................................................................................ 125
Effects of short-term exposure................................................................................................................................................................................. 125
Effects of long-term exposure (noncancer) ............................................................................................................................................................. 127
Genotoxicity............................................................................................................................................................................................................. 133
Carcinogenic potential ............................................................................................................................................................................................. 134
Interactions with other chemicals ............................................................................................................................................................................ 134
Odour perception ..................................................................................................................................................................................................... 134
Summary of Naphthalene Dose Response Assessment .......................................................................................................................................... 135
6. Risk Characterization................................................................................................................................................................................................ 136
Health hazard evaluation of short-term exposure.................................................................................................................................................... 136
Health hazard and cancer risk evaluation of long-term exposure........................................................................................................................... 136
Result ....................................................................................................................................................................................................................... 136
Relevance of EU-population exposure to naphthalene ........................................................................................................................................... 137
Acetaldehyde .............................................................................................................................................................. 138
1. Compound identification .......................................................................................................................................................................................... 138
2. Physical and Chemical properties............................................................................................................................................................................. 138
3. Indoor Air Exposure assessment .............................................................................................................................................................................. 138
Contribution of inhalation exposure to total exposure............................................................................................................................................ 138
Emission sources...................................................................................................................................................................................................... 139
Indoor air and exposure concentrations................................................................................................................................................................... 139
4. Toxicokinetics........................................................................................................................................................................................................... 141
Absorption................................................................................................................................................................................................................ 141
Distribution .............................................................................................................................................................................................................. 141
Metabolism and elimination .................................................................................................................................................................................... 141
5. Health effects ............................................................................................................................................................................................................ 142
Effects of short-term exposure................................................................................................................................................................................. 142
Effects of long-term exposure ................................................................................................................................................................................. 143
Carcinogenic potential ............................................................................................................................................................................................. 148
Genotoxicity............................................................................................................................................................................................................. 148
Interactions with other chemicals ............................................................................................................................................................................ 149
The INDEX project Final report
6
Odour perception ..................................................................................................................................................................................................... 149
Summary of Acetaldehyde Dose Response Assessment......................................................................................................................................... 151
6. Risk Characterization................................................................................................................................................................................................ 152
Cancer risk and health hazard evaluation................................................................................................................................................................ 152
Percentage of population exposed beyond given threshold levels.......................................................................................................................... 152
Comments ................................................................................................................................................................................................................ 152
Result ....................................................................................................................................................................................................................... 153
Toluene .............................................................................................................................................................. 154
1. Compound identification .......................................................................................................................................................................................... 154
2. Physical and Chemical properties............................................................................................................................................................................. 154
3. Indoor Air Exposure assessment .............................................................................................................................................................................. 154
Emission sources...................................................................................................................................................................................................... 154
Indoor air and exposure concentrations................................................................................................................................................................... 154
4. Toxicokinetics........................................................................................................................................................................................................... 158
Absorption................................................................................................................................................................................................................ 158
Distribution .............................................................................................................................................................................................................. 158
Metabolism and elimination .................................................................................................................................................................................... 159
5. Health effects ............................................................................................................................................................................................................ 160
Effects of short-term exposure................................................................................................................................................................................. 160
Effects of long-term exposure ................................................................................................................................................................................. 162
Carcinogenic potential ............................................................................................................................................................................................. 168
Genotoxicity............................................................................................................................................................................................................. 168
Interactions with other chemicals ............................................................................................................................................................................ 168
Odour perception ..................................................................................................................................................................................................... 169
Summary of Toluene Dose Response Assessment.................................................................................................................................................. 170
6. Risk Characterization................................................................................................................................................................................................ 171
Health hazard evaluation of short- and long-term exposure ................................................................................................................................... 171
Relevance for the EU-population exposure............................................................................................................................................................. 172
Result ....................................................................................................................................................................................................................... 172
Xylenes (ortho-, meta- and para-) ..............................................................................................................................173
1. Compound identification .......................................................................................................................................................................................... 173
2. Physical and Chemical properties............................................................................................................................................................................. 173
3. Indoor Air Exposure assessment .............................................................................................................................................................................. 173
Emission sources...................................................................................................................................................................................................... 173
Indoor air and exposure concentrations................................................................................................................................................................... 174
4. Toxicokinetics........................................................................................................................................................................................................... 179
Absorption................................................................................................................................................................................................................ 179
Distribution .............................................................................................................................................................................................................. 179
Metabolism and elimination .................................................................................................................................................................................... 179
5. Health effects ............................................................................................................................................................................................................ 181
Differences among individual xylene isomers ........................................................................................................................................................ 181
Effects of short-term exposure................................................................................................................................................................................. 181
Effects of long-term exposure ................................................................................................................................................................................. 183
Carcinogenic potential and genotoxicity................................................................................................................................................................. 188
Interactions with other chemicals ............................................................................................................................................................................ 189
Odour perception ..................................................................................................................................................................................................... 189
Summary of Xylenes Dose Response Assessment.................................................................................................................................................. 190
6. Risk Characterization................................................................................................................................................................................................ 191
Health hazard evaluation of short- and long-term exposure ................................................................................................................................... 191
Relevance of EU-population exposure to xylenes .................................................................................................................................................. 191
Result ....................................................................................................................................................................................................................... 192
7. Aromatic compounds: Comparison of relevant data................................................................................................................................................ 193
The reciprocal calculation approach for mixtures................................................................................................................................................... 193
Styrene .............................................................................................................................................................. 195
1. Compound identification .......................................................................................................................................................................................... 195
2. Physical and Chemical properties............................................................................................................................................................................. 195
3. Indoor Air Exposure assessment .............................................................................................................................................................................. 195
Emission sources...................................................................................................................................................................................................... 195
Indoor air and exposure concentrations................................................................................................................................................................... 196
4. Toxicokinetics........................................................................................................................................................................................................... 197
Absorption................................................................................................................................................................................................................ 197
Distribution .............................................................................................................................................................................................................. 197
Metabolism and elimination .................................................................................................................................................................................... 197
5. Health effects ............................................................................................................................................................................................................ 198
Effects of short-term exposure................................................................................................................................................................................. 198
Effects of long-term exposure (noncancer) ............................................................................................................................................................. 200
Macromolecular adducts and genetic toxicity......................................................................................................................................................... 207
Carcinogenic potential ............................................................................................................................................................................................. 208
Interactions with other chemicals ............................................................................................................................................................................ 210
Odour perception ..................................................................................................................................................................................................... 210
Summary of Styrene Dose Response Assessment .................................................................................................................................................. 211
6. Risk Characterization................................................................................................................................................................................................ 212
Health hazard and cancer risk evaluation of short- and long-term exposure.......................................................................................................... 212
Relevance of EU-population exposure to styrene ................................................................................................................................................... 213
Result ....................................................................................................................................................................................................................... 213
The INDEX project Final report
7
Ammonia .............................................................................................................................................................. 214
1. Compound identification .......................................................................................................................................................................................... 214
2. Physical and Chemical properties............................................................................................................................................................................. 214
3. Indoor Air Exposure assessment .............................................................................................................................................................................. 214
Indoor air and exposure concentrations................................................................................................................................................................... 214
4. Toxicokinetics........................................................................................................................................................................................................... 216
Absorption................................................................................................................................................................................................................ 216
Distribution .............................................................................................................................................................................................................. 216
Metabolism and elimination .................................................................................................................................................................................... 216
5. Health effects ............................................................................................................................................................................................................ 217
Effects of short-term exposure................................................................................................................................................................................. 217
Effects of long-term exposure ................................................................................................................................................................................. 220
Reproductive, mutagenic and carcinogenic effects................................................................................................................................................. 224
Interactions with other chemicals ............................................................................................................................................................................ 224
Odour perception ..................................................................................................................................................................................................... 224
Summary of Ammonia Dose Response Assessment............................................................................................................................................... 225
6. Risk Characterization................................................................................................................................................................................................ 226
Health hazard evaluation of short-term exposure.................................................................................................................................................... 226
Health hazard evaluation of long-term exposure..................................................................................................................................................... 226
Relevance of EU-population exposure to Ammonia............................................................................................................................................... 226
Result ....................................................................................................................................................................................................................... 226
Limonene .............................................................................................................................................................. 227
1. Compound identification .......................................................................................................................................................................................... 227
2. Physical and Chemical properties............................................................................................................................................................................. 227
3. Indoor Air Exposure assessment .............................................................................................................................................................................. 227
Contribution of inhalation exposure to total exposure............................................................................................................................................ 227
Terpene/ozone reaction products........................................................................................................................................................................... 228
Indoor air and exposure concentrations................................................................................................................................................................... 228
4. Toxicokinetics........................................................................................................................................................................................................... 230
Absorption................................................................................................................................................................................................................ 230
Distribution .............................................................................................................................................................................................................. 230
Metabolism and elimination .................................................................................................................................................................................... 230
5. Health effects ............................................................................................................................................................................................................ 231
Effects of short-term exposure................................................................................................................................................................................. 231
Effects of long-term exposure ................................................................................................................................................................................. 232
Carcinogenic and genotoxic effects......................................................................................................................................................................... 233
Interactions with other chemicals ............................................................................................................................................................................ 234
Odour perception ..................................................................................................................................................................................................... 234
Summary of Limonene Dose Response Assessment .............................................................................................................................................. 234
6. Risk Characterization................................................................................................................................................................................................ 235
Health hazard evaluation of long-term exposure..................................................................................................................................................... 235
Relevance of EU-population exposure to limonene................................................................................................................................................ 235
Result ....................................................................................................................................................................................................................... 235
alpha-Pinene .............................................................................................................................................................. 236
1. Compound identification .......................................................................................................................................................................................... 236
2. Physical and Chemical properties............................................................................................................................................................................. 236
3. Indoor Air Exposure assessment .............................................................................................................................................................................. 237
Terpene/ozone reaction products............................................................................................................................................................................. 237
Indoor air and exposure concentrations................................................................................................................................................................... 237
4. Toxicokinetics........................................................................................................................................................................................................... 238
Absorption................................................................................................................................................................................................................ 238
Distribution .............................................................................................................................................................................................................. 238
Metabolism and elimination .................................................................................................................................................................................... 238
5. Health effects ............................................................................................................................................................................................................ 239
Effects of short-term exposure................................................................................................................................................................................. 239
Effects of long-term exposure ................................................................................................................................................................................. 240
Carcinogenic and genotoxic potential ..................................................................................................................................................................... 240
Interactions with other chemicals ............................................................................................................................................................................ 241
Odour perception ..................................................................................................................................................................................................... 241
Summary of alpha-Pinene Dose Response Assessment.......................................................................................................................................... 241
6. Risk Characterization................................................................................................................................................................................................ 242
Health hazard evaluation of long-term exposure..................................................................................................................................................... 242
Relevance of EU-population exposure to α-Pinene ................................................................................................................................................ 242
Result ....................................................................................................................................................................................................................... 242
5. Risk management tools .......................................................................................................................................... 243
6. Recommendations and management options....................................................................................................... 246
6.1 High priority chemicals .......................................................................................................................................................................................... 246
6.2 Second priority chemicals....................................................................................................................................................................................... 247
6.3 Chemicals requiring further research with regard to human exposue or dose response........................................................................................ 248
The INDEX project Final report
8
Annex 1. Phase 1: Exposure and dose-response data for indoor air pollutants collected in the literature review...................................................... 249
Annex 2. Phase 2: The selection of compounds to the further analysis....................................................................................................................... 258
Annex 3. Phase 3: Compounds selected into the detailed risk assessment.................................................................................................................. 265
Annex 4. The EXPOLIS study: parameters describing the indoor air concentration distributions plotted to the graphs of this report..................... 270
Annex 5. The National Survey of air pollutants in English homes: parameters describing the indoor air concentration distributions
plotted to the graphs of this report................................................................................................................................................................ 271
Annex 6. The French National Survey on Indoor Air Quality (preliminary results from the ongoing project): parameters describing the indoor
air concentration distributions plotted to the graphs of this report (Golliot et al 2003, Kirchner 2004).................................................... 272
Annex 7. Relevant National and International Guidelines and Recommendations .................................................................................................... 273
Bibliographic references.................................................................................................................................................................................. 275
References in introductory chapters (chapters 1 to 4).................................................................................................................................................. 275
References by compounds (chapter 4).......................................................................................................................................................................... 276
Formaldehyde .......................................................................................................................................................................................................... 276
Carbon monoxide..................................................................................................................................................................................................... 282
Nitrogen dioxide ...................................................................................................................................................................................................... 286
Benzene.................................................................................................................................................................................................................... 291
Naphthalene ............................................................................................................................................................................................................. 294
Acetaldehyde............................................................................................................................................................................................................ 298
Toluene..................................................................................................................................................................................................................... 303
Xylenes..................................................................................................................................................................................................................... 309
Styrene ..................................................................................................................................................................................................................... 315
Ammonia.................................................................................................................................................................................................................. 321
Limonene ................................................................................................................................................................................................................. 325
alpha-Pinene............................................................................................................................................................................................................. 328
References in Annex 1, 2 and 3 .................................................................................................................................................................................... 330
The INDEX project Final report
9
Executive Summary
Introduction and objectives
The INDEX project (Critical Appraisal of the Setting and Implementation of Indoor exposure Limits in the EU) started
in December 2002 and had a duration of two years, until December 2004.The project was financially supported by DG
SANCO and it was coordinated and carried out by the JRC in collaboration with a Steering Committee of leading
European experts in the area of indoor air pollution. Scope of INDEX was to identify priorities and to assess the needs
for a Community strategy and action plan in the area of indoor air pollution by:
- setting up a list of compounds to be regulated in indoor environments with priority on the basis of health
impact criteria
- providing suggestions and recommendations on potential exposure limits for these compounds, and
- providing information on links with existing knowledge, ongoing studies, legislation etc. at world scale.
Methodology
The main steps to be followed in the project as they have been defined by the Steering Committee were:
- literature review (step 1)
- setting up criteria to select compounds (step 2)
- review of exposure and dose/response data (step 3)
- risk characterization of the selected compounds (step 4)
- prioritisation of the selected compounds (step 5) and
- recommendations and risk management options on potential exposure limits (step 6)
In the first step the world literature was reviewed to collect background information on exposure concentrations and
dose/response data of the chemical pollutants that have strong indoor sources. In the second step, the Steering
Committee has defined the criteria for the selection of pollutants for further risk analysis. After careful examination of
exposure and toxicological dose response data (step 3), the Steering Committee ended up to a short list of pollutants that
were selected for the risk characterization (step 4). The compounds that probably cause the highest health risk in
European population according to the risk characterization were prioritized (step 5). Finally, recommendations and risk
management options on European exposure limits should be given for a few selected compounds (step 6).
In order to accomplish with the tasks mentioned above the Steering Committee was divided into two working groups,
one for exposure assessment and one for dose/response assessment.
Exposure to selected indoor air pollutants was estimated by reviewing exposure data collected from scientific literature,
from available databases, and by personal communications. The aim of this work was to summarise prevailing indoor
air and personal exposure concentrations of these compounds in Europe and also worldwide. Results from population-
based studies have been preferred to be able to generalise the results from studied individuals to larger populations,
targeting to assess exposures of all the Europeans. Considering the fact that no new data could be generated in this
project, the steering group defined the following criteria to be able to select the chemical compounds to the risk
analyses:
1. Only single compounds will be considered
2. The compound should have common indoor sources, which dominate the exposures of at least significant fraction
of the population
3. The compound should have known health effects.
It was also decided that compounds, which have been regulated by specific guidelines or regulations would be excluded
from theses analyses. For example, radon and tobacco smoke were excluded from the risk assessment process due to
these criteria.
In preparing the dose-response assessment fact sheets of the selected chemicals, information were retrieved from
scientific literature (mainly by electronic search), comprehending toxicological reviews of leading health organizations,
risk evaluation documents and available databases. In addition, Toxline and Medline were searched for relevant
scientific communications published up to September 2004.
Nearly all key-studies referred to in the present assessment, establishing effect levels for appropriate toxicological
endpoints, are those selected by health organizations for the derivation of health based limits of exposure or among risk
The INDEX project Final report
10
assessment requirements. Although not specifically addressing health hazards and risks associated with indoor air
exposure, i.e. not being designed for the expression of effects at lowermost exposure concentrations, nearly all studies
were aimed at identifying the most sensitive endpoint considered to be of relevance to humans. Where relevant, studies
conducted on susceptible sub-populations (e.g. asthmatics, infants, children, pregnant women etc.) were quoted and
taken into consideration in the risk characterization.
In the final step of the general risk assessment process, the incidence of health hazards and risks in the European
population, associated with indoor exposure to individual chemicals, was estimated. Limits of exposure (ELs, following
short- and long-term exposure) were derived for each chemical after selection of a critical study describing the
appropriate toxicological endpoint and by applying the “no-observed-adverse-effect level (NOAEL) / assessment factor
(AS)” approach. Where no NOAEL observation was documented, a lowest-observed-adverse-effect level (LOAEL) was
taken and an additional assessment factor of 10 used for EL derivation. Only for one compound (benzene) the
characterization was based on population cancer risk estimation. Where supported by scientific evidence, susceptible
subpopulations were accounted for, in particular: asthmatic individuals, infants, children, individuals with heart
diseases, pregnant women, individuals with enzyme deficiencies.
On the basis of the available information and after careful examination of the existing data, the steering committee
finally decided to include into a detailed assessment 14 out of initial 41 candidate compounds (phase 4) i.e.:
acetaldehyde, alpha-pinene, benzene, carbon monoxide, d-limonene, formaldehyde, meta-and para xylene, ortho-
xylene, naphthalene, ammonia, nitrogen dioxide, styrene and toluene.
Risk assessment of the selected compounds
Information from the exposure assessment (Chapters 1-3) and toxicity assessment (Chapters 4-5) were integrated and a
risk characterization (Chapter 6) performed on each chemical. Based on the conclusions of the assessments and on the
completeness of individual databases, a priority ranking was arranged with the 14 chemicals assigned to three groups as
given hereafter.
Group 1: High priority chemicals
Formaldehyde : Because of its high chemical reactivity, formaldehyde is the most important sensory irritant
among the chemicals assessed in the present report. Due to being ubiquitous pollutant in
indoor environments and to the increasing evidence indicating that children may be more
sensitive to formaldehyde respiratory toxicity than adults it is considered a chemical of
concern at levels exceeding 1 µg/m
3
, a concentration more or less corresponding with the
background level in rural areas. Results from available exposure data, although limited,
confirm that almost the entire population is exposed indoors at levels (Median level±sd: 26±6
µg/m
3
; 90th±sd: 59±7 µg/m
3
; N = 6) higher than this background level, here established as
the limit of exposure, with at least 20% of the European population exposed at levels
exceeding the no-observed-effect-level (NOAEL: 30 µg/m
3
). Within the concentration range
measured, mild irritation of the eyes could be experienced by the general population as well
as the odour perceived above 30 µg/m
3
.
Reported formaldehyde concentrations were lower (99
th
< 150 µg/m
3
)
than a presumed
threshold for cytotoxic damage to the nasal mucosa (about 1 mg/m
3
) and hence
considered low enough to avoid any significant risk of upper respiratory tract cancer in
humans. The last statement could be subjected to changes due to the current IARC
revision of the carcinogenicity of formaldehyde.
Carbon monoxide : Available exposure data confirm that Carbon Monoxide (CO) sources in EU-residences are
contributing to short-term rather than to long-term exposures. Personal exposure outcomes
averaged over 1-hour were considered of moderate concern even for the most susceptible
subpopulations. Nevertheless, uncertainties resulting from the predictive capabilities of the
CFK-model* in individuals exposed at low
CO concentrations and its applicability to
sensitive subpopulations, suggest that about 10% of the general non-smoking population
experience CO levels which could be hazardous for individuals with heart diseases. Increased
exposures could be expected for residences in the vicinity of busy city streets.
In addition, there is no evidence that long-term CO exposures in EU residences contribute to
carboxyhaemoglobin levels in blood higher than the baseline levels resulting from
endogenous production in normal, non-smoking individuals.
The INDEX project Final report
11
On the other hand and in contrast with all other chemicals assessed in the present report,
carbon monoxide causes a considerable number of deaths and acute poisonings in the general
population (with complications and late sequel). Also, individuals suffering from CO
poisoning are often unaware of their exposure because symptoms are similar to those
associated with viral illness or clinical depression. In indoor environments, these health risks
are nearly completely associated with the incorrect use of combustion devices or faulty
unvented gas appliances.
* The physiologically based pharmacokinetic (PBPK) model of Coburn, Forster, and Kane (CFK-model) is a reliable
method for predicting COHb blood levels for exposure to a given ambient carbon monoxide concentration. This
model has been extensively validated over many years. Precision is acceptable, providing that the original conditions
of use are rigorously applied.
Nitrogen dioxide : Reported maximum nitrogen dioxide (NO
2
) levels associated with the use of gas appliances
in homes (gas cooking and heating) are in the range 180-2500 µg/m
3
. Exposure at these
levels could generate effects in the pulmonary function of asthmatics, considered to be the
subjects most susceptible to acute NO
2
exposure, with the lower end of the range
approximating the WHO guideline (200 µg/m
3
, 1-hour average), established for the
protection of asthmatic individuals and the upper end starting to affect health in normal
individuals.
For long-term exposures, increased respiratory symptoms and lung function decreases in
children were documented to be the most sensitive effect in the general population. Measured
background
levels in European homes indicate that a remarkable portion of the population is
exposed at NO
2
levels higher than current guideline values protecting from respiratory effects
in children. In up to 25% of the investigated residences (45% in an Italian study) NO
2
levels
exceeded the German indoor-related guideline value (GV II: 60 µg/m
3
, 1-week average),
what would have resulted in immediate action i.e. the examination of the situation with
regard to a need for control measures.
On the other hand, safe levels in homes, i.e. < 40 µg/m
3
(following the WHO recommended
annual value), are not likely to be achievable everywhere (e.g. in areas with intense
automotive traffic) given that ventilation alone may introduce outdoor air containing such
concentrations.
Benzene : Benzene is ubiquitous in the athmosphere, mainly due to anthropogenic sources (90%), with
concentrations in the European continental pristine air ranging from 0.6 to 1.9 µg/m
3
. It is a
genotoxic carcinogen and hence no safe level of exposure could be recommended. Results
from nine monitoring surveys indicate that the European population is experiencing in their
homes an increased risk in contracting benzene induced leukaemia, with respect to the
estimated background lifetime risk of 7-8 cases per one million people (considering the WHO
unit risk factor). Based on the available exposure data (Median levels±sd: 4.2±3.2 µg/m
3
;
90
th
levels±sd: 11.5±11.1 µg/m
3
; N = 9) two main scenarios could be described as follows:
People living in highly trafficked urban areas are expected, on average, to
experience an estimated 6 to 30-fold increase in contracting benzene induced
leukaemia during their life, the benzene levels encountered in these areas not being
expected to produce chronic effects other than cancer, in particular haematological
effects, nor acute sensory effects such as odour perception (odour threshold: 1.2
mg/m
3
) and sensory irritation. Also, a reduced contribution of specific indoor
sources is likely to be expected, given that ventilation alone may introduce increased
outdoor benzene levels.
People living in rural areas or poorly trafficked towns were expected, on average, to
experience an estimated 1 to 5-fold increase in contracting benzene induced
leukemia during their life, this factor depending principally on the presence of
indoor sources.
Naphthalene : With regard to the general population a long-term exposure limit has been set at 10 µg/m
3
,
according to the assumption that nasal effects observed in mice are consistent with the health
effects reported among exposed workers. Available exposure data indicate that, on average,
the European population is exposed at naphthalene levels 10 times lower than this EL,
although an important exception resulted from a survey held in Athens, were levels
exceeding the EL were measured in nearly all residences. It is assumed that increased
residential exposures originate from the use of naphthalene based moth-repellents, a
The INDEX project Final report
12
widespread use occurring in certain countries of the Mediterranean area.
An important source of uncertainty in establishing safe exposure limits is the potentially
greater sensitivity of certain subpopulations to naphthalene toxicity, including infants and
neonates, and individuals deficient in glucose-6-phosphate dehydrogenase (G6PD), the
prevalence of this inherited deficiency reported to be 2 to 20% in defined Mediterranean
subpopulations. In these latter cases manifested effects are hemolytic anemia and its sequel.
In relation to carcinogenicity, naphthalene is not genotoxic in vivo and thus tumour
development, observed in rodents, is considered to arise via a non-genotoxic mechanism.
Also, the underlying mechanism for the development of nasal tumours in the rat is considered
to be the chronic inflammatory damage seen at this site. It follows that prevention of local
tissue damage would prevent subsequent development of tumours.
Group 2: Second priority chemicals
Acetaldehyde : The results from only three indoor air monitoring surveys allow a crude estimate of average
acetaldehyde concentrations in European residences. Median concentrations (10-20 µg/m
3
)
are one order of magnitude lower than the Exposure Limit set here at 200 µg/m
3
and are
within the same range of concentrations occurring in exhaled breath following its
endogenous production in the general population, not taking into account increases resulting
from the consumption of alcoholic beverages. Considering that exogenous acetaldehyde peak
exposures are mainly associated with tobacco smoke, concentrations in the order of the
Exposure Limit could be expected following intense cigarette consumption.
Assuming that the available exposure data are indicative of the population residential
exposure it is concluded that people in Europe do not experience increased health hazards
associated with acetaldehyde levels in their homes, although additional work should be
warranted for a better characterization of exposure and dose response.
Also, measured indoor levels are lower than a presumed threshold for cytotoxic damage to
the nasal mucosa, and hence considered low enough to avoid any significant risk of upper
respiratory tract cancer in humans.
Toluene : Human effects on the central nervous system are considered as the most sensitive effect in
both short- and long-term inhalatory exposure to toluene. Available exposure data indicate
that the European population is not experiencing health effects of concern resulting from the
exposure to toluene in their homes. Results from ten monitoring surveys show that toluene
levels in the order of the established exposure limit of 300 µg/m
3
could be reached under
worse-case conditions and in a limited number of urban residences. On average, median
concentrations (90th percentile) were found to be 16 (5) times lower than the EL. Also,
short-term exposures associated with human indoor activities are not expected to exceed the
acute EL set here at 15.000 µg/m
3
.
Xylenes : A chronic exposure limit of 200 µg/m
3
has been derived based on generally mild adverse
effects associated with CNS and increase in the prevalence of eye irritation and sore throat.
The results of eight monitoring surveys indicate that background levels of xylenes in
European residences are of no concern to human health since median (90th percentile) levels
are, on average, 20 (6) times lower than the EL established. Acute exposure data indicate that
it is very unlikely that xylenes emissions associated with human indoor activities would
generate levels in the order of the proposed short-term EL of 20 mg/m
3
, considered
protective for irritative effects in the general population.
Although human exposure most likely occurs to the mixture of xylene isomers, animal and
human toxicity data suggest that mixed xylenes and the different xylene isomers produce
similar effects.
Styrene : A long-term exposure limit (EL) of 250 µg/m
3
has been derived based on the assumption that
neurological effects are probably the most sensitive indicator of styrene toxicity. When
examining the results of eight monitoring surveys it can be concluded that background
styrene concentrations in European residences are of no concern to human health since
median levels are, on average, two orders of magnitude below the established EL. Although
no acute exposure data were available, it is unlikely that styrene emissions associated with
human indoor activities would generate levels up to the proposed short-term EL of 2000
The INDEX project Final report
13
µg/m
3
, considered protective for irritative effects in asthmatics.
Although genotoxic effects in humans have been observed at relatively low concentrations,
they were not considered as critical endpoints for the derivation of the exposure limit, in
view of the equivocal evidence for the carcinogenicity of styrene in humans (WHO).
Group 3: Chemicals requiring further research with regard to human exposure or dose response
Ammonia : There is a lack of knowledge concerning indoor concentrations and exposures of ammonia.
Exposure data are limited on only one monitoring survey describing concentrations of
ammonia in Finnish homes with and without known indoor air quality (IAQ) problems. In
both cases measured concentrations were within the same order of magnitude with both
exposure limits here established for short- and long-term effects (70 and 100 µg/m
3
,
respectively), relating on irritative effects and pulmonary functions and taking into account
the particular susceptibility of asthmatic subjects. It is assumed that exposure concentrations
in the order of the short-term EL could easily be attained during domestic activities making
use of ammonia containing household products.
Limonene : An attempt has been done in deriving an exposure limit (EL) for long-term effects associated
with limonene exposure by referring to a study on volunteers exposed at sub-acute (2 hours)
inhalation doses. When comparing this EL (450 µg/m
3
) with the results from seven indoor
surveys it is concluded that no neurological effects would be expected at background
limonene levels encountered in European homes, with median (90
th
percentile) levels at least
10 (3) times lower than the proposed EL. It is assumed that at 10-fold the level set as the EL,
health effects could be expected following acute exposure. Due to its widespread use as a
flavouring agent in numerous consumer products, short-term exposures at levels in the order
of some mg/m
3
could not be excluded, although significative exposure data are lacking.
An exacerbation of effects (not better defined) could be expected following the concomitant
presence in residences of ozone emitting sources, due to the formation of irritant reaction
products.
α-Pinene : An attempt has been done in deriving an exposure limit (EL) for long-term effects associated
with α-pinene exposure by referring to a study on volunteers exposed at sub-acute (2 hours)
inhalation doses. When comparing this EL (450 µg/m
3
) with the results from six indoor
surveys it is concluded that no irritative effects to the eyes, nose and throat would be
expected at background α-pinene levels encountered in European homes, with median (90
th
percentile) levels at least 40 (10) times lower than the proposed EL. It is assumed that at 10-
fold the level set as the EL, health effects could be expected following acute exposure. Due
to its widespread use as a flavouring agent in numerous consumer products, short-term
exposures at levels in the order of some mg/m
3
could not be excluded, although significative
exposure data are lacking.
An exacerbation of effects (not better defined) could be expected following the concomitant
presence in residences of ozone emitting sources, due to the formation of irritant reaction
products.
The INDEX project Final report
14
Risk management tools
The indoor air risk management tools include:
IAQ Standards and guidelines: The huge quantity and privacy of indoor spaces makes enforceable indoor air quality
standards impossible. Guideline values for key pollutants, however, are useful when matching potential sources,
occupant needs and ventilation rates in the design of new building or renovation of an existing one. Sometimes they can
also be applied in judging the acceptability of indoor air quality in a building.
Building codes and ventilation standards apply to every new building, and can therefore be powerful means of
preventing or controlling indoor sources and ensuring sufficient contaminant dilution.
Building and household equipment standards and permits cover new installations, and can therefore ensure that
improper - hazardous or avoidable - emissions are avoided or properly controlled, e.g. exhausted.
Mandatory maintenance and inspections of, e.g., gas appliances or fireplaces & chimneys, help avoid gradual
deterioration of the equipment, and accumulation of its risks, and ensure that safe operation requirements are met
through their lifetime.
Limits, labelling and reporting of the contents of or releases from building products, furnishing materials,
equipment and consumer products can be quite inflexible and heavy instruments when mandated by law, but flexible
and effective on voluntary basis, as long as commonly agreed (between consumers, builders, industry and public health
authorities), widely published and generally understood criteria are used. Clear product labels guide informed selections
of both builders and consumers.
Public awareness raising and information is the key to safe indoor environments, because, due to their sheer
numbers, a great majority of potential indoor pollution risks must be identified, assessed and managed by occupants
themselves. They are best helped by widespread and general awareness raising information, and easily found and
understood sources (leaflets, internet sites) for more specific technical information.
Recommendations and management options
The recommendations and management options proposed protect the general population and most individuals most of
the time, but they will not prevent all harm to every individual. Besides, the proposed measures will not prevent health
damage from outdoor air pollutants which penetrate indoors. Following general recommendations apply to many indoor
air contaminants:
Restrict tobacco smoking in all indoor spaces.
Restrict the construction of attached garages, or isolate them from living and working spaces.
Ensure that ventilation dilutes predictable indoor emissions below the guideline levels.
Raise public awareness about indoor air risks.
High priority chemicals
The high priority compound list consists of 5 chemicals, with potential of high indoor concentrations, uncontested
health impacts, and effective risk management.
Formaldehyde: The non-carcinogenic no-effect level is 30 µg/m
3
. Pending on IARC revision of formaldehyde
carcinogenecity, a guideline should be as low as reasonably achievable. Management options are to restrict and avoid
the use of formaldehyde containing materials and products in buildings:
Nitrogen Dioxide: Proposed guideline values are 40 µg/m
3
(1-week) and 200 µg/m
3
(1-hour). Management options are
to connect each indoor combustion device/appliance to chimney or vented hood, and to ensure sufficient local extract
ventilation in kitchens with gas stove.
Carbon Monoxide: Proposed guideline values are 10 mg/m
3
(8-hour) and 30 mg/m
3
(1-hour). Management options are
to connect each combustion equipment/appliance to chimney or vented hood, to ensure sufficient local extract
ventilation in kitchens with gas stove, mandatory inspection and maintenance of indoor combustion devices, and CO
alarms.
Benzene: Benzene is a carcinogen, its indoor air concentration should be kept as low as reasonably achievable, and not
exceed outdoor concentrations. Management options are to ban benzene sources indoors, and lower the permissible
benzene content in any building material and consumer product.
The INDEX project Final report
15
Naphtalene: Proposed long term guideline value is 10 µg/m
3
. Management option is to restrict the use of naphthalene
containing household products.
Second priority chemicals,
which are not considered to urgently require regulatory risk management actions
specifically in indoor air are Acetaldehyde, o-, p- and m-Xylene, Toluene and Styrene.
Additional chemicals of interest
for indoor air risk management, which are considered to require further research with
regard to human exposure or dose response before recommendations can be made, are Ammonia, delta-Limonene, and
alpha-Pinene.
The INDEX project Final report
16
The INDEX project Final report
17
1. Introduction
Human exposure to air pollution occurs over 90% indoors, but it depends on both indoor and outdoor air pollution.
Outdoor air pollution is important mainly because indoor air is linked to outdoor air via ventilation. The strength of the
personal exposure-outdoor air association varies considerably between the individuals, their activities,
microenvironments and pollutants. On one hand leisure time exposures of active individuals to various pollutants are
mainly associated with outdoor air pollution levels. On the other hand workday exposures, exposures of individuals
leading sedentary lifestyles in closed or air conditioned spaces and exposures to pollutants like formaldehyde and radon
are essentially independent of outdoor air pollution levels.
Past European Air Quality Directives have not taken population exposure sufficiently into account. They have up to
now been mainly oriented on outdoor air pollution. EU legilslation addresses inter alia air quality standards, national
emission ceilings and emissions from vehicles and industries. However, the Sixt Environmental Action Plan and the
new launched Environmental and Health Strategy are oriented towards the impact of environmental risk factors on
human health, and DG SANCO and other relevant DGs are developing proposals for a public health policy. So, in
addition to ambient air pollution, the pollution inside confined environments as well as the extent of personal mobility
and specific activities all play a significant role in exposure to air pollutants. The relative importance of these pollutants
varies greatly depending on the sources, pollutants, and individuals or populations of concern. Concentrations of e.g.
volatile organic compounds (VOC), in general, are higher indoors than outdoors, yet for some VOCs outdoor air levels
may significantly affect respective indoor levels. For many chemicals occurring indoors the risk for human health and
comfort is almost unknown and difficult to predict, in particular, the risk associated with chronic low dose exposures to
these compounds because of a quite limited toxicological data and information on dose-response characteristics in
human or animal models. Only very recently some few data have been become available, which partly make possible to
carry out reliable exposure and risk assessments. Due to the missing risk assessments, it has been difficult to regulate
the presence of these chemicals in indoor environments up to now.
It is, therefore, recommended to develop a strategy for indoor air quality assessment and management in Europe and
that future clean air policies take into account the total air exposure of European citizens, which will necessarily include
exposures to pollutants from both outdoor and indoor sources. This report offers background information for this
strategy planning.
The INDEX project Final report
18
2. Objectives
The aim of the project was to create a network of European leading scientists in the area of indoor air pollution and the
herewith-associated health impacts in order to identify priorities for a Community strategy and action plan. The key
issues that were addressed within the project were:
- to provide information on links with existing knowledge, ongoing studies, legislation etc.,
- the setting up of a list of priority substances to be regulated in indoor environments on the basis of health
impact criteria,
- to provide suggestions and recommendations on potential exposure limits or other risk management
procedures for these substances, and
- to assess essential research needs for pollutants with high risk potential, but insufficient information for
carrying out risk assessment and setting regulatory objectives or selecting regulatory options.
The INDEX project Final report
19
3. Methodology
3.1 Risk Assessment
Risk assessment was based on indoor air quality and exposure data from the scientific literature, databases and directly
from the researchers. Similarly, dose/response data was reviewed from scientific literature. Two working groups (WG)
of experts from several countries were established to assess these data. Finally, risk characterisation, recommendations
for risk management and conclusions were drawn by the scientific steering committee based on the work of the working
groups for exposure assessment and dose-response assessment. The main steps of the project are presented in Figure
3.1.1. The risk assessment approach applied to this project is presented in more detail in the following paragraphs.
Step 1:
Literature
review
Step 2: Setting up
criteria to select
compounds
Step 3: Review of
exposure and
dose/response data
Step 4: Risk
characterisation of the
selected compounds
Step 5: Prioritisation of
the selected compounds
Step 6:
Recommendations and
risk management
options on potential
exposure limits
Figure 3.1.1: The main steps of the INDEX project.
3.1.1 Selection of Indoor Air Chemicals for Consideration (Hazard
Identification)
The hazard identification of the indoor air pollutants was assessed combining the information of the prevalence of
pollutants in European homes with the available knowledge of adverse health effects that these compounds had been
linked to in toxicological or epidemiological studies. If a compound was present in the indoor air and it had had adverse
health effects, it was considered as a potential hazard to European populations and was thus included in the risk
assessment process. The process and the criteria to include or exclude each compound to the risk assessment process in
this project are presented in the following paragraphs.
The INDEX project Final report
20
The selection criteria of the compounds to be included in the risk analysis
Considering the time limit of this exercise, and the fact that no new data could be generated in this project, the steering
group defined the following criteria to be able to select the chemical compounds to the risk analyses:
1. Only single compounds will be considered
2. The compound should have common indoor sources, which dominate the exposures of at least significant
fraction of the population
3. The compound should have known health effects.
It was also decided that compounds, which have been regulated by specific guidelines or regulations would be excluded
from this analysis. For example, radon and tobacco smoke were excluded from the risk assessment process due to these
criteria.
Phase 1: Literature review
In the first phase, a literature review was carried out to collect information about candidate pollutants to be assessed in
the later stages of the project. The scientific literature of the indoor air pollutants was reviewed by using several search
engines in the Internet and by searching from the web pages of the relevant journals. In addition to electronic search,
numerous study reports concerning indoor air pollutants were reviewed.
The main focus of the review was on recent population-based studies to be able to evaluate current population
exposures to selected pollutants in Europe. Only compounds with known indoor sources and known health effects were
taken into consideration.
Based on the literature review, a summary table of the concentrations in residential indoor, workplace indoor,
residential outdoor and personal exposures was created for the compounds meeting the first criteria (see Annex 1).
Simultaneously, some dose/response data were collected for the selected compounds. Also the existing international
(i.e. EU and WHO) and national guidelines for those compounds in indoor air were reviewed. Finally, the output of the
literature review was used as an input for the next steps of the risk assessment.
Establishment of the working groups
For the evaluation and development of the reviewed literature, two working groups (WG) were established, one for
exposure assessment (WG
ea
) and one for dose/response assessment (WG
dr
). The following experts were nominated to
the working groups:
WG
ea
on exposure assessment
Members: M. Jantunen (co-ordinator, KTL), Finland, C. Cochet, France, S. Kirchner, France, K. Koistinen, Finland, J.
Mc Laughlin, Ireland, S. Kephalopoulos, EU/JRC-Ispra, E. Oliveira-Fernandez, Portugal, B. Seifert, Germany, and
WG
dr
on dose/response assessment
Members: P. Carrer (co-ordinator, UniMi), Italy, T. Lindvall, Sweden, M. Maroni, Italy, L. Mølhave, Denmark and C.
Schlitt, Italy.
Phase 2: The selection of compounds to the further analysis
In the second phase of the selection process, the working groups assessed the reviewed data and collected more detailed
information for the previously selected compounds. The aim of the work was to select about 20 compounds for further
analyses. In this phase the steering group excluded more compounds using the following criteria:
- no expressed concerns for health at present levels (for example acetone, decane, ethylbenzene, phenol ,
propylbenzene, trimethylbenzene)
- compound already regulated by use restrictions for indoor materials (pentachlorophenol)
- incomplete or no dose-response data available at present levels (methyl-ethyl-ketone, propionaldehyde)
- the main route/media for the exposure to the compound is other than indoor air (lead, mercury).
After detailed review and discussion of the available information, the working groups selected the 25 compounds
presented in Annex 2 to the more detailed analysis.
The INDEX project Final report
21
Phase 3: Compounds selected for a detailed risk assessment
In addition to exposure and dose/response data, also data about odour threshold values were considered important and
thus, these data were added to the background information. The standardised human odour threshold values were taken
from Devos et al. (1990).
On the basis of the available information and after an extensive discussion on the chemical substances, the steering
group decided to conduct detailed assessment of the 14 compounds presented in Figure 3.1.1.1.(phase 3).
Based on the potential/estimated population risk caused by concentrations from indoor sources, toxicological properties
including hypersensitivity for allergy and asthma, other known health effects and comfort, the steering group decided to
separate the 14 compounds into two groups: 1) compounds to be considered with high priority and 2) compounds that
could be included in the final prioritisation after further examination and new published findings of their hazardous
potential and the other criteria set within the project.
After a first evaluation the compounds were initially classified as follows:
Group 1 (high priority) Benzene, Acetaldehyde, Formaldehyde, CO and NO
2
Group 2 (further information needed) m&p-Xylenes, o-Xylene, Naphthalene, Styrene, Toluene, a-Pinene, d-Limonene,
NH
3.
Flame-retardants were regarded as an emerging issue, which will require further consideration in the future. The
compound Tris-(2-chloroethyl) phosphate belongs to this group, but because reliable data on its sources and occurrence
in indoor environments, exposure routes and on toxicological properties were lacking, the compound was not included
in the evaluation procedure in this project.
Figure 3.1.1.1: The indoor originated compounds that were assessed and considered the most hazardous in the three
phases of the hazard identification process.
Phase 1
Phase 2
Phase 3
1. priority
Formaldehyde
Carbon monoxide
Nitrogen dioxide
Benzene
Naphtalene
Acetaldehyde
Ammonia
a-Pinene
Benzene
Carbon monoxide
d-Limonene
Formaldehyde
m&p-Xylene
Naphtalene
Nitrogen dioxide
o-Xylene
Styrene
Toluene
1-Butanol
2-Ethyl-1-hexanol
3-Carene
Acetaldehyde
Ammonia
a-Pinene
Benzaldehyde
Benzene
Cadmiuim
Carbon monoxide
Dichloromethane
Diisocyanate
d-Limonene
Formaldehyde
Hexaldehyde
m&p-Xylene
Naphtalene
Nitrogen dioxide
o-Xylene
Styrene
Tetrachloroethylene
Toluene
Trichloroethylene
Tris-(2-chloroethyl) phosphate
1-Butanol
2-Buthoxyethanol
2-Ethyl-1-hexanol
2-Methyl-1-propanol
3-Carene
Acetaldehyde
Acetone
Ammonia
a-Pinene
Benzaldehyde
Benzene
Benzo[a]pyrene
Cadmiuim
Carbon monoxide
Decane
Dichloromethane
Diisocyanate
d-Limonene
Ethylbenzene
Formaldehyde
Hexaldehyde
Lead
m&p-Xylene
Mercury
Methyl-ethyl-ketone
Naphtalene
Nitrogen dioxide
Nonane
o-Xylene
Pentachlorophenol
Phenol
Propionaldehyde
Propylbenzene
Styrene
Tetrachloroethylene
Toluene
Trichloroethylene
Trimethylbenzenes
Tris-(2-chloroethyl) phosphate
Undecane
The INDEX project Final report
22
3.1.2 Exposure Assessment
Exposure to selected indoor air pollutants was estimated by collecting exposure data from scientific literature, from
available databases, and by personal communications. The aim of this work was to summarise prevailing indoor air and
personal exposure concentrations of these compounds in Europe and also worldwide. These reviews are mainly focused
on indoor air and exposure concentrations measured recently in European population based studies such as EXPOLIS
(Jantunen et al 1998), German Environmental Surveys, GerES, (Seifert et al 2000), the German study on Indoor Factors
and Genetics in Asthma, INGA (Schneider et al 1999 and 2001), and a national survey of air pollutants in English
homes (Raw et al 2002). Also some preliminary results of the French National Survey (Golliot et al 2003, Kirchner
2004) were available during this project. Some comparisons have been also done to the TEAM (Wallace et al 1991) and
the NHEXAS (Sexton et al 1995, Pellizzari et al 1995) studies carried out in the USA. Results from population-based
studies have been used to be able to generalise the results from studied individuals to larger populations, targeting to
assess exposures of all the Europeans.
Population exposures are typically reported in literature using parameters such as arithmetic or geometric mean and
standard deviation. Mean concentrations give us a general picture of the concentration levels, but due to presence of
subpopulations that are exposed to much higher concentrations, the whole exposure distribution is needed when linking
these exposures to toxicological or epidemiological dose-response data. The distributions presented in this report are
drawn using arithmetic or geometric means and respective standard deviations, reported in the literature or extracted
from the databases, assuming that the measured data is log-normally distributed, which is a typical shape for the
distributions of the naturally occurring pollutants. These parameters taken from the large scale European population
based studies are presented in Annexes 4, 5, and 6.
Concentrations have been linked to the main emission sources if possible. Short time concentration peak values are
presented in tables and graphs to be used in the assessment of acute health effects.
European Union has recently published risk assessment reports (RAR) for some of the chemicals that are reviewed in
this report. Final RARs are available for naphthalene (EU 2003a), styrene (EU 2002a), toluene (EU 2003b) and a draft
report for benzene (EU 2002b). These reports were used as a data source in this project, and therefore the contents
presented in those reports have not been repeated in this report.
3.1.3 Dose/Response Assessment
In preparing the dose-response assessment fact sheets of 12 chemicals (m,p, and o-xylenes handled together),
information were retrieved from scientific literature (mainly by electronic search), comprehending toxicological reviews
of leading health organizations, risk evaluation documents and available databases, as outlined in Table 3.1.3.1. In
addition, Toxline and Medline were searched for relevant scientific communications published up to September 2004.
Nearly all key-studies referred to in the present assessment, establishing effect levels for appropriate toxicological
endpoints, are those selected by health organizations for the derivation of health based limits of exposure (WHO/GV,
IRIS/REL, OEHHA/RfC, ATSDR/MRL, HC/TC, UBA/GVII&I) or among risk assessment requirements (ECB).
Although not specifically addressing health hazards and risks associated with indoor air exposure, i.e. not being
designed for the expression of effects at lowermost exposure concentrations, nearly all studies were aimed at identifying
the most sensitive endpoint considered to be of relevance to humans. Summary definitions of exposure limits/guidelines
established by these health organisations are given in Table 3.1.3.2. Where relevant, studies conducted on susceptible
sub-populations (e.g. asthmatics, infants, children, pregnant women etc.) were quoted and taken into consideration in
the risk characterization.
The INDEX project Final report
23
Table 3.1.3.1: Toxicological reviews, risk evaluation documents
and databases consulted and referred to in the dose response assessment
World Health Organization
Air Quality Guidelines for Europe
International Program on Chemical Safety
Environmental Health Criteria, Concise International
Chemical Assessment Documents and Health and Safety
Guides
European Chemicals Bureau - Institute for Health and
Consumer Protection
European Union Risk Assessment Reports
Agency for Toxic Substances and Disease Registry
Toxicological Profiles
U.S.EPA - Integrated Risk Information System
Office of Environmental Health Hazard Assessment of
the Californian EPA
Acute and Chronic Toxicity Summaries
IARC monographs
Summaries and Evaluations
WHO IPCS ECB ATSDR IRIS OEHHA Others when relevant IARC
Formaldehyde
2000
EHC 89
1989
CICADS
40 2002
1999
being
reassessed
acute &
chronic
1999
NIWL 2003
c
Vol. 88
1995
2004 in
preparation
Acetaldehyde
EHC 167
1995
HSG 90
1995
1991
being
reassessed
chronic
1997
Vol. 71
1999
Ammonia
EHC 54
1986
HSG 37
1990
2002
draft
2003
acute &
chronic
1999
Carbon monoxide
2000
EHC 213
1999
acute
a
1999
Nitrogen dioxide
2000
EHC 188
1997
1993
acute
a
1999
UBA 1998
Benzene
2000
EHC 150
1993
2004
draft
1997 2003
b
acute &
chronic
1999
Vol. 29
Suppl.7
1987
Toluene
2000
EHC 52
1986
2003 2000
1994
being
reassessed
acute &
chronic
1999
UBA 1996
d
Vol. 71
1999
Xylenes
EHC 190
1997
1995 2003
acute &
chronic
1999
Vol. 71
1999
Styrene
2000
EHC 26
1983
1992
1993
being
reassessed
acute &
chronic
1999
Vol. 82
2002
Naphthalene
2003
2003
draft
1998
being
reassessed
chronic
1999
UBA 2004
e
Vol. 82
2002
Limonene
CICADS
5
1998
1993 NICNAS 2002
Vol. 73
1999
α-Pinene
UBA 2003
f
NIEHS 2002
g
a
including: Evaluation of current California Air Quality Standards with respect to protection of children (2000)
b
including: EPA-Toxicological review of benzene/noncancer effects (2002) and EPA-Carcinogenic Effects of Benzene: An Update (1998)
c
National Institute for Working Life - The Nordic Expert Group for Criteria Documentation of Health Risks from Chemicals and The Dutch Expert
Committee on Occupational Standards: 132. Formaldehyde - Anton Wibowo
d
Umweltbundesamt: Bundesgesundheitsblatt 11/96: Richtwerte für die Innenraumluft: Toluol. H.Sagunski,
e
Umweltbundesamt: Bundesgesundheitsblatt 7 · 2004: Richtwerte für die Innenraumluft: Naphthalin. H.Sagunski, W.Heger
The INDEX project Final report
24
f
Umweltbundesamt: Bundesgesundheitsblatt 4 · 2003: Richtwerte für die Innenraumluft: Bicyclische Terpene (Leitsubstanz α-Pinen). H.Sagunski,
B.Heinzow
g
National Institute of Environmental Health Sciences, Toxicological Summary For Turpentine, Review of Toxicological Literature, February 2002
Table 3.1.3.2: Exposure limits/guidelines established by health organisations and their summary definitions
World Health Organization - Air Quality Guidelines for Europe
Guidelines: The term “guidelines”, in the context of the WHO - Air Quality Guidelines for Europe, implies not only numerical values (guideline
values), but also any kind of guidance given. Accordingly, for some substances the guidelines encompass recommendations of a more general nature
that will help to reduce human exposure to harmful levels of air pollutants. For some pollutants no guideline values are recommended, but risk
estimates are indicated instead.
The starting point for the derivation of guideline values is to define the lowest concentration at which adverse effects are observed. On the basis of the
body of scientific evidence and judgements of uncertainty factors, numerical guideline values were established to the extent possible. Compliance
with the guideline values does not, however, guarantee the absolute exclusion of undesired effects at levels below the guideline values. It means only
that guideline values have been established in the light of current knowledge and that uncertainty factors based on the best scientific judgements have
been incorporated, though some uncertainty cannot be avoided. The numerical values for the various air pollutants should be considered in the context
of the accompanying scientific documentation giving the derivation and scientific considerations. Any isolated interpretation of numerical data should
therefore be avoided, and guideline values should be used and interpreted in conjunction with the information contained in the appropriate sections.
Guidelines based on carcinogenic effects are indicated in terms of incremental unit risks in respect of those carcinogens that are considered to be
genotoxic. To allow risk managers to judge the acceptability of risks, this edition of the guidelines has provided concentrations of carcinogenic air
pollutants associated with an excess lifetime cancer risk of 1 per 10 000, 1 per 100 000 and 1 per 1 000 000.
Agency for Toxic Substances and Disease Registry (ATSDR)
Minimal Risk Levels (MRLs): During the development of toxicological profiles, Minimal Risk Levels (MRLs) are derived when reliable and sufficient
data exist to identify the target organ(s) of effect or the most sensitive health effect(s) for a specific duration for a given route of exposure. An MRL is
an estimate of the daily human exposure to a hazardous substance that is likely to be without appreciable risk of adverse noncancer health effects over
a specified duration of exposure. MRLs are based on noncancer health effects only and are not based on a consideration of cancer effects. These
substance-specific estimates, which are intended to serve as screening levels, are used by ATSDR health assessors to identify contaminants and
potential health effects that may be of concern at hazardous waste sites. It is important to note that MRLs are not intended to define clean-up or action
levels. MRLs are derived for hazardous substances using the no-observed-adverse-effect level/uncertainty factor approach. They are below levels that
might cause adverse health effects in the people most sensitive to such chemical-induced effects. MRLs are derived for acute (1–14 days),
intermediate (15–364 days), and chronic (365 days and longer) durations and for the oral and inhalation routes of exposure.
U.S.EPA - Integrated Risk Information System (IRIS)
The inhalation Reference Concentration (RfC) is analogous to the oral RfD and is likewise based on the assumption that thresholds exist for certain
toxic effects such as cellular necrosis. The inhalation RfC considers toxic effects for both the respiratory system (portal-of-entry) and for effects
peripheral to the respiratory system (extrarespiratory effects). It is expressed in units of mg/cu.m. In general, the RfC is an estimate (with uncertainty
spanning perhaps an order of magnitude) of a daily inhalation exposure of the human population (including sensitive subgroups) that is likely to be
without an appreciable risk of deleterious effects during a lifetime. Inhalation RfCs were derived according to the Interim Methods for Development
of Inhalation Reference Doses (EPA/600/8-88/066F August 1989) and subsequently, according to Methods for Derivation of Inhalation Reference
Concentrations and Application of Inhalation Dosimetry (EPA/600/8-90/066F October 1994). RfCs can also be derived for the noncarcinogenic
health effects of substances that are carcinogens. Therefore, it is essential to refer to other sources of information concerning the carcinogenicity of
this substance. If the U.S. EPA has evaluated this substance for potential human carcinogenicity, a summary of that evaluation will be contained in
Section II of this file.
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency (OEHHA)
The concentration, at or below which no adverse health effects are anticipated in the general human population, is termed the reference exposure level
(REL). RELs are based on the most sensitive relevant adverse health effect reported in the medical and toxicological literature. RELs are designed to
protect the most sensitive individuals in the population by the inclusion of margins of safety. Protection against carcinogenicity and against adverse
health effects of short-term exposures are not considered in these guidelines. For this reason, chemicals should be evaluated separately for their
carcinogenic potential and additional acute health effects that may occur.
Methods for the evaluation of acute and chronic health effects and for the carcinogenic potential of chemicals are provided in the OEHHA documents
entitled Air Toxics Hot Spots Program Risk Assessment Guidelines:
Acute Reference Exposure Levels (RELs): three categories of acute severity levels are developed in accordance with criteria established by NRC
(1993): the level protective against mild adverse effects, the level protective against severe adverse effects, and the level protective against life
threatening effects. Each of these three acute exposure levels is determined for one-hour exposure duration. However, the major focus of this
document is in developing acute RELs for the preparation of risk assessments for non-emergency routine releases. Thus, the RELs used in the risk
assessment are generally levels protective against mild adverse effects; a few are based on severe effects (e.g., reproductive/developmental).
Chronic Reference Exposure Levels (RELs): Chronic reference exposure levels are concentrations or doses at or below which adverse health effects
are not likely to occur. A central assumption is that a population threshold exists below which adverse effects will not occur in a population; however,
such a threshold is not observable and can only be estimated. Areas of uncertainty in estimating effects among a diverse human population exposed
continuously over a lifetime are addressed using extrapolation and uncertainty factors.
Federal Environmental Agency of Germany (Umweltbundesamt – UBA)
In Germany, an important framework for the setting of indoor-related guideline values is given by the building codes (which are under the jurisdiction
of the German States). The building codes demand that there be no health hazard to occupants from the building. Hence the work of the ad-hoc group
focused on defining concentration levels at which such hazard would probably occur. The introduction of a safety margin would then allow the
definition of a concentration where there would be no more concern for adverse health effects. The following two concentration levels were defined:
- Guideline Value II (GV II): GV II is a health-related value based on current toxicological and epidemiological knowledge. If the concentration
corresponding to GV II is reached or exceeded immediate action must be taken because permanent stay in a room at this concentration level is
likely to represent a threat to health, especially for sensitive people. In this context, taking action means an immediate examination of the situation
with regard to a need for control measures. It may include the evacuation of the room in question. If by measurement GV II has been found to be
The INDEX project Final report
25
exceeded, the results should be checked by repetitive measurements carried out immediately under normal conditions of occupancy. If possible and
deemed meaningful, biological monitoring of the occupants should be carried out in addition.
- Guideline Value I (GV I): GV I is the concentration level at which a substance – taken individually – does not give rise to adverse health effects
even at life-long exposure. An exceedance of GV I is linked with an exposure beyond normal which is undesirable from a hygienic viewpoint.
Thus, there is also a need for action at concentrations between GV II and GV I. GV I is obtained by dividing GV II by a factor of 10. This factor is
a convention. However, for odorous substances GV I must be defined based on the odour threshold (“odour detection”) if the odour threshold has a
lower numerical value than the concentration derived according to the general scheme. GV I can be used as the level to be reached after control
measures have been applied. The level should not be “filled up”; rather, the final concentration should fall below.
Environment Canada - Health Canada (EC-HC)
Different approaches were adopted for assessments of Priority Substances for chemicals for which the critical effect is believed to have a threshold
and those for which it is considered not to have a threshold. For substances for which the critical effect is considered to have a threshold (i.e., non-
neoplastic effects), Tolerable Daily Intakes (TDIs) or Tolerable Concentrations (TCs) have been developed by dividing effect levels observed in
studies in exposed populations or animal species, by uncertainty factors.
Priority Substances are classified into one of 6 categories, based on the weight of evidence of carcinogenicity (see Section 3). For genotoxic
carcinogens (i.e., primarily compounds which are considered “carcinogenic to humans” or “probably carcinogenic to humans”, Groups I or II of the
scheme for classification of carcinogenicity under CEPA), quantitative estimates of the carcinogenic potency within or close to the experimental
range have been developed. This approach was adopted for several reasons, one of the most important of which was to avoid expressing risk in
precise absolute terms (i.e., predicted excess numbers of cancers per unit of the population) based on uncertain low-dose extrapolation procedures.
Tolerable Concentration (TC): Tolerable concentrations (Section 3a) (often expressed in mg/m3) are generally airborne concentrations to which it is
believed that a person can be exposed continuously over a lifetime without deleterious effect. They are based on non-carcinogenic effects.
Tumorigenic Concentration 05 (TC05): The Tumorigenic Concentration 05(TC05) is the concentration generally in air (expressed, for example, in
mg/m3) associated with a 5% increase in incidence of, or deaths due to, tumours considered to be associated with exposure, observed in
epidemiological studies in human populations or bioassays in experimental animals Values derived based on division of the TC05s by a suitable
margin (e.g., 5.000 to 50.000)* can provide a benchmark against which the adequacy of indoor or ambient air can be judged, with respect to potential
carcinogenicity.
It should be noted that Health Canada does not necessarily deem as “acceptable” from a societal viewpoint health risks associated with these values
and that the Health Protection Branch continues to subscribe to the position that exposure to substances for which the critical effect has no threshold
be reduced to the extent possible.
* Since Tumorigenic Concentration05s were computed directly from the curve within or close to the experimental region, division by an additional
factor of 2 would equate approximately to the lower 95% confidence limit.
Key-studies were summarised among each chapter treating effects of short- and long-term exposure and itemised in
tables at the end of the chapter. One-page fact sheets resuming the most relevant toxicological properties are given at
the end of each D/R assessment chapter. In the key-study Tables, subscripts were assigned to effect levels (NOAELs
and LOAELs), stating on whether occupational average levels or experimental concentrations are quoted or identifying
the extrapolation process applied for the given value. Details on these subscripts are given in Table 3.1.3.3.
Table 3.1.3.3: Subscripts used in summary tables and fact sheets quoting key-studies
and the exposure limit derivation process accounted for by health organisation
Subscript Description Details
Study
average concentration time weighted average concentration with no information on
maximum concentrations, in chronic studies generally estimated
from numerous intermittent measurements in occupational settings,
even over years
EXP
experimental concentration concentrations artificially generated in inhalation/exposure chambers
in animal or volunteer studies
ADJ
concentration adjusted from an intermittent to a
continuous exposure
When extrapolating from occupational to population based
exposures on a [hour/day and days/7 days] basis (generally: division
by 4.2)
1h-ADJ
concentration adjusted to 1-hour exposure duration For the extrapolation from sub-acute to acute
HEC
human equivalent concentration Generally a blood-to-air partition coefficient of the chemical for the
experimental animal species was used in the HEC derivation of an
RfC
MLE
maximum likelihood estimate for 5% response A statistical best estimate of the value of a parameter from a given
data set.
BC05
BC05 is the 95% lower confidence limit of the
concentration expected to produce a response rate
of 5%
Following a Benchmark (BM) approach, alternative to the traditional
NOAEL/LOAEL approach. A Benchmark Concentration (BMC) is a
statistical lower confidence limit on the dose producing a
predetermined, altered response for an effect.
STAT
lowest statistically significant effect concentration
The INDEX project Final report
26
3.1.4 Risk Characterization and prioritisation of chemicals
In this final step of the general risk assessment process, the incidence of health hazards and risks in the European
population, associated with indoor exposure to individual chemicals, was estimated. It is pointed out that the assessment
of risk is a scientific one, which has been kept separate from any consideration regarding the risk management process,
including the setting or the proposal of Indoor Exposure Limits. Namely, an important uncertainty, not accounted for in
the assessment, is the possibility of antagonistic and synergistic effects arising from the exposure to mixtures of
chemicals, little scientific information existing in this area. Multiple contaminants are typically occurring in indoor
environments (although at low concentrations) and the resulting uncertainty (uncontrolled factor) should be taken into
consideration in the risk management.
Nevertheless, Limits of Exposure (ELs) had to be established in order to perform the risk characterization associated
with individual chemicals, following inhalatory exposure in indoor environments, for both short-term (indoor-activity
related) and long-term exposures (background indoors).
An EL was derived for each chemical after the identification of key-studies (critical-study) describing the appropriate
toxicological endpoints (among those selected by health organizations for the derivation of health based reference
concentrations).
Uncertainty factors (here named assessment factors, AF) applied in the present assessment to account for the adequacy
of the critical study are the product of the individual factors outlined in Table 3.1.4.1.
Table 3.1.4.1: Elements considered for the derivation of Uncertainty (Assessment) Factors (AS)
Description Detail Factor
Extrapolation from a LOAEL to a NOAEL When in the critical study no NOAEL could be observed 10
Interspecies extrapolation
Critical study = experimental animal study
(no human study available or appropriate)
10
Inter-interindividual (intraspecies) variability
in humans
Always, unless the critical study was performed on individuals of the sub-
population considered susceptible
10
Susceptible population
asthmatic individuals, infants, children, individuals with heart diseases,
individuals with (hereditary) enzyme deficiencies, pregnant women
10; 3; 2
Adequacy or quality of toxicological data Old study 2
Extrapolation from sub-acute to chronic Deficiencies in toxicological database 10
Extrapolation from sub-acute to acute Deficiencies in toxicological database 10
For all chemicals a threshold-level of action could be identified, enabling a “no-observed-adverse-effect level
(NOAEL)/assessment factor (AF)” approach, i.e. EL derived by dividing the critical effect level by the AF, with the AF
based on the available scientific evidence. Where no NOAEL observation was documented, a lowest-observed-adverse-
effect level (LOAEL) was taken into consideration and an additional assessment factor of 10 used for EL derivation.
For one compound only (benzene) the characterization has been based on population cancer risk estimation rather than
on an EL.
In those cases where large differences in sensitivity for different susceptible groups were documented, a bimodal
distribution of population responses was supposed to exist and a tenfold difference in sensitivity, usually accepted as
higher than the encountered range, taken in account in the AF derivation. Susceptible subpopulations considered in the
present characterization are: asthmatic individuals, infants, children, individuals with heart diseases, individuals with
(hereditary) enzyme deficiencies, pregnant women.
Information from the exposure assessment (Chapters 1-3) and toxicity assessment (Chapters 4-5) were integrated and a
risk characterization performed for each chemical (Chapter 6). Based on the conclusions of the assessments and on the
completeness of individual databases a priority ranking of the 14 chemicals was established with the chemicals assigned
to one of three groups, and the results of the assessment handed over to Risk Management.
Group 1: High priority chemicals
Group 2: Low priority chemicals
Group 3: Chemicals requiring further research with regard to human exposure or dose response
The INDEX project Final report
27
3.2 Risk Management
Buildings are built for shelters from the cold (or heat), wind and rain of the outdoor air. Typically indoor air contains
higher levels of most air pollutants than outdoor air. As long as people feel that they are in control of their own indoor
environment, however, there is usually a higher tolerance to poor indoor air quality compared to outdoor air quality -
after all one can always go outdoors or open a door or window. This tolerance is greatly decreased if self-control is
lacking.
Risk assessment disentangles an identified risk complex (e.g. intoxication from a high indoor CO concentration peak)
into its details (e.g. formation of CO in a faulty combustion device, release of CO into room instead of chimney,
exposure to CO in the room, absorption to bloodstream in the lung, subsequent reduction of oxygen delivery, and
hypoxia), and analyses these details one by one and in relation to each other. Risk management must entangle these
details back again into an efficient and enforceable policy to reduce the risk (e.g. building codes requiring combustion
devices to be vented to chimneys, regular maintenance & inspections, CO-alarms, etc.).
While risk assessment is a science based exercise, risk management by necessity involves also technical, economic,
social and legal realities, as well as need for actions in the face of incomplete information. Risk management decisions
concerning a specific risk are based on one hand on risk characterisation, produced by risk assessment, and on the other
hand on the political and administrative processes that generate, evaluate, select and implement policies in the society
(EU Commission directive 93/67/EEC). The indoor air risk management options include, in principle:
IAQ Standards and guidelines
Building codes and ventilation standards
Equipment standards and permits
Mandatory maintenance and inspections
Limits, labelling and reporting of the contents of or releases from building products, furnishing materials,
equipment and consumer products
Public awareness raising and information
These alternatives will be discussed separately for each of the final shortlist components in Chpt. 6.1. The following
discussion looks at indoor air risk management in general.
The core of ambient air risk management is in the ambient air quality guidelines and standards. The ambient air
concentration limits have questionable applicability for indoor environments, because they are derived for different
pollution mixtures and exposure patterns, because comparable monitoring and implementation alternatives do not exist
indoors, and because indoor air quality for an individual may depend strongly on the behaviour of the same individual.
When ambient air quality standards are applied for indoor air quality (as determined by indoor sources and activities),
short term (24 h) measurements of indoor pollutants should in most cases be related to long term (annual) outdoor air
guidelines.
The occupational exposure limits (OEL, TLV, MAK, etc.), although derived for indoor environments, are not directly
applicable either, because they have been developed for healthy adult populations and more or less controlled 40 h
exposure in a 168 h week.
Risk managers can intervene in many points (that lead from the primary source of risk to its materialisation). They can
prevent the hazardous process, reduce exposures, modify effects, alter perceptions or valuations through education and
public relations or compensate for damage after the fact. (Morgan 1993).
The issue, which is most specific for indoor air risk management, becomes obvious by visualising the millions of indoor
environments in just one city, and the daily multitudes of potential hazards in them. A vast majority of both indoor air
risk observations (assessments) as well as decisions (management) will remain to be made by occupants themselves -
and this is particularly true for the acute effects from short term hazardous exposures. No imaginable public service or
professional expert resources can ever cover but a tip of the iceberg of all hazardous IAQ situations. At best there are
enforceable governmental regulations, respected industrial/professional standards/practices and/or authoritative
examples.
In contrast to ambient air quality management, which relies on air quality standards and monitoring networks, the
capabilities of public services and experts to manage most indoor air risks lie crucially on their abilities to establish and
maintain public confidence, to communicate the risks in a way that the public is able to comprehend, to relate to their
own living conditions and willing to accept, and to present practical solutions which are technically, financially and
legally feasible, and which sufficiently acknowledge the diversity of the buildings, the occupants, expectations and
risks.
The largest investments in the life of most individuals are their homes, and buildings represent in the order of 2/3 of
national property in most European countries. This fact sets very concrete requirements for any interventions into the
The INDEX project Final report
28
residential building stock, as well as for the allocation of financial burdens and benefits. The fact that most indoor
environments are private property and protected by privacy of the individuals significantly limits the options of public
indoor air risk management.
The public services can, however, rely more on standards and regulations in managing risks in rented apartments,
hotels, public buildings, workplaces, schools, hospitals, shopping malls etc. These buildings should be ensured to be
safe for a great majority of the population (excluding only exceptionally sensitive individuals) by public health,
occupational health and building inspection authorities as the last resort. When credible concerns arise they should be
addressed promptly and properly for the sake of public health, confidence and liability. One of the most important
special tasks for the public services and experts is prediction, identification, assessment and management of the
situations where susceptible population groups and risky indoor environments coincide.
Risk - benefit: In managing risks from indoor air pollutants, there is a case for erring on the side of caution and acting
to reduce risks if there is any reasonable suspicion that a hazard exists. However:
- any chemical can be hazardous at sufficient concentration, and yet attempt to reduce all measurable concentrations
to zero is not feasible;
- blind application of precaution may divert resources into irrelevant actions, and thus lead to sub-optimal protection
of health and/or the environment. It may also unjustifiably deny the benefits of using products or techniques; and
- consideration should be given to the fact that limiting the use of one chemical may entail the application of a
substitute that may not have lower, but only less well-characterised risks.
Hence, legislation in Europe has moved towards approaches based on overall risk rather than mere hazard
identification. Article 10 of the Council Regulation (EEC) 793/93 (Existing Substances Regulation) requires that:
"Where such control measures include recommendations for restrictions on the marketing and use of the substance in
question the rapporteur" [on behalf of the body carrying out the risk assessment] "shall submit an analysis of the
advantages and drawbacks and the availability of replacement substances".
Risk Communication: Risk communication is by far the most important tool for the society to manage indoor air
health risks. Successful communication will not raise undue concerns, but suggests behaviour, which reduces avoidable
risks, yet restricts nonessential interference into the lives of the individuals or to the alternatives of the enterprises. Poor
risk communication may not only fail to produce the wanted risk reduction, it may also unnecessarily limit individuals'
options, add public and private costs, and generate new risks due to non-productive public concerns and potentially
harmful behavioural changes.
The essence of good risk communication is simple: Learn what people believe and the decisions that they are facing and
tailor the language and message to this knowledge. Increased (decreased) trust in the managing organisation of the
activity under concern - including indoor air risk management - will lead to lower (higher) levels of perceived risk and
thus increase public support (opposition). Understanding the dimensions of trust is essential for developing policy
strategies that will gain public acceptance. (Flynn et al. 1992)
Economically and technically sound [indoor air risk management] policies may be doomed if people believe that they
distribute benefits and burdens unfairly. Three concepts emerge from the philosophical and cultural basis of risk
sharing: parity, in which each individual, group or country is treated equally; priority, or giving the burden to those
most deserving of it; and proportionality, or the sharing of burdens according to need or contribution. (IIASA 1993)
The INDEX project Final report
29
4. Risk Assessment of the selected chemicals
The exposure and dose/response data reviewed during the project are presented in this chapter. The summary of the
typical concentrations in Europe for the selected compounds are presented in Table 4.0.1. To be able to compare
recently published results reviewed in this report to the ‘normal’ indoor concentrations defined by the Working Group
of WHO (WHO 1989), these concentrations are summarised in Table 4.0.2.
Table 4.0.1. Typical European microenvironmental and exposure concentrations (µg/m
3
, except CO in mg/m
3
)
summarised from population based studies reviewed in this report.
In
1
W
1
Out
1
P
1
Aromatics
Benzene 2-13 4-14 1-21 3-23
Naphthalene 1-90 2-8 1-4 2-46
Styrene 1-6 3-7 1-2 1-5
Toluene 15-74 25-69 3-43 25-130
m&p-Xylenes 4-37 25-121 2-23 25-55
o-Xylene 2-12 7-29 1-8 8-15
Aldehydes
Acetaldehyde 10-18 3 1-2 8
Formaldehyde 7-79 12 2-4 21-31
Terpenes
a- Pinene 11-23 1-17 1-7 7-18
Limonene 6-83 11-23 5-9 19-56
Classical pollutants
CO 0.5-1 1 2 0.8-1.7
NO
2
13-62 27-36 24-61 25-43
1
In, W, Out, P = Indoor, workplace, outdoor and personal exposure concentrations
Table 4.0.2. ‘Normal ‘residential indoor air concentrations of selected organic pollutants reviewed by a Working Group
of WHO in 1987 (adopted from WHO 1989 and Maroni et al 1995).
Pollutant 10% median 90%
A
M Sources
Aromatics
Benzene 2 10 20 10 fuel component, tobacco
Naphtalene 2 5 solvent moth balls
Styrene < 1 1 5 fuel component
Toluene 30 65 150 80 fuel component, solvent
m,p-xylene 10 20 40 20 fuel component, solvent
o-xylene 3 5 10 10 fuel component, solvent
Aldehydes
acetaldehyde 10 30 cigarette smoke
formaldehyde 25 60 40 chipboard, urea-formaldehyde insulation
Terpenes
a-pinene 2 10 20 10 wax, wood product
limonene 2 15 70 30 odorant, detergent
10% and 90% = ith percentile, AM = arithmetic mean
Concentration (µg/m
3
)
The INDEX project Final report
30
Formaldehyde
Synonyms: Formalin, methanal, oxoymethane, oxomethylene, methylene oxide, formic
aldehyde, methyl aldehyde
CAS Registry Numbers: 50-00-0
Molecular Formula: CH
2
O
1. Compound Identification
Formaldehyde is a colourless, odorous, highly reactive, flammable and easily polymerised gas at room temperature and
pressure. It is formed by oxidation or incomplete combustion of hydrocarbons. Formaldehyde is soluble in water,
ethanol and diethyl ether and is used in solution or in polymerised form (paraformaldehyde). In atmosphere
formaldehyde is photo-oxidised in sunlight to carbon dioxide. Its half-life in urban air, under the influence of sunlight,
is short. In the absence of nitrogen dioxide, the half-life of formaldehyde is approximately 50 minutes during the
daytime, but in the presence of nitrogen dioxide, it decreases down to 35 minutes (WHO 1989).
In solution formaldehyde has a wide range of uses: in the manufacture of resins and textiles, as a disinfectant, and as a
laboratory fixative or preservative in consumer products, such as food, cosmetics, and household cleaning agents (SIS
2003, EPA/Cal 2003).
Formaldehyde is used commonly in homes as an adhesive resin in pressed wood products. There are two types of
formaldehyde resins: urea formaldehyde (UF) and phenol formaldehyde (PF). Products made of urea formaldehyde
readily release formaldehyde. Urea-formaldehyde foam is used to insulate buildings. It can continue to emit
formaldehyde after installation or constituting a source of persistent emission. Formaldehyde is used also in phenolic
and polyacetal plastics, but they are not expected to release formaldehyde (WHO 1989, EPA 2002).
2. Physical and Chemical Properties
Molecular weight (g/mol) 30.03
Melting point (°C) -118
Boiling point (°C) -19.2
Density (g/l at 20 °C, 1 atm) 0.815
Relative density (air =1) 1.04
Explosivity range in air (vol %) 7-73
Solubility: Soluble in water, ethanol, ether, and acetone
Conversion factors at 20 °C and 760 mm Hg:
1 ppb = 1.248 µg/m
3
1 µg/m
3
= 0.815 ppb
Sources: (CRC, 1994, WHO 1989, Verschueren 2001, HSDB 2003,)
3. Indoor Air Exposure Assessment
Emission sources
There are several indoor sources that can result in increased human exposure to formaldehyde including cigarette
smoke, insulating materials, particle board or plywood furniture containing formaldehyde-based resins, water based
paints, fabrics, household cleaning agents, disinfectants, pesticide formulations, paper products and adhesives
containing formaldehyde used for plastic surfaces, parquet, carpets and other building materials containing urea-
formaldehyde resins. Also gas cookers and open fireplaces emit formaldehyde to indoor air (WHO 1989, COMEAP
1997, Jurvelin et al 2001, EPA/Cal 2003).
The INDEX project Final report
31
There might be also formaldehyde sinks in indoor environment. Zwiener et al (1999) reported reduction of
formaldehyde concentration between 80% and 87% in chamber air in two hours after the test had begun. This reduction
is due to chemical reaction of formaldehyde and the wool proteins.
Building age was identified a factor for indoor air formaldehyde concentration in UK (Brown VM et al 2002). Indoor
air concentration increased with decreasing age, having highest concentrations in buildings built in 1990’s. A source
identified in this study was particleboard flooring. These findings show that formaldehyde in indoor air is still an issue
in European homes and should not be neglected in the forthcoming indoor air surveys.
Emission rates of 55 diverse materials and consumer products were determined in a chamber study in California (Kelly
et al 1999). The tests showed that among dry products highest formaldehyde emissions were found from bare pressed-
wood materials made with urea-formaldehyde (UF) resins, and from new permanent press fabrics. An acid-cured floor
finish showed the highest emissions from wet products, clearly exceeding those of any dry product.
Puhakka and Karkkainen (1993) studied factors that increased formaldehyde concentrations in indoor air. Their results
showed the amount of chipboard and the air exchange efficiency in a room being the dominant factors. Formaldehyde
emissions from chipboard could be reduced by painting the surfaces. On the other hand elevated relative humidity
increased formaldehyde emissions significantly.
Increased formaldehyde concentrations may be present in hospitals and scientific facilities where formaldehyde is used
as a sterilising and preserving agent, and in living spaces, such as schools, kindergartens, and mobile homes or
apartments where there may be uncontrolled emissions of formaldehyde from tobacco smoking, building materials, and
furniture (WHO 1989).
A French study carried out in 61 recently refurbished ats with no previous history of complaint for odour or specic
symptoms, showed that aldehyde levels of formaldehyde, acetaldehyde, pentanal, and hexanal were depend on the age
of wall or oor coverings (renovations less than 1 year old), smoking, and ambient parameters such as carbon dioxide
levels and temperature. Geometric mean and geometric standard deviation of formaldehyde concentrations in bedrooms
were 24.5 µg/m
3
and 2.0 respectively (Clarisse et al 2003).
Formaldehyde is formed naturally by photochemical reactions in the atmosphere. It is also emitted by fuel combustion
such as motor vehicles and by industrial fuel combustion such as power generators, oil refining processes, copper
plating, and incinerators (EPA/Cal 2003).
Indoor air and exposure concentrations
A large-scale population based indoor air survey has been carried out in UK (Brown VM et al 2002). Geometric mean
and the maximum value of 3-day samples (n=833) were 22.2 µg/m
3
and 171 µg/m
3
, respectively. In Helsinki, average
indoor air concentration of formaldehyde, 41.4 µg/m
3
, was more than tenfold compared to the respective ambient
concentration. Average personal exposure, 26.8 µg/m
3
, was slightly lower than the respective residential indoor
concentration, but anyway clearly higher than the workplace concentration, 15 µg/m
3
. In Sweden (Gustafson et al 2004)
average 24-hour personal exposure (n=24, AM=47 µg/m
3
, std=111) was higher than the respective indoor air (bedroom)
concentration, (n= 24, AM= 26 µg/m
3
, std = 14). In another campaign of the same study, average 6-day exposure (n=40,
AM=31 µg/m
3
, std=18) was slightly higher than the respective indoor air concentration (n=40, AM=35 µg/m
3
, std=22).
Both of these levels were about an order of magnitude higher than respective outdoor air concentration (n=20, AM=3.8
µg/m
3
, std=1.2). These results show the dominating role of residential indoor emissions for population exposure to
formaldehyde.
In a German study (GerES IV) weekly mean formaldehyde concentration in residential indoor air in a randomly
selected population of children and teenagers was 36 µg/m
3
(n=58) (Ullrich et al 2002). This level was only slightly
lower than the respective concentrations in last few years demonstrating the limitations of the source control actions.
A large-scale population based formaldehyde study was also carried out in Austria (Hutter et al 2002). Formaldehyde
concentrations in Austrian homes ranged from 8.8 µg/m
3
to 115 µg/m
3
(n= 160, AM= 31.3 µg/m
3
and median =25
µg/m
3
).
Sakai et al (2004) compared indoor air concentrations of formaldehyde in Swedish and Japanese dwellings, showing
higher geometric mean values in Japan than in Sweden being, 17.6 µg/m
3
(n=37) and 8.3 µg/m
3
(n=27) respectively.
The highest concentration was measured in a detached house with an unvented kerosene heater. In general,
The INDEX project Final report
32
formaldehyde concentrations were higher in newer (< 10 years) dwellings than in older ones, possibly due to less
natural ventilation and more emissions from building materials.
In Paris, geometric mean and geometric standard deviation of formaldehyde concentrations in recently refurbished ats
in bedrooms were 24.5 µg/m
3
and 2.0 respectively (Clarisse et al 2003). In a study of 174 homes in UK, the annual
mean indoor concentration ranged from 7 µg/m
3
to 76 µg/m
3
, all of them being clearly higher than the mean annual
outdoor concentration of 2 µg/m
3
(COMEAP 1997). In an Italian study (Cavallo et al 1993), formaldehyde
concentrations in schools and office buildings ranged 8 – 210 µg/m
3
and 1.5 – 71 µg/m
3
respectively. VOC emissions
were related to cleaning materials in schools and to carpeting and furniture in offices. Based on the results of a
European survey indoor concentration of formaldehyde in homes and schools ranged from 10 µg/m
3
to 100 µg/m
3
(COMEAP 1997). Similar concentration levels of formaldehyde were reported by WHO in conventional homes with no
urea-formaldehyde foam insulation ranging from 25 to 60 µg/m
3
(WHO 2000).
In 1970’s and 1980’s residential indoor formaldehyde concentrations were high, and therefore formaldehyde in indoor
air became an issue. It has been suggested to be due to off-gassing from new materials and decreased ventilation in
order to decrease energy consumption of house heating. Maximum formaldehyde concentrations were measured in new
mobile homes in 1980’s showing formaldehyde concentration of several mg/m
3
(WHO 2000). In the NHEXAS study in
Arizona, USA (Gordon et al 1999) median and 90
th
percentile indoor concentrations, 21 µg/m
3
and 46 µg/m
3
, were
about the same levels than those measured in EXPOLIS in European cities. In a Taiwanese study (Li et al 2002), mean
8-hour formaldehyde concentrations in offices ranged 75 µg/m
3
– 300 µg/m
3
. Maximum short time concentrations were
as high as 1000 µg/m
3
based on the continuous monitoring. Building materials were identified as the main contributor
to these high formaldehyde concentrations. Lee and Wang (2004) studied emissions of incense burning in chamber (18
m
3
) tests. They found that six tested incense types caused concentrations exceeding the Recommended Indoor Air
Quality Objectives for Office Buildings and Public Places in Hong Kong (HKIAQO) of 100 µg/m
3
for formaldehyde.
Maximum concentration exceeded 250 µg/m
3
during incense burning.
Brown et al (2002) studied emissions and concentrations caused by unflued gas heaters in a room chamber of 32.4 m
3
.
In normal conditions formaldehyde concentrations ranged from <10 µg/m
3
to 180 µg/m
3
. When the gas supply was
restricted due to the backpressure, as high as 2100 µg/m
3
of formaldehyde was measured.
Elevated formaldehyde concentrations, up to 375 µg/m
3
, in manufactured houses in US were related to engineered
wood products such as particleboard and plywood bonded with urea-formaldehyde resin in 1970’s (Sexton et al 1989).
Recently measured formaldehyde concentrations in site-built houses were clearly lower, ranging from 8 – 66 µg/m
3
and
33 – 81 µg/m
3
prior to occupancy and five months after occupancy respectively (Lindstrom et al 1995). Similar results
were reported by Hodgson et al (2000) showing indoor concentrations in site-built houses in eastern United States,
ranging from 17 µg/m
3
to 73 µg/m
3
. Cabinetry materials, passage doors and plywood subfloor were the predominant
sources of formaldehyde in a new manufactured house studied by Hodgson et al (2002).
Studies in Denmark (Granby and Kristensen 1997), Finland (Viskari et al 2000), and Italy (Possanzini et al 1996)
showed average (3-8 months periods) ambient concentrations of formaldehyde being 3.3 µg/m
3
, 1.3 – 2.8 µg/m
3
, and
13.7 µg/m
3
, respectively. In the USA ambient concentrations ranged from 3.3 to 3.7 µg/m
3
(Grosejan et al 1993, CARB
1999). In Chinese studies ambient levels ranged from 4.1 µg/m
3
to 13.3 µg/m
3
, while the mean indoor concentration of
formaldehyde in hotels was more than twofold (Ho et al 2002, Feng et al 2004). Relatively high ambient concentrations
were found in Japan, (Satsumabayashi et al 1995), Brazil (Grosejan et al 2002), Greece (Bakeas et al 2003) and in
Mexico (Baez et al 1995) 3.1–14 µg/m
3
, 10.8 µg/m
3
, 17.2 µg/m
3
and 43.5 µg/m
3
, respectively.
Cumulative distributions of the indoor and personal exposure concentrations in Europe are presented in Figure 3.1 and
Figure 3.2. Indoor air concentrations of formaldehyde in European countries before 1990 are summarised in Table 3.1
and short time peak concentrations in Table 3.2.
The INDEX project Final report
33
Table 3.1. Indoor air concentrations of formaldehyde measured before 1990 (adopted from ECA 1990).
Country Environment
Expected source
A
Mmax
Denmark apartments 9.9
Germany dwellings 48-hour 56 279
Greece dwellings 30-minute 6 - 9 22
France nursery school 30-minute 29
dwellings 24-hour 22 < 70
apartments with complaints 600 2800 Particleboard
collective sites with complaints 3000 UFFI
Norway dwellings < 60 110 Particleboard
Switzerland dwellings 480 2760 UFFI
new buildings 840 Particleboard
The Netherlands 50% of the dwellings with complaints > 120 1000
66% of the schools with complaints > 120 2500
UK buildings without UFFI 57
buildings with UFFI 114 UFFI
AM = aritmethic mean, max = maximum value, UFFI = urea formaldehyde foam insulation
Concentration (µg/m
3
)Averaging time
Table 3.2. Short time formaldehyde concentrations related to specific microenvironments or emission sources.
Averaging time Reference
A
MGMmax
Unflued gas heater Room chamber tests Brown et al 2002
- normal burning <10 - 180
- gas supply restricted 2100
Offices Taiwanese study 8-hour 75 - 300 Li et al 2002
peak 2000
Incense burning chamber tests 250 Lee and Wang 2004
Public buildings Schools 5-hour 8 - 210 Cavallo et al 1993
Offices 5-hour 1.5 - 71
Homes in UK with UF foam insulation 30 - 60 min 130 Brown et al 1993
without UF foam insulation 31 - 60 min 70
Houses in US manufactured 30-min 26 - 59 45 Hodgson et al 2000
site-built 30-min 17 - 73 43
Urban dwellings Sweden 24-hour 8.3 19 Sakai et al 2004
Japan 24-hou
r
18 73
AM = arithmetic mean, GM = geometric mean
Environment or
emission source
Concentration (µg/m
3
)
The INDEX project Final report
34
0
20
40
60
80
100
0 102030405060708090
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Hel
UK
Fre
Swe
Figure 3.1. Cumulative frequency distributions of indoor air concentrations of formaldehyde in Helsinki (Hel, n=15,
Jurvelin et al 2001), in England (UK, n=833, Brown VM et al 2002), in the French Survey (Fre, n=201, Kirchner 2004),
and in Sweden (Swe, n=40, Gustafson et al 2004).
0
20
40
60
80
100
-20 0 20 40 60 80 100 120 140 160 180 200
Exposure (µg/m
3
)
Cumulative frequency (%)
Hel
Swe-24h
Swe-6day
Figure 3.2. Cumulative frequency distribution of personal exposure concentrations of formaldehyde in Helsinki (Hel,
48-hour average, n= 15, Jurvelin et al 2001) and Sweden (Swe-24h = 24-hour average, n=24, and Swe-6day = 6-day
average, n=40, Gustafson et al 2004).
The INDEX project Final report
35
4. Toxicokinetics
Absorption
Owing to its solubility in water, formaldehyde is rapidly absorbed in the respiratory and gastrointestinal tracts and
metabolized. Over 90% of inhaled formaldehyde gas is absorbed in the upper respiratory tract of rats and monkeys. In
rats, it is absorbed in the nasal passages; in monkeys, it is also absorbed in the nasopharynx, trachea and proximal
regions of the major bronchi. Although formaldehyde or its metabolites can penetrate human skin – it induces allergic
contact dermatitis in humans – dermal absorption appears to be very slight (WHO, 1989; IARC, 1995).
Distribution
Owing to its rapid metabolism, exposure of humans, monkeys or rats to formaldehyde by inhalation does not alter the
concentration of endogenous formaldehyde in the blood, which is about 2–3 mg/litre for each of the three species.
Intravenous administration of formaldehyde to dogs, cats and monkeys does not result in accumulation of formaldehyde
in the blood, because of its rapid conversion to formate. In dogs, orally administered formaldehyde results in a rapid
increase in formate levels in the blood. Following a 6-hour inhalation exposure of rats to
14
C-formaldehyde,
radioactivity was extensively distributed in other tissues, the highest concentration occurring in the oesophagus,
followed by the kidneys, liver, intestine and lung, indicating that absorbed
14
C-formaldehyde and its metabolites are
rapidly removed by the mucosal blood supply and distributed throughout the body (WHO, 1989).
Metabolism and elimination
Formaldehyde reacts virtually instantaneously with primary and secondary amines, thiols, hydroxyls and amides to
form methylol derivatives. Formaldehyde acts as an electrophile and can react with macromolecules such as DNA,
RNA and protein to form reversible adducts or irreversible cross-links. Absorbed formaldehyde can be oxidized to
formate along three different pathways, and can be exhaled as carbon dioxide or incorporated into biological
macromolecules via tetrahydrofolate-dependent one-carbon biosynthetic pathways. In the body, formaldehyde is
produced in small quantities as a normal metabolite and also in the oxidative demethylation of xenobiotics; it may
therefore be found in the liver (IARC, 1995). Formaldehyde disappears from the plasma with a half-time of about 1–1.5
minutes, most of it being converted to carbon dioxide and exhaled via the lungs. Smaller amounts are excreted in the
urine as formate salts and several other metabolites (WHO, 1987).
5. Health effects
Effects of short-term exposure
Predominant signs of short-term exposure to formaldehyde in humans are irritation of the eyes, nose and throat, together
with concentration-dependent discomfort, lachrymation, sneezing, coughing, nausea, dyspnoea and finally death (Table
5.1). Symptoms are often more severe at the start of exposure than after minutes or hours, when they gradually
diminish.
Table 5.1: Effects of formaldehyde in humans after short-term exposure
Concentration range
or average (mg/m
3
)
Time range or average Health effects in general population
0.03 Repeated exposure Odour detection threshold (10th percentile)
a
0.18 Repeated exposure Odour detection threshold (50th percentile)
a
0.6 Repeated exposure Odour detection threshold (90th percentile)
a
0.1 – 3.1 Single and repeated exposure Throat and nose irritation threshold
0.6–1.2 Single and repeated exposure Eye irritation threshold
0.5 – 2 3–5 hours Decreased nasal mucus flow rate
2.4 40 minutes on 2 successive days with
10 minutes of moderate exercise on
second day
Post-exposure (up to 24 hours) headache
The INDEX project Final report
36
2.5 – 3.7
b
Biting sensation in eyes and nose
3.7 Single and repeated exposure Decreased pulmonary function only at heavy
exercise
5 – 6.2 30 minutes Tolerable for 30 minutes with lachrymation
12 – 25
– b
Strong lachrymation, lasting for 1 hour
37 – 60
– b
Pulmonary oedema, pneumonia, danger to life
60 – 125
– b
Death
a
Frequency of effect in population.
b
Time range or average unspecified.
Sources: WHO, 1989; IARC, 1995; WHO, 1987
Numerous acute controlled and occupational human exposure studies have been conducted with both asthmatic and
normal subjects to investigate formaldehyde’s irritant effects on the eyes and the upper respiratory tract. Feinman
(1988) states that most people cannot tolerate exposures to more than 6 mg/m
3
(5 ppm) formaldehyde in air
A series of pulmonary function studies has been conducted in healthy nonsmokers and asthmatics exposed to
formaldehyde vapour; generally, lung function was unaltered. Fifteen healthy nonsmokers and 15 asthmatics were
exposed to 2.4 mg/m
3
formaldehyde for 40 minutes to determine whether acute exposures could induce asthmatic
symptoms (Schachter et al. 1986; Witek. et al. 1987). No significant airway obstruction, changes in pulmonary function
or bronchial hyperreactivity were noted. Similar observations were made on a group of 15 hospital laboratory workers
who had been exposed to formaldehyde (Schachter et al. 1987).
Healthy nonsmokers and asthmatics were exposed to 3.7 mg/m
3
formaldehyde for 1 or 3 hours, either at rest or when
engaged in intermittent heavy exercise. No significant changes in pulmonary function and nonspecific airway reactivity
were observed among asthmatic subjects. Small decreases (< 5%) in pulmonary function were observed in healthy non-
smokers exposed to formaldehyde while engaged in heavy exercise. Two normal and two asthmatic subjects had
decrements greater than 10% (Sauder et al. 1986; Green et al. 1987).
When 15 asthmatic subjects were exposed for 90 minutes to concentrations of 0.008–0.85 mg/m
3
formaldehyde, no
change in pulmonary function was seen, and there was no evidence of an increase in bronchial reactivity (Harving, et
al., 1990).
In a study of controlled exposure to formaldehyde, 18 subjects, 9 of whom had complained of adverse effects from
urea–formaldehyde foam insulation installed in their homes, were exposed to 1.2 mg/m
3
formaldehyde, or to off-gas
products of urea–formaldehyde foam insulation containing 1.5 mg/m
3
formaldehyde, for 90 minutes (Day et al., 1984).
No statistically or clinically significant change in pulmonary function was seen either during or 8 hours after exposure,
and no evidence was obtained that urea–formaldehyde foam insulation off-gas acts as a lower airway allergen.
Kulle et al. (1987; 1993) exposed 19 healthy subjects to 0, 1.2, and 2.5 mg/m
3
(0, 1.0, and 2.0 ppm) for 3-hour periods
and asked them to note symptoms of eye and nose/throat irritation and to rate severity on a 0-3 scale: 0=none; l=mild
(present but not annoying); 2=moderate (annoying); and 3=severe debilitating). Ten of the subjects were also exposed
to 0.6 mg/m
3
(0.5 ppm) and nine were exposed to 4 mg/m
3
(3 ppm) for 3-hour periods. The frequencies of subjects
reporting eye irritation or nose/throat irritation increased with increasing exposure concentration, especially at
concentrations 1.2 mg/m
3
. Under nonexposed conditions, 3/19 subjects noted mild nose/throat irritation and l/19
noted mild eye irritation. At 0.6 mg/m
3
, l/10 subjects noted mild nose/throat irritation, but none reported eye irritation.
Frequencies for subjects with mild or moderate eye irritation were 4/19 at 1.2 mg/m
3
(1 moderate), 10/19 at 2.5 mg/m
3
(4 moderate), and 9/9 at 4 mg/m
3
(4 moderate). The increased frequency for eye irritation (compared with controls) was
statistically significant at 2.5 mg/m
3
. Frequencies for mild nose/throat irritation were l/19 at 1.2 mg/m
3
, 7/19 at 2.5
mg/m
3
, and 2/9 at 4 mg/m
3
. Compared with control frequency for nose/throat irritation, only the response at 2.5 mg/m
3
was significantly elevated.
Weber-Tschopp et al. (1977) exposed a group of 33 healthy subjects for 35 minutes to concentrations of formaldehyde
that increased during the period from 0.04 to 3.9 mg/m
3
(0.03 to 3.2 ppm); another group of 48 healthy subjects was
exposed to 0.03, 1.2, 2.1, 2.8, and 4.0 ppm for 1.5 minute intervals. Eye and nose irritation were reported on an l-4 scale
(l=none to 4=strong) in both experiments, and eye blinking rate was measured in the second experiment. Average
indices of eye and nose irritation were increased in both experiments to a small, but statistically significant at 1.2 ppm
compared with indices for nonexposed controlled conditions. The published report of this study graphically showed
average severity scores of about 1.3-l.4 for both indices at 1.2 ppm compared with 1.0-l.1 for non exposed conditions.
The average severity score was increased to a greater degree at higher concentrations, but was less than about 2.5 at the
The INDEX project Final report
37
highest exposure concentration, 4 ppm. Average rates of eye blinking were not significantly affected at 1.2 ppm, but
were statistically significantly increased at 2.1 ppm (about 35 blinks/minute at 2.1 ppm versus about 22 blinks/minutes
under nonexposed conditions).
The relation of chronic respiratory symptoms and pulmonary function to formaldehyde (HCHO) in homes was studied
in a sample of 298 children (6-15 years of age) and 613 adults. HCHO measurements were made with passive samplers
during two 1-week periods. Data on chronic cough and phlegm, wheeze, attacks of breathlessness, and doctor diagnoses
of chronic bronchitis and asthma were collected with self-completed questionnaires. Peak expiratory flow rates (PEFR)
were obtained during the evenings and mornings for up to 14 consecutive days for each individual. Significantly greater
prevalence rates of asthma and chronic bronchitis were found in children from houses with HCHO levels 74-147 µg/m³
(60-120 ppb) than in those less exposed, especially in children also exposed to environmental tobacco smoke. In
children, levels of PEFR decreased linearly with HCHO exposure, with the estimated decrease due to 74 µg/m³ of
HCHO equivalent to 22% of PEFR level in nonexposed children. The effects in asthmatic children exposed to HCHO
below 61 µg/m³ were greater than in healthy ones. The effects in adults were less evident: decrements in PEFR due to
HCHO over 49 µg/m³ were seen only in the morning, and mainly in smokers (Krzyzanowski et al., 1990).
Studies in children, including the Krzyzanowski study above, indicate adverse health impacts in children at
concentrations as low as 37 µg/m
3 (
30 ppb). Wantke et al. (1996) reported that formaldehyde-specific IgE and
respiratory symptoms were reduced when children transferred from schools with formaldehyde concentrations of 53 to
92 µg/m
3 (
43 to 75 ppb) to schools with concentrations of 28 to 36 µg/m
3
(23 to 29 ppb). Garrett et al. (1999) reported
increased sensitization associated with the formaldehyde level in children’s homes which had a median value of 15.8
µg/m
3
(12.6 ppb). And Franklin et al. (2000) reported significantly higher exhaled nitric oxide, an indicator of airway
inflammation, in the breath of children living in homes with formaldehyde concentrations greater than 61 µg/m
3
(50
ppb) than in the breath of those children living in homes with formaldehyde levels below 61 µg/m
3
. These human
studies are not entirely consistent with each other, and there is potential for confounding in each. Nevertheless, taken
together, they suggest that children may be more sensitive to formaldehyde toxicity than adults.
Pulmonary function has been assessed in residents of mobile and conventional homes (Broder et al., 1988) and mobile
offices (Main & Hogan, 1983)) exposed to concentrations of 0.007–2.0 mg/m
3
. No changes were seen in pulmonary
function or airway resistance.
Eleven healthy subjects and nine patients with formalin skin sensitization were exposed to 0.5 mg/m³ formaldehyde for
2 hours (Pazdrak et al., 1993). Nasal lavage was performed prior to and 5 to 10 minutes, 4 hours, and 18 hours after
exposure. Rhinitis was reported and increases in the number and proportion of eosinophils, elevated albumin and
increased protein levels were noted in nasal lavage fluid 4 and 18 hours after exposure. No differences were found
between patients with skin sensitization and healthy subjects.
Summary of short-term exposure effect levels
Studies of humans under controlled conditions clearly indicate that acute exposures to air concentrations ranging from
0.5 to 3.7 mg/m
3
(0.4 to 3 ppm):
- induce reversible eye, nose, and throat irritation (Andersen and Mølhave 1983; Bender et al. 1983; Day et al.
1984; Gorski et al. 1992; Krakowiak et al. 1998; Kulle 1993; Kulle et al. 1987; Pazdrak et al. 1993; Schachter et
al. 1986; Weber-Tschopp et al. 1977; Witek et al. 1986);
- produce changes in nasal lavage fluid contents, indicative of irritation of the nasal epithelium (Gorski et al. 1992;
Krakowiak et al. 1998; Pazdrak et al. 1993); and
- do not consistently or markedly affect pulmonary function variables in most individuals (Andersen and Mølhave
1983; Day et al. 1984; Gorski et al. 1992; Green et al. 1987; Harving et al. 1986, 1990; Kulle et al. 1987;
Nordman et al. 1985; Sauder et al. 1986; Schachter et al. 1986; Witek et al. 1986).
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
0.6
EXP
1.2
EXP
0.53
BC05
0.94
1h-ADJ-BC05
Mild and moderate eye irritation Volunteers, 3h Kulle et al. (1987) OEHHA 1999;
REL: 0.094
0.5
EXP
Nasal and eye irritation
Volunteers, 2h
(potentially sensitive
population)
Pazdrak et al. (1993) ATSDR 1999;
MRL: 0.05
0.24
STAT
Respiratory tract irritation cited in Sloof et al.
(1992)
RIVM 1992
0.1 Nose and throat irritation Human, short-term cited in IPCS/WHO, WHO 2000
The INDEX project Final report
38
1989
(0.037) Respiratory symptoms (depending on study) Children; additional
investigation requested
Krzyzanowski et al.,
(1990); Wantke et al.
(1996); Garrett et al.
(1999); Franklin et al.
(2000)
UCLA 2001
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
OEHHA (1999)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Derivation of Acute Reference Exposure Level (for a 1-hour exposure)
Study Kulle et al. (1987)
Study population 19 nonasthmatic, nonsmoking human subjects
Exposure method 0.6- 3.6 mg/m³ (0.5-3.0 ppm)
Critical effects mild and moderate eye irritation
LOAEL 1.2 mg/m³ (1 ppm)
NOAEL 0.6 mg/m³ (0.5 ppm)
Benchmark concentration 0.44 ppm (BC
05)
Exposure duration 3 hours
Extrapolated 1 hour concentration 0.94 mg/m³ [0.76 ppm (0.44
2
ppm* 3 h = C
2
* 1 h )]
LOAEL uncertainty factor not required in BC approach
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 10
Cumulative uncertainty factor 10
Reference Exposure Level 0.076 ppm (0.094 mg/m³; 94 µg/m³)
The recommended REL was estimated by a benchmark concentration (BC
05) approach, using logprobit analysis
(Crump, 1984; Crump and Howe, 1983). The BC
05 is defined as the 95% lower confidence limit of the concentration
expected to produce a response rate of 5%. The resulting BC05 from this analysis was 0.53 mg/m³ formaldehyde. This
value was adjusted to a 1-hour duration using the formula Cn * T = K, where n = 2 (AICE, 1989), resulting in a value of
0.94 mg/m³. An uncertainty factor (UF) of 10 was used to account for individual variation.
Generally an uncertainty factor of 3 would be used with the BC
05 for intraindividual variability, since the BC05 accounts
for some degree of individual variation. However, information from the literature indicates a wide variability in
response to formaldehyde irritancy including reports of irritation (NIOSH HHE reports 1981-1996; Liu et al. 1991;
Horvath et al, 1985) or cellular changes associated with irritation and an immune response at levels below the one-hour
extrapolated BC
05 (Pazdrak et al, 1993; Gorski et al, 1992). For these reasons, we used an uncertainty factor of 10 to
account for intraindividual variability in the human population.
ATSDR (1999)
Agency for Toxic Substances and Disease Registry
Derivation of the Minimal Risk Level (MRL):
ATSDR has derived an acute inhalation MRL of 0.05 mg/m³ (0.04 ppm) on the basis of clinical symptoms (increased
itching, sneezing, mucosal congestion, transient burning sensation of the eyes and of the nasal passages) and nasal
alterations (elevated eosinophil counts and a transient increase in albumin content of nasal lavage fluid) in humans
(Pazdrak et al. 1993). This MRL is based on a minimal LOAEL of 0.5 mg/m³ and an uncertainty factor of nine (three
for use of a minimal LOAEL and three for human variability).
The Anderson and Mølhave (1983) study identified an apparent effect level at 0.3 mg/m³ (0.2 ppm), based on subjective
reports of irritation that is lower than the effect levels at 0.43-0.5 mg/m³ (0.35-0.4 ppm) in the studies by Pazdrak et al.
(1993), Krakowiak et al. (1998), and Bender et al. (1983), which used more objective measures of acute irritation
(eosinophil counts and protein concentrations in nasal lavage fluid or time to first reporting of irritation). Because of the
use of objective measures of toxicity and the general weight of the available data indicating that some people will not
experience eye or upper respiratory tract irritation from formaldehyde even at 1.2 mg/m³ (see Day et al. 1984; Kulle et
al. 1987, Weber Tschopp et al. 1977, and Witek et al. 1986), the Pazdrak et al. (1993) LOAEL of 0.5 mg/m³ was
The INDEX project Final report
39
considered a minimal LOAEL in a group of potentially sensitive individuals (some subjects had dermal hypersensitivity
to formaldehyde) and selected as the basis of the acute MRL.
RIVM (1992)
National Institute of Public Health and the Environment - The Netherlands
Sensory reactions are apparently the most typical effects in the non-industrial indoor environment (Sloof et al., 1992). A
large number of observations of people in various settings support a conclusion that the generally observed range over
which more than 95 % of people respond is 125-3750 µg/m
3
(0.10-3.1 ppm). According to the Dutch Health Council
(1984) and WHO (1989) the lowest statistically significant effect concentration for respiratory tract irritation is 240
µg/m
3
(LOAEL) (0.20 ppm) (Sloof et al., 1992).
The Dutch Health Council (1984) recommended the following three limit values (Sloof et al., 1992):
120 µg/m
3
as ceiling value (based on 30 min. means);
40 µg/m
3
as 98-percentile value (based on 24 h means);
30 µg/m
3
as 95-percentile value (based on 24 h means).
WHO (2001)
Air Quality Guidelines for Europe 2000
The lowest concentration that has been associated with nose and throat irritation in humans after short-term exposure is
0.1 mg/m
3
, although some individuals can sense the presence of formaldehyde at lower concentrations.
To prevent significant sensory irritation in the general population, an air quality guideline value of 0.1 mg/m
3
as a 30-
minute average is recommended. Since this is over one order of magnitude lower than a presumed threshold for
cytotoxic damage to the nasal mucosa, this guideline value represents an exposure level at which there is a negligible
risk of upper respiratory tract cancer in humans.
Effects of long-term exposure (noncancer)
Long term exposure to elevated levels of formaldehyde in the occupational setting has been shown to result in upper
and lower airway irritation and eye irritation in humans; degenerative, inflammatory and hyperplastic changes of the
nasal mucosa in humans and animals. The voluminous body of data describing these effects has been briefly
summarized below. For the sake of brevity, only the studies that best represent the given effects are presented.
Ritchie and Lehnen (1987) reported a dose-dependent increase in health complaints (eye and throat irritation, and
headaches) in 2000 residents living in 397 mobile and 494 conventional homes, that was demonstrated by logistic
regression. Complaints of symptoms of irritation were noted at concentrations of 0.12 mg/m³ formaldehyde or above.
Similarly, Liu et al. (1991) found that exposure to 0.135 mg/m
3
formaldehyde exacerbated chronic respiratory and
allergy problems in residents living in mobile homes.
Employees of mobile day-care centers (66 subjects) reported increased incidence of eye, nose and throat irritation,
unnatural thirst, headaches, abnormal tiredness, menstrual disorders, and increased use of analgesics as compared to
control workers (Olsen and Dossing, 1982). The mean formaldehyde concentration in these mobile units was 0.43
mg/m
3
(range = 0.24 - 0.55 mg/m3). The exposed workers were exposed in these units a minimum of 3 months. A
control group of 26 subjects in different institutions was exposed to a mean concentration of 0.06 mg/m
3
formaldehyde.
Occupants of houses insulated with urea-formaldehyde foam insulation (UFFI) (1726 subjects) were compared with
control subjects (720 subjects) for subjective measures of irritation, pulmonary function (FVC, FEV
1, FEF25-75, FEF50),
nasal airway resistance, odour threshold for pyridine, nasal cytology, and hypersensitivity skin-patch testing (Broder et
al., 1988). The mean length of time of exposure to UFFI was 4.6 years. The mean concentration of formaldehyde in the
UFFI-exposed group was 0.053 mg/m³, compared with 0.043 mg/m³ for the controls. A significant increase in
symptoms of eye, nose and throat irritation was observed in subjects from UFFI homes, compared with controls. No
other differences from control measurements were observed.
An increase in severity of nasal epithelial histological lesions, including loss of cilia and goblet cell hyperplasia (11%),
squamous metaplasia (78%), and mild dysplasia (8%), was observed in 75 wood products workers exposed to between
0.1 and 1.1 mg/m
3 formaldehyde for a mean duration of 10.5 years (range = 1 - 39 years), compared to an equal number
of control subjects (Edling et al., 1988). Only three exposed men had normal mucosa. A high frequency of symptoms
relating to the eyes and upper airways was reported in exposed workers. Nasal symptoms included mostly a runny nose
The INDEX project Final report
40
and crusting. The histological grading showed a significantly higher score for nasal lesions when compared with the
referents (2.9 versus 1.8). Exposed smokers had a higher, but non-significant, score than ex-smokers and non-smokers.
When relating the histological score to duration of exposure, the mean histological score was about the same regardless
of years of employment. In addition, no difference in the histological scores was found between workers exposed only
to formaldehyde and those exposed to formaldehyde and wood dust.
Alexandersson and Hedenstierna (1989) evaluated symptoms of irritation, spirometry, and immunoglobulin levels in 34
wood workers exposed to formaldehyde over a 4-year period. Exposure to 0.5 – 0.6 mg/m³ (0.4 - 0.5 ppm)
formaldehyde resulted in significant decreases in FVC, FEV
1, and FEF25-75. Removal from exposure for 4 weeks
allowed for normalization of lung function in the non-smokers.
Symptoms of irritation were reported by 66 workers exposed for 1 - 36 years (mean = 10 years) to a mean concentration
of 0.26 mg/m
3
formaldehyde (Wilhelmsson and Holmstrom, 1992). Controls (36 subjects) consisted of office workers in
a government office and were exposed to a mean concentration of 0.09 mg/m
3
formaldehyde. The significant increase in
symptoms of irritation in exposed workers did not correlate with total serum IgE antibody levels. However, 2 exposed
workers, who complained of nasal discomfort, had elevated IgE levels. In another occupational health study, 37
workers, who were exposed for an unspecified duration to formaldehyde concentrations in the range of 0.004 to 0.090
mg/m³, reported ocular irritation; however, no significant serum levels of IgE or IgG antibodies to formaldehyde-human
serum albumin were detected (Grammer et al., 1990). An epidemiological study of the effects of formaldehyde on 367
textile and shoe manufacturing workers employed for a mean duration of 12 years showed no significant association
between formaldehyde exposure, pulmonary function (FVC, FEV
1, and PEF) in normal or asthmatic workers, and
occurrence of specific IgE antibodies to formaldehyde (Gorski and Krakowiak, 1991). The concentrations of
formaldehyde did not exceed 0.75 mg/m
3
. Workers (38 total) exposed for a mean duration of 7.8 years to 0.14 – 2.60
mg/ (mean = 0.41 mg/m³) formaldehyde were studied for their symptomatology, lung function, and total IgG and IgE
levels in the serum (Alexandersson and Hedenstierna, 1988). The control group consisted of 18 unexposed individuals.
Significant decrements in pulmonary function (FVC and FEV
1) were observed, compared with the controls. Eye, nose,
and throat irritation was also reported more frequently by the exposed group, compared with the control group. No
correlation was found between duration of exposure, or formaldehyde concentration, and the presence of IgE and IgG
antibodies.
Holmstrom et al. (1989) examined histological changes in nasal tissue specimens from a group of 70 workers in a
chemical plant that produced formaldehyde and formaldehyde resins for impregnation of paper, a group of 100 furniture
factory workers working with particle board and glue components, and a nonexposed control group of 36 office workers
in the same village as the furniture factories. Mean durations of employment in the groups were 10.4 years (SD 7.3,
range 1–36 years) for the chemical workers and 9.0 years (SD 6.3, range 1–30 years) for the furniture workers.
Estimates of personal breathing zone air concentrations ranged from 0.05 to 0.5 mg/m³ (median 0.29±0.16 ppm) for the
chemical workers, from 0.20 to 5 mg/m
3
(median 0.25±0.05 ppm) for the furniture workers, and from 0.09 to 0.16
mg/m³ in the late summer for the office workers with a year-round office worker median reported as 0.09 mg/m
3
with
no standard deviation. The mean wood dust concentration in the furniture factory was reported to have been between 1
and 2 mg/m
3
. Nasal mucosa specimens were taken from the medial or inferior aspect of the middle turbinate. Histology
scores were assigned to each specimen based on a 0–8 scale, identical to the scale used by Edling et al. (1988; described
previously). Nasal mucosal biopsy sections for each subject were examined and assigned scores as follows: 0 - normal
respiratory epithelium; 1- loss of ciliated epithelium cells; 2 - mixed cuboid/squamous epithelium, metaplasia; 3 -
stratified squamous epithelium; 4 - keratosis; 5 - budding of epithelium; 6 - mild or moderate dysplasia; 7 - severe
dysplasia; and 8 - carcinoma.
Nasal histology scores ranged from 0 to 4 (mean 2.16; n=62) for the chemical workers, from 0 to 6 (mean 2.07; n=89)
for the furniture workers, and from 0 to 4 (mean 1.46; n=32) for the office workers. The mean histological score for the
chemical workers, but not the furniture workers, was significantly different from the control score, thus supporting the
hypothesis that the development of the nasal lesions is formaldehyde-related and not obligatorily related to possible
interaction between formaldehyde and wood dust. The most severe epithelial change found (light or moderate epithelial
dysplasia) was found in two furniture workers. Among the chemical workers (not exposed to wood dust), loss of cilia,
goblet cell hyperplasia and cuboidal and squamous cell metaplasia replacing the columnar epithelium occurred more
frequently than in the control group of office workers. Within both groups of formaldehyde-exposed workers, no
evidence was found for associations between histological score and duration of exposure, index of accumulated dose, or
smoking habit.
The INDEX project Final report
41
Summary of long-term exposure effect levels (noncancer)
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
0.09
Study
0.032
ADJ
0.26
Study
0.093
ADJ
Nasal and eye irritation, nasal obstruction, and
lower airway discomfort; histopathological nasal
lesions including rhinitis, squamous metaplasia,
and dysplasia
Occupational, 10y
average;
Wilhelmsson and
Holmstrom, 1992;
supported by Edling et
al., 1988
OEHHA 1999;
REL: 0.003
0.3
Study
Mild irritation of the eyes and upper respiratory
tract and mild damage to the nasal epithelium
Occupational, 10.4y Holmstrom et al., 1989
ATSDR 1999;
MRL: 0.01
0.31
STAT
Sensory irritation
for low but significant
percentage of exposed
workers
Weighting the total
body of data
NIWL 2003
0.12 Symptoms of irritation
LOAEL may be lower
only for a very small
proportion of the
population
well conducted studies Health Canada
1999; (noTC)
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
OEHHA (1999)
Office of Environmental Health Hazard Assessment
Derivation of Chronic Reference Exposure Level (REL):
Studies Wilhelmsson and Holmstrom, 1992; supported by Edling et al.,
1988
Study population Human chemical plant workers (66 subjects)
Exposure method Discontinuous occupational exposure
Critical effects Nasal and eye irritation, nasal obstruction, and lower airway
discomfort; histopathological nasal lesions including rhinitis,
squamous metaplasia, and dysplasia
LOAEL Mean of 0.26 mg/m
3
(range = 0.05 to 0.6 mg/m
3
) (described as
exposed group)
NOAEL Mean of 0.09 mg/m
3
(described for control group of office workers)
Exposure continuity 8 hours/day, 5 days/week (assumed)
Exposure duration 10 years (average); range = 1-36 years
Average occupational concentration 0.032 mg/m
3
for NOAEL group (0.09 x 10/20 x 5/7)
Human equivalent concentration 0.032 mg/m
3
LOAEL uncertainty factor 1
Subchronic uncertainty factor 1
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 10
Cumulative uncertainty factor 10
Inhalation reference level 0.003 mg/m
3
(3 µg/m
3
; 0.002 ppm; 2 ppb)
The Wilhelmsson and Holmstrom study was selected by the Office of Environmental Health Hazard Assessment
(OEHHA, reference) for the derivation of the Chronic Reference Exposure Level (REL), because it was a human
occupational study that contained a LOAEL and a NOAEL and contained a reasonable number of subjects. The
supporting occupational study by Edling et al. (1988) noted similar sensory irritation results due to long-term
formaldehyde exposure. In addition, nasal biopsies from exposed workers in the Edling et al. (1988) study exhibited
nasal epithelial lesions similar to those found in subchronic and chronic animal studies.
For comparison with the proposed REL of 3 µg/m
3
, we estimated a REL from Edling et al. (1988). A median
concentration of 0.6 mg/m
3
was determined for the LOAEL from the TWA range of 0.1- 1.1 mg/m
3
. A NOAEL was not
reported. The average continuous occupational concentration was 0.2 mg/m
3
(0.6 x 10/20 x 5/7) and the exposure
duration was 10.5 years (range = 1 – 39 years). Application of a UF of 10 for intraspecies variability and a UF of 10 for
estimation of a NOAEL from the LOAEL would result in a REL of 2 µg/m
3
(2 ppb).
Data Strengths and Limitations for Development of the REL: The strengths of the inhalation REL include the use of
human exposure data from workers exposed over a period of years and the observation of a NOAEL. In addition, a
The INDEX project Final report
42
number of well conducted animal studies supported the derivation of the REL. The major areas of uncertainty are the
uncertainty in estimating exposure in the occupational studies and the potential variability in exposure concentration.
Table 5.2 presents a summary of potential RELs based on chronic and subchronic animal studies. The toxicological
endpoint was nasal lesions, consisting principally of rhinitis, squamous metaplasia, and dyplasia of the respiratory
epithelium.
Table 5.2: Summary of chronic and subchronic formaldehyde studies in experimental animals
Study Animal Exposure
Duration
LOAEL/
NOAEL
(mg/m
3
)
HEC
adj.
(mg/m
3
)
Cumulativ
e
UF
REL
(mg/m3)
Woutersen et al.,
1989
rat 28 m 9.8 / 1.0 0.06 30 2
Kerns et al., 1983 rat 24 m 2.0 / NA 0.1 300 0.3
Monticello et al.,
1996
rat 24 m 6.01 / 2.05 0.1 30 4
Kamata et al., 1997 rat 24-28 m 0.30 / NA 0.02 100 0.2
Appelman et al.,
1988
rat 52 w 9.4 / 1.0 0.06 30 2
Rusch et al., 1983 rat 26 w 2.95 / 0.98 0.2 30 7
Kimbell et al., 1997 rat 26 w 6 / 2 0.1 30 3
Wilmer et al., 1989 rat 13 w 4 / 2 0.2 300 0.7
Woutersen et al.,
1987
rat 13 w 9.7 / 1.0 0.03 100 0.3
Zwart et al., 1988 rat 13 w 2.98 / 1.01 0.2 300 0.7
Kerns et al., 1983 mouse 24 m 2.0 / NA 0.05 100 0.5
Maronpot et al., 1986 mouse 13 w 10.1 / 4.08 0.09 100 0.9
Rusch et al., 1983 monkey 26 w 2.95 / 0.98 none 300 4
The most striking observation is the similarity of potential RELs among the rat chronic studies (exposures > 26 weeks)
that contain a NOAEL. The range of RELs from these animal studies, 2 – 7 mg/m
3
, is comparable to the proposed REL
based on a human study. Another related observation is that the NOAEL and LOAEL are similar among all the studies,
regardless of exposure duration. The NOAEL and LOAEL are generally in the range of 1-2 mg/m3 and 2-10 mg/m3,
respectively, with the exception of the study by Kamata et al. (1997). These results indicate that the formation of
formaldehyde-related nasal lesions are more concentration dependent than time, or dose, dependent. A limitation of a
majority of the occupational studies is their high reliance on surveys and other methods that focus on sensory irritation.
Such sensory irritant results, as exhibited in the Wilhelmsson and Holmstrom (1992) study, may be more related to
recurrent acute injury rather than a true chronic injury. The concentration dependent nature of the nasal lesions in the
supporting animal studies, and suggested in the supporting human nasal biopsy study, would also imply that the nasal
cavity endpoint may be a recurrent acute effect. However, Kerns et al. (1983) and Kamata et al. (1997) clearly
demonstrated that near the LOAEL, increasing exposure durations would result in nasal lesions at lower formaldehyde
concentrations. Also, the rat study by Woutersen et al. (1989) demonstrated that subchronic exposure to formaldehyde
concentrations that produce nasal lesions could result in lifelong changes of the nasal epithelium. These findings
substantiate the chronic nature of the nasal/upper airway injury that results from long-term formaldehyde exposure.
ATSDR (1999)
Agency for Toxic Substances and Disease Registry
Derivation of the chronic Minimal Risk Level (MRL).
The chronic inhalation MRL of 0.01 mg/m
3
was derived from a LOAEL of 0.3 mg/m
3
in humans for nasal effects
(Holmstrom et al., 1989). This minmal LOAEL was divided by a total uncertainty factor of 30 (3 for use of a LOAEL
and 10 for human variability). The exposure concentration was not adjusted to a continuous exposure basis based on
evidence that concentration is more important than the product of concentration and duration of exposure in
determining the severity of formaldehyde-induced epithelial damage in the upper respiratory tract (Wilmer et al. 1987).
The INDEX project Final report
43
National Institute for Working Life (NIWL) - Sweden (2003)
Nordic Expert Group for Criteria Documentation of Health Risks from Chemicals (NEG) and Dutch Expert Committee
on Occupational Standards (DECOS)
Summary of the report n°132.Formaldehyde (Scientific basis for an occupational exposure limit):
Inhaled formaldehyde is almost completely absorbed in the upper respiratory tract in rodents. Formaldehyde is a normal
metabolite in mammalians and is rapidly metabolised to formate, which may be further oxidised to carbon dioxide.
Final elimination occurs via exhalation and via the kidneys. The target organs of formaldehyde vapour are the eyes,
nose, and throat. The effects of concern are sensory irritation and cytotoxicity-induced regenerative hyperplasia and
metaplasia of the nasal respiratory epithelium accompanied by nasal carcinomas in rats. Weighting the total body of
data 0.31 mg/m
3
(0.25 ppm) formaldehyde is the LOAEL at which sensory irritation may occur in a low but significant
percentage of exposed workers. The majority of short- and long-term inhalation animal studies reveal a NOAEL of 1.2-
2.5 mg/m
3
(1-2 ppm). However, in a few studies slight histopathological changes of the nasal respiratory epithelium
were observed at 0.4-2.5 mg/m
3
(0.3-2 ppm).
Formaldehyde is genotoxic in a variety of experimental systems, including rodents in vivo. There is overwhelming
evidence that high, cytotoxic formaldehyde vapour concentrations (10 ppm) can induce nasal cancer in rats. A large
body of data suggests an association between the cytotoxic, genotoxic, and carcinogenic effects of formaldehyde. The
crucial role of tissue damage followed by hyperplasia and metaplasia of the nasal respiratory epithelium in
formaldehyde carcinogenesis has been demonstrated in a convincing way. Thus, formaldehyde in non-cytotxic
concentrations most probably cannot act as a complete carcinogen. However, if human exposure to formaldehyde is
accompanied by recurrent tissue damage at the site of contact, formaldehyde may be assumed to have carcinogenic
potential in man.
Formaldehyde-induced allergic contact dermatitis has been estimated to occur in 3-6% of the population, and skin
sensitisation by direct skin contact has been induced with formaldehyde solutions. There is no consistent evidence of
formaldehyde being a respiratory sensitiser.
Health Canada (1999)
Canadian Environmental Protection Act (CEPA).
Health Canada did not derive a Tolerable Concentration (TC) for formaldehyde. However, there are considered to be
sufficient data from clinical studies and cross-sectional surveys of human populations, as well as supporting
observations from experimental studies conducted with laboratory animals, to indicate that the irritant effects of
formaldehyde on the eyes, nose and throat occur at lowest concentration. Although individual sensitivity and exposure
conditions such as temperature, humidity, duration and co-exposure to other irritants are likely to influence response
levels, in well-conducted studies, only a very small proportion of the population experiences symptoms of irritation
following exposure to less than or equal to 0.12 mg/m
3
formaldehyde. This is less than the levels that reduce
mucociliary clearance in the anterior portion of the nasal cavity in available clinical studies in human volunteers (0.3
mg/m
3
) and induce histopathological effects in the nasal epithelium in cross-sectional studies of formaldehyde-exposed
workers (Andersen and Mølhave, 1983). Additional investigation of preliminary indication of effects on pulmonary
function in children in the residential environment associated with lower concentrations of formaldehyde (0.048-0.072
mg/m
3
) (Krzyzanowski et al., 1990) is warranted.
Carcinogenic effects
On June 15, 2004, IARC announced there was sufficient evidence that formaldehyde causes nasopharyngeal cancer in
humans and re-classified it as a Group 1, known human carcinogen (previous classification: Group 2A). IARC also
reported there was limited evidence that formaldehyde exposure causes nasal cavity and paranasal cavity cancer and
“strong but not sufficient“ evidence linking formaldehyde exposure to leukemia.
EPA (1991) classified formaldehyde in Group B1 - probable human carcinogen, based on an evaluation of limited
human evidence and sufficient laboratory animal evidence. More recently, in a collaborative review and evaluation of
the formaldehyde epidemiology studies, EPA and CIIT (CIIT 1998) concluded, “It appears that a weak association
between nasopharyngeal cancer and formaldehyde exposure cannot be completely ruled out.” Adopting another view,
McLaughlin (1994) concluded, “Clearly, the causal criteria used by epidemiologists to evaluate an association, such as
strength of the association, consistency, specificity, dose-response, plausibility, and coherence, are not satisfied by the
epidemiologic studies in the formaldehyde-cancer research domain”. ECETOC (1995) similarly concluded, “After a
The INDEX project Final report
44
careful review of the cytologic, cytogenic and epidemiological studies there is an absence of evidence to support the
judgement of an etiologic relationship between formaldehyde and human cancer risk.”
Human carcinogenicity data
A series of papers on the cytogenetic effects of formaldehyde in humans has been published (IARC, 1995). The studies
mostly concern occupational exposures (plywood makers, makers of wood splinter materials, morticians, paper makers
and hospital autopsy service workers) and measure chromosomal anomalies, micronuclei in peripheral lymphocytes,
buccal epithelium (Norppa, et al., 1993) or nasal epithelium and sperm abnormalities. Both positive and negative
results were obtained, but their interpretation is difficult because of the small number of subjects studied,
inconsistencies in the findings, inadequate reporting of the data, and concomitant exposure to other chemicals. No
increase in urinary mutagenicity was observed in autopsy workers as compared to controls (Connor, et al. 1985). No
data are available on DNA–protein cross-links in humans. The overall conclusion is that adequate data on genetic
effects of formaldehyde in humans are not available (IARC, 1995).
The relationship between exposure to formaldehyde and cancer has been investigated in over 25 cohort studies of
professionals (pathologists, anatomists and embalmers) and industrial groups (formaldehyde producers, formaldehyde
resin makers, plywood and particle board manufacturers, garment workers and workers in the abrasives industry).
Relative risks have been estimated as standardized mortality ratios (SMRs), proportionate mortality ratios (PMRs),
proportionate cancer mortality ratios (PCMRs) and standardized incidence ratios (SIRs). In some studies, exposure was
not assessed but was assumed on the basis of the subject’s occupation or industry; in others, it was based on duration of
exposure and quantitative estimates of historical exposure levels. Mortality in several of the cohorts was followed
beyond the period covered by the original report; only the latest results are reviewed below, unless there were important
differences in the analyses performed or changes in the cohort definition (IARC, 1995,).
Recent meta-analyses by Blair et al. (Blair et al. 1990) and Partanen (Partanen, 1993) contain most of the available data.
The International Agency for Research on Cancer (IARC) summarized the results of the two meta-analyses in tabular
form. IARC (IARC, 1995) also systematically reviewed case-control studies of cancer of the oral cavity, pharynx and
respiratory tract. Also, McLaughlin (McLaughlin, 1994) recently published a critical review on formaldehyde and
cancer.
Excess numbers of nasopharyngeal cancers were associated with occupational exposure to formaldehyde in two of six
cohort studies of industrial or professional groups, in three of four case-control studies and in meta-analyses. In one
cohort study performed in 10 plants in the United States, the risk increased with category of increasing cumulative
exposure. In the cohort studies that found no excess risk, no deaths were observed from nasopharyngeal cancer. In three
of the case-control studies, the risk was highest in people in the highest category of exposure and among people
exposed 20–25 years before death. The meta-analyses found a significantly higher risk among people estimated to have
had substantial exposure than among those with low/medium or no exposure. The observed associations between
exposure to formaldehyde and risk of cancer cannot reasonably be attributed to other occupational agents, including
wood dust, or to tobacco smoking. Limitations of the studies include misclassification of exposure and disease and loss
to follow-up, but these would tend to diminish the estimated relative risks and dilute exposure–response gradient. Taken
together, the epidemiological studies suggest a causal relationship between exposure to formaldehyde and
nasopharyngeal cancer, although the conclusion is tempered by the small numbers of observed and expected cases in
the cohort studies (IARC, 1995).
Of six case-control studies in which the risk for cancer of the nasal cavities and paranasal sinuses in relation to
occupational exposure to formaldehyde was evaluated, three provided data on squamous cell tumours and three on
unspecified cell types. Of the three studies of squamous cell carcinomas, two (from Denmark and the Netherlands)
showed a positive association, after adjustment for exposure to wood dust, and one (from France) showed no
association. Of the three studies of unspecified cell types, one (from Connecticut, United States) gave weakly positive
results and two (also from the United States) reported no excess risk. The two case-control studies that considered
squamous cell tumours and gave positive results involved more exposed cases than the other case-control studies
combined. In the studies of occupational cohorts overall, however, fewer cases of cancer of the nasal cavities and
paranasal sinuses were observed than were expected. Because of the lack of consistency between the cohort and case-
control studies, the epidemiological studies can do no more than suggest a causal role of occupational exposure to
formaldehyde in squamous cell carcinoma of the nasal cavities and paranasal sinuses (IARC, 1995).
The cohort studies of embalmers, anatomists and other professionals who use formaldehyde tended to show low or no
risk for lymphatic or haematopoietic cancers, and excess risks for cancers of the brain, although they were based on
small numbers. These findings are countered by consistent lack of excess risk for brain cancer in the studies of the
industrial cohorts, which generally included more direct and quantitative estimates of exposure to formaldehyde than
did the cohort studies of embalmers and anatomists (IARC, 1995).
The INDEX project Final report
45
IARC (IARC, 1995) interpreted the above data as limited evidence for the carcinogenicity of formaldehyde in humans,
and classified formaldehyde as “probably carcinogenic to humans” (Group 2A).
In a published cohort study (Hansen & Olsen, 1995), a significantly increased relative risk of 3.0 (1.4–5.7; 95%
confidence limits) for sinonasal cancer was found among blue-collar workers potentially exposed to formaldehyde, but
with no probable exposure to wood dust, the major confounder.
Animal carcinogenicity data
Formaldehyde was tested for carcinogenicity by inhalation in mice, rats and hamsters, by oral administration in
drinking-water in rats, by skin application in mice, and by subcutaneous injection in rats. In additional studies in mice,
rats and hamsters, modification of the carcinogenicity of known carcinogens was tested by administration of
formaldehyde in drinking-water, by application to the skin or by inhalation (IARC, 1995).
Several carcinogenicity studies in which formaldehyde was administered to rats by inhalation produced evidence of
carcinogenicity, particularly induction of squamous cell carcinomas of the nasal epithelium, usually only at the highest
exposure level which ranged from about 18 to 25 mg/m
3
. In one long-term inhalation study, a high incidence of nasal
squamous cell carcinomas (15/58) was found in rats exposed to 12.3 mg/m
3
, the nasal mucosa of which had been
severely damaged by electrocoagulation before the exposure to formaldehyde started. In a comparable group of rats
exposed to the same formaldehyde concentration but with an undamaged nasal mucosa, no formaldehyde-related nasal
tumours were found (IARC, 1995). In one study in mice, no statistically significantly increase in the incidence of nasal
tumours was found after chronic (2-year) exposure to formaldehyde concentrations up to 17.6 mg/m
3
. Similar long-term
studies in hamsters showed no evidence of carcinogenicity (IARC, 1995).
In rats administered formaldehyde in the drinking-water, increased incidences were seen of forestomach papillomas in
one study and of leukaemias and gastrointestinal tract tumours in another; two other studies in which rats were treated
through the drinking-water gave negative results. Studies in which formaldehyde was applied to the skin or injected
subcutaneously were inadequate for evaluation (IARC, 1995).
In experiments to test the effect of formaldehyde on the carcinogenicity of known carcinogens, oral administration of
formaldehyde concomitantly with N-nitrosodimethylamine to mice increased the incidence of tumours at various sites;
skin application in addition to 7,12-dimethylbenz[a]anthracene reduced the latency of skin tumours. In rats, concomitant
administration of formaldehyde and N-methyl-N´-nitro-N-nitrosoguanidine in the drinking water increased the
incidence of adenocarcinoma of the glandular stomach. Exposure of hamsters by inhalation to formaldehyde increased
the multiplicity of tracheal tumours induced by subcutaneous injections of N-nitrosodiethylamine (IARC, 1995).
Genotoxic effects
Formaldehyde induces DNA–protein cross-links in mammalian cells in vitro and in vivo (2). The precise nature of these
cross-links is unknown. They are removed from normal cells with a half-time of 2–3 hours; the removal rates were
similar at nontoxic and toxic concentrations of formaldehyde (Grafström et al., 1984).
DNA–protein cross-links were measured in the mucosa of several regions of the respiratory tract of rats exposed by
inhalation to 0.4, 0.9, 2.4, 7.3 or 12.2 mg/m
3
14
C-formaldehyde, and of rhesus monkeys exposed by inhalation (mouth-
only) to 0.9, 2.4 or 7.3 mg/m
3
14
C-formaldehyde for 6 hours. The concentration of cross-links increased non-linearly
with the airborne concentration in both species. The concentrations of cross-links in the turbinates and anterior nasal
mucosa were significantly lower in monkeys than in rats. Cross-links were also formed in the nasopharynx and trachea
of monkeys, but they were not detected in the sinus, proximal lung or bone marrow. The investigators suggested that the
differences between rats and monkeys with respect to DNA–protein cross-link formation may be due to differences in
nasal cavity deposition and in the elimination of absorbed formaldehyde (Casanova et al., 1991; Heck et al., 1989).
In addition to DNA–protein cross-links, formaldehyde induced DNA single-strand breaks, chromosomal aberrations,
sister chromatid exchanges and gene mutations in human cells in vitro. It induced cell transformation, chromosomal
aberrations, sister chromatid exchanges, DNA strand breaks, DNA–protein cross-links and gene mutations in rodent
cells in vitro. Administration of formaldehyde in the diet to Drosophila melanogaster induced lethal and visible
mutations, deficiencies, duplications, inversions, translocations and crossing-over in spermatogonia. Formaldehyde
induced mutations, gene conversion, DNA strand breaks and DNA–protein cross-links in fungi, and mutations and
DNA damage in bacteria. Assays for dominant lethal mutations in rodents in vivo gave inconclusive results. In single
studies, formaldehyde induced sperm head abnormalities in rats but not in mice (IARC, 1995).
While there is conflicting evidence that formaldehyde can induce chromosomal anomalies in the bone marrow of
rodents exposed by inhalation in vivo, recent studies have shown that it induced cytogenetic damage in the cells of
The INDEX project Final report
46
tissues that are more locally exposed, either by gavage or by inhalation. Rats given 200 mg per kg body weight
formaldehyde orally were killed 16, 24 or 30 hours after treatment and examined for the induction of micronuclei and
nuclear anomalies in cells of the gastrointestinal epithelium. The frequency of mitotic figures was used as an index of
cell proliferation. Treated rats had significant (greater than five-fold) increases in the frequency of micronucleated cells
in the stomach, duodenum, ileum and colon; the stomach was the most sensitive, with a 20-fold increase in the
frequency of micronucleated cells 30 hours after treatment, and the colon the least sensitive. The frequency of nuclear
anomalies was also significantly increased at these sites. These effects were observed in conjunction with signs of
severe local irritation (Migliore et al., 1989).
In another experiment, rats were exposed to 0, 0.62, 3.7 or 18.5 mg/m
3
formaldehyde for 6 hours per day on five days
per week, for one and eight weeks. There was no significant increase in chromosomal abnormalities in the bone-marrow
cells of formaldehyde-exposed rats relative to controls, but there was a significant increase in the frequency of
chromosomal aberrations in pulmonary lavage cells (lung alveolar macrophages) from rats that inhaled 18.5 mg/m
3
formaldehyde. Aberrations, which were predominantly chromatid breaks, were seen in 7.6% and 9.2% of the scored
pulmonary lavage cells from treated animals, and in 3.5% and 4.8% of the cells from controls, after one and eight
weeks, respectively (Dallas et al., 1992).
The spectrum of mutations induced by formaldehyde has been studied in human lymphoblasts in vitro, in Escherichia
coli, and in naked pSV2gpt plasmid DNA (IARC, 1995; Crosby et al. 1988). About 50% of formaldehyde-induced
tumours of the nasal epithelium of rats appeared to have a point mutation in the p53 tumour suppressor gene (Recio et
al., 1992).
Overall, formaldehyde was genotoxic in a variety of experimental systems, ranging from bacteria to rodents, in vivo.
Formaldehyde given by inhalation or gavage to rats in vivo induced chromosomal anomalies in lung cells and
micronuclei, respectively, in the gastrointestinal tract (IARC, 1995).
Interactions with other chemicals
A group of 24 healthy nonsmokers were exposed while engaged in intermittent heavy exercise for 2 hours to
formaldehyde at 3.7 mg/m
3
or to a mixture of formaldehyde and 0.5 mg/m
3
of respirable carbon aerosol, in order to
determine whether adsorption of formaldehyde on respirable particles elicits a pulmonary response. Small (<5%)
decreases were seen in forced vital capacity and forced expiratory volume, but these effects were not considered to be
clinically significant (Green et al. 1989), Risby et al. (1990) and Rothenberg et al. (1989) estimated that the amount of
formaldehyde adsorbed on to carbon black or dust particles and delivered to the deep lung by particle inhalation is
minuscule in relation to the amount that remains in the vapour phase and is adsorbed in the upper respiratory tract.
The industrial manufacture of furniture, plywood and particle board may entail simultaneous exposure to formaldehyde
and wood dust, both being nasal carcinogens, the former in rats and the latter in humans (IARC, 1995). While the
epidemiological studies in woodworkers revealed the cancer excess to be attributable to wood dust per se rather than to
other exposures in the workplace such as formaldehyde, it is also true that exposure to wood dust is often an important
confounder in epidemiological studies on formaldehyde in industrial groups (IARC, 1995), indicating that exposure to
formaldehyde may enhance the nasal cancer risk associated with wood dust exposure.
Lam et al. (1985) studied the effects of inhalation co-exposure to acrolein and formaldehyde in male Fischer 344 rats.
Rats were exposed for 6 hours to room air (controls), 2 ppm acrolein, 6 ppm formaldehyde, or a combination of 2 ppm
acrolein and 6 ppm formaldehyde. The animals were sacrificed immediately after completion of exposure and their
nasal tissues were harvested. Exposure to formaldehyde significantly increased the percentage of interfacial DNA (a
measure of DNA-protein cross linking) compared to rats exposed to room air only (12.5 versus 8.1%, p<0.05). Co-
exposure to acrolein resulted in further increases in the percentage of interfacial DNA (18.6%) which were significantly
greater than the effect of formaldehyde alone (p<0.05). The authors concluded that simultaneous exposure to acrolein
enhanced formaldehyde-induced DNA-protein cross linking and that depletion of glutathione by acrolein inhibited the
metabolism of formaldehyde, thereby increasing formaldehyde-induced DNA-protein cross link formation.
To investigate the possibility of additive or potentiating interactions between inhaled aldehydes, Cassee et al. (1996)
compared responses in nasal epithelial histopathology and cell proliferation in groups of male Wistar rats exposed for 3
days (6 hours/day) to 1.0, 3.2, or 6.4 ppm formaldehyde alone; to 0.25, 0.67, or 1.40 ppm acrolein alone; to 750 or
1,500 ppm acetaldehyde alone; or to several mixtures of these aldehydes. At the concentrations tested, the histological
and cell proliferation responses measured in the nasal epithelium of rats exposed to the mixture which produced effects
(3.2 ppm formaldehyde; 1,500 ppm acetaldehyde; 0.67 ppm acrolein) were attributed by the investigators to the acrolein
alone with no additional effects from the formaldehyde or acetaldehyde. The investigators concluded that combined
The INDEX project Final report
47
exposures to these aldehydes at exposure levels in the vicinity of individual no-effect-levels was not associated with a
greater hazard than that associated with exposure to the individual chemicals.
Odour perception
Source: WHO (2001)
Formaldehyde has a pungent odour with odour detection thresholds as specified in Table 5.1 (0.03 – 0.6 mg/m
3
); the
odour recognition threshold is not known. Formaldehyde poses nuisance problems in indoor environments owing to its
release from building materials or furnishings. Indoor air usually contains other organic compounds which, in
combination with formaldehyde or by themselves, may have odorous and irritating properties causing discomfort. It has
been reported that some sensitive individuals can sense formaldehyde concentrations of 0.01 mg/m
3
and perhaps even
lower as a “warm” feeling on the face (Ahlström et al., 1986).
Source: Devos et al. (1990)
Odour threshold: 35 µg/m
3
.
Source: IPCS/WHO (1989)
The absolute odour threshold for formaldehyde is between 0.06 and 0.22 mg/m
3
(a group of observers detected the
odour in 50% of the presentations, 10% of an untrained population detected a level of 0.03 mg/m
3
). There is a low
probability that human beings will be able to detect formaldehyde in air at concentrations below 0.01 mg/m
3
.
Source: Moncrieff (1955)
The difference between odour and irritation concentration may be noticeable, but there is no evidence that there is a
threshold at which odour is superceded by irritation. However, for most inhaled odorous compounds, the trigeminal
nerve has a higher threshold than the olfactory nerve. When the formaldehyde concentration is increased and affects
both the eyes and the nostrils, sensory irritation is first experienced in the eyes, then the odour is perceived, and
finally nasal irritation occurs.
Summary of Formaldehyde Dose Response Assessment
________________________________________________________________________________________________
Exposure other than inhalation: Endogeneous formaldehyde levels in blood: 2-3 mg/litre
Toxicokinetics: Almost completely absorbed (>90%) in the upper respiratory tract; endogenous formaldehyde levels in
blood not altered after exposure; does not accumulate, rapid conversion to formate
Health effect levels of short- and long-term exposure (noncancer):
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
Short-term exposure
0.6
EXP
1.2
EXP
0.53
BC05
0.94
1h-ADJ-BC05
Mild and moderate eye irritation Volunteers, 3h Kulle et al. (1987) OEHHA 1999;
REL: 0.094
0.5
EXP
Nasal and eye irritation
Volunteers, 2h
(potentially sensitive
population)
Pazdrak et al. (1993) ATSDR 1999;
MRL: 0.05
0.24
STAT
Respiratory tract irritation cited in Sloof et al.
(1992)
RIVM 1992
0.1 Nose and throat irritation Human, short-term cited in IPCS/WHO,
1989
WHO 2000
(0.037) Respiratory symptoms (depending on study) Children; additional
investigation requested
Krzyzanowski et al.,
(1990); Wantke et al.
(1996); Garrett et al.
(1999); Franklin et al.
(2000)
UCLA 2001
Long-term exposure
0.09
Study
0.032
ADJ
0.26
Study
0.093
ADJ
Nasal and eye irritation, nasal obstruction,
and lower airway discomfort;
histopathological nasal lesions including
rhinitis, squamous metaplasia, and dysplasia
Occupational, 10y
average;
Wilhelmsson and
Holmstrom, 1992;
supported by Edling et
al., 1988
OEHHA 1999;
REL: 0.003
0.3
Study
Mild irritation of the eyes and upper Occupational, 10.4y Holmstrom et al., 1989 ATSDR 1999;
The INDEX project Final report
48
respiratory tract and mild damage to the
nasal epithelium
MRL: 0.01
0.31
STAT
Sensory irritation
for low but significant
percentage of exposed
workers
Weighting the total
body of data
NIWL 2003
0.12 Symptoms of irritation
LOAEL may be lower
only for a very small
proportion of the
population
well conducted studies Health Canada
1999; (noTC)
Carcinogenicity: IARC: 1 (15.06.04); U.S.EPA: B1; Unit risk (EPA-IRIS): 1.3E-5 (µg/m
3
)
-1
; An association exists
between cytotoxic, genotoxic, and carcinogenic effects. Most probably HCHO cannot act as a complete carcinogen at
non-cytotoxic concentrations. If exposure is accompanied by recurrent tissue damage at the site of contact, it may be
assumed to have carcinogenic potential in humans. Evidence of carcinogenicity in rats: induction of squamous cell
carcinoma at > 18 mg/m
3
.
In June 2004, IARC announced there was sufficient evidence that formaldehyde causes nasopharyngeal cancer in
humans and re-classified it as a Group 1, known human carcinogen (previously classified as Group 2A). IARC also
reported there was limited evidence that formaldehyde exposure causes nasal cavity and paranasal cavity cancer and
strong but not sufficient evidence linking formaldehyde exposure to leukemia.
Genotoxicity: Mutagenic in vitro, in vivo only in tissue of contact at doses adapt to give cytotoxic effects (no primary
mutagen)
Odour threshold: range: 0.03 - 0.6 mg/m
3
(WHO); 0.035 mg/m
3
(Devos); > 6 mg/m
3
untolerable to most people
Susceptible population: Wide variability in response to HCHO irritancy; Children seem to bee more sensitive than
adults (still additional investigation requested; results would be expected from recent German environmental survey
GerES IV); Asthmatics: no significant changes in pulmonary function and non-specific airway reactivity at 3.7 mg/m
3
;
population with allergic contact dermatitis (3-6%) seem not to react differently at 0.5 mg/m
3
(Pazdrak, 1993: 9
volunteers with formalin skin sensitization).
Remarks: RD
50
: 4 mg/m
3
; HCHO is a reaction product of ozone and unsaturated VOCs (e.g. limonene, pinene,
styrene); neither dose addition nor potentiating interactions occured upon exposure to aldehyde mixtures
(formaldehyde, acetaldehyde, acrolein) at exposure levels around the MOEL.
________________________________________________________________________________________________
The INDEX project Final report
49
6. Risk Characterization
Health hazard evaluation
Predominant symptoms of formaldehyde exposure in humans are irritation of the eyes, nose and throat, together with
concentration-dependent discomfort, lachrymation, sneezing, coughing, nausea and dyspnoea. Although there is
substantial variation in individual responses to formaldehyde in humans, there are sufficient data to indicate that the
irritant effects on the nose and throat after short-term exposure occur at concentrations as low as 0.1 mg/m
3
(Health
Canada; WHO), set as the LOAEL. A NOAEL of 0.03 mg/m
3
is defined according to the lowest concentration supposed
not to give effects in the normal population (OEHHA). This level has been further devided by an assessment factor of
30, taking into account the evidence indicating that children may be more sensitive to formaldehyde respiratory toxicity
than adults (3) and intraspecies variation (10) resulting in an Exposure Limit (EL) of 1 µg/m
3
. For this chemical the
observed acute effects and endpoints are consistent with the pathology seen in long-term studies. Consequently, no
distinction has been done between short-term and long-term ELs.
Percentage of population exposed beyond given threshold levels
Table 6.1
Population-based studies
LOAEL
a
NOAEL
b
NOAEL
3
NOAEL
30
Description (Study, Year) N 0.1 mg/m
3
0.03 mg/m
3
0.01 mg/m
3
0.001 mg/m
3
England (BRE) 528 < 35 % 90 % 100 %
England (BRE, 2002) 833 < 35 % 85 % 100 %
Vienna – bedrooms (Hutter, 2001) 160 < 40 % 90 % 100 %
Paris – refurbished flats (Clarisse, 03) 61 < 40 % 90 % 100 %
Helsinki (Expolis, 97) 15 < 65 % 100 % 100 %
French National Survey 7-d TWA (IAQ
observatory, 2003-04)
201 < 20 % 90 % 100 %
< out of the evaluation range (i.e. <5% of the environments investigated)
a
corresponds with 30-min average WHO guideline value
b
corresponds with lowest odour threshold reported
Cancer risk evaluation
On June 15, 2004, IARC announced there was sufficient evidence that formaldehyde causes nasopharyngeal cancer in
humans and re-classified it as a Group 1, known human carcinogen (previous classification: Group 2A). IARC also
reported there was limited evidence that formaldehyde exposure causes nasal cavity and paranasal cavity cancer and
“strong but not sufficient“ evidence linking formaldehyde exposure to leukemia.
Pending the outcome of the current IARC revision, the position taken in the present indoor risk evaluation is as follows:
Inhalation of formaldehyde under conditions that induce cytotoxicity and sustained regenerative proliferation within the
respiratory tract is considered to present a carcinogenic risk to humans. At airborne levels for which the prevalence of
sensory irritation is low (i.e., 0.1 mg/m
3
), risks of respiratory-tract cancers for the general population are exceedingly
low.
Comments
Among the available exposure data (see chapter 3), results from a limited number of indoor air surveys enabled the
construction of frequency distributions and hence the evaluation of population percentages exposed at given threshold
levels (see Table 6.1). When taking into account the whole body of published exposure data, the formaldehyde levels
reported in these surveys could be considered as indicative for “indoor air quality non-problem” buildings in the EU.
As summarised in Table 6.1, the results of these measurements indicate that at least more than 20% of the population is
exposed at formaldehyde levels higher than the NOAEL. A concentration slightly higher than the established NOAEL
has been recently proposed as a LOAEL (37 µg/m
3
) for effects on pulmonary functions in children, although additional
investigation is required to confirm this level. Concerning childrens’ susceptibility, important results are expected from
the German environmental survey GerES IV.
An exposure limit of 1 µg/m
3
has been derived, which corresponds more or less with the background outdoor level in
rural areas. It could be assessed that indoors almost the entire population in the EU is exposed at levels which are higher
than this limit.
The INDEX project Final report
50
For safety check purposes indoor formaldehyde levels are habitually refered to the WHO-air quality guideline value *
(100 µg/m
3
as a 30-minute average), with the assumption that transient sensory effects would be avoided for most
people. It is here asserted that this level should be regarded as hazardous for susceptible individuals (children) when
evidence exists that concentrations are maintained over prolonged periods.
* WHO-air quality guideline value:
The lowest concentration that has been associated with nose and throat irritation in humans after short-term exposure is 0.1 mg/m
3
,
although some individuals can sense the presence of formaldehyde at lower concentrations. To prevent significant sensory irritation
in the general population, an air quality guideline value of 0.1 mg/m
3
as a 30-minute average is recommended. Since this is over one
order of magnitude lower than a presumed threshold for cytotoxic damage to the nasal mucosa, this guideline value represents an
exposure level at which there is a negligible risk of upper respiratory tract cancer in humans.“
Result
Because of its high chemical reactivity, formaldehyde is the most important sensory irritant among the chemicals
assessed in the present report. Due to its ubiquitousness in indoor environments and to the increasing evidence
indicating that children may be more sensitive to formaldehyde respiratory toxicity than adults, it is considered a
chemical of concern at levels exceeding 1 µg/m
3
, a concentration more or less corresponding with background levels in
rural areas. Results from available exposure data, although limited, confirm that almost the entire population is exposed
indoors at levels (Median level±sd: 26±6 µg/m
3
; 90th±sd: 59±7 µg/m
3
; N = 6) higher than this background level, here
established as the limit of exposure, with more than 20% of the European population exposed at levels exceeding the
no-observed-effect-level (NOAEL: 30 µg/m
3
). Within the reported concentration range mild irritation of the eyes could
be experienced by the general population as well as the odour perceived starting from about 30 µg/m
3
.
Reported formaldehyde concentrations are lower (99th < 150 µg/m
3
) than a presumed threshold for cytotoxic damage to
the nasal mucosa and hence considered low enough to avoid any significant risk of upper respiratory tract cancer in
humans. The last statement could be subjected to changes due to the current IARC revision of the carcinogenicity of
formaldehyde.
The INDEX project Final report
51
Carbon monoxide
Synonyms: Carbon oxide, Carbonic oxide
CAS Registry Number: 630-08-0
Molecular Formula: CO
1. Compound identification
Carbon monoxide is a colourless, practically odourless and tasteless gas that is poorly soluble in water, but it is soluble
in alcohol and benzene. It is a product of incomplete combustion of carbon-containing fuels. Carbon monoxide burns
with a violet flame and it is classified as an inorganic compound. It has a slightly lower density than air. Anthropogenic
emissions are responsible about two-thirds of the CO in the atmosphere and natural emissions account for the remaining
one-third Small amounts of carbon monoxide are also produced endogenously (EPA 2000, Alm 1999).
Accidental high exposures to CO can lead to acute health effects that can be fatal. Carbon monoxide exposure causes
unintentional and suicidal poisonings, and a large number of deaths annually both in Europe and in the United States. It
is estimated that more than half of the 6000 annual deaths from fires in the Unites States is caused by CO poisoning
(Raub et al 2000). It is obvious that such homes exist where CO concentrations are high enough to increase chronic
health effects, especially among sensitive populations such as pregnant women, the fetus, children, the elderly, and
individuals suffering from anemia or other diseases that restrict oxygen transport between blood and cells (IEH 1998).
In the human body, CO reacts with haemoglobin to form carboxyhaemoglobin (HbCO). The relationship of CO
exposure and the HbCO concentration in blood is presented in Table 1.1.
The scientific literature on carbon monoxide sources, concentrations, and human exposures and health effects in
outdoor and indoor environments has been comprehensively reviewed by the US Environmental Protection Agency
(EPA) and the health risk assessment has been carried out by WHO (EPA 2000, WHO 1979 and 2000). Raub et al
(2000) have also recently reviewed literature about carbon monoxide poisoning from a public health perspective.
Table 1.1. The relationship between carbon monoxide exposure and carboxyhaemoglobin levels in blood (adopted from
WHO 1979)
% HbCO
mg/m
3
ppm % in air
0.87 5.7 5 0.0005
1.73 11.5 10 0.001
3.45 23 20 0.002
5.05 34.5 30 0.003
6.63 46 40 0.004
8.16 57.5 50 0.005
9.63 69 60 0.006
11.08 80 70 0.007
12.46 92 80 0.008
13.8 103 90 0.009
15.11 114.5 100 0.01
16.37 126 110 0.011
17.6 130 120 0.012
18.78 149 130 0.013
19.95 160 140 0.014
21.05 172 150 0.015
22.15 183 160 0.016
23.23 195 170 0.017
24.26 206 180 0.018
25.25 218 190 0.019
26.22 229 200 0.02
The INDEX project Final report
52
2. Physical and Chemical properties
Molecular weight (g/mol) 28.01
Melting point (oC ) -205.1
Boiling point (oC) -191.5
Density, at 0°C, 1 atm (kg/m
3
) 1.250
at 25°C, 1 atm (kg/m
3
) 1.145
Relative density (air =1) 0.967
Solubility in water (at 0°C, 1 atm, ml/100 ml) 3.54
(at 25°C, 1 atm, ml/100 ml) 2.14
(at 37°C, 1 atm, ml/100 ml) 1.83
Conversion factors at 20 °C and 760 mm Hg:
1 ppm = 1.164 mg/m
3
1 mg/m
3
= 0.859 ppm
Sources: WHO 1979, 2000a, Verschueren 2001
3. Indoor Air Exposure assessment
Indoor air and exposure concentrations
Residential indoor concentrations of CO were typically lower in Northern Europe than in Central Europe, where they
were again lower than in Southern Europe based on the European exposure study, EXPOLIS (Georgoulis et al 2002).
Average residential indoor concentration in Helsinki was 1.2 mg/m
3
for non-ETS population. Average 48-hour
exposure to CO, being 1.4 mg/m
3
, was slightly higher than the respective indoor concentration. In Basle and Prague
average exposures to CO were higher than in Helsinki, but lower than in Milan and Athens. The highest geometric
mean exposure concentrations were found in a subpopulation of smokers in Athens, being 4.0 mg/m
3
(Georgoulis et al
2002). Average residential indoor CO concentrations in Milan vary from 2.1 to 3.9 mg/m
3
. In another Italian study
carried out by Maroni et al (1996) an average personal exposure of 2.2 mg/m
3
was reported for office workers. Weekly
average CO concentrations in 14 homes in UK ranged from 0.2 to 2.7 mg/m
3
in a study published by Ross (1996). Alm
et al (2000) reported mean exposures of 15-min, 1-hour and 8-hour periods for preschool children in Finland, being 8.4
mg/m
3
, 6.0 mg/m
3
and 3.3 mg/m
3
, respectively.
In UK typical ambient background CO levels range between 0.01 and 0.23 mg/m
3
, but in traffic environments, the 8-
hour mean concentrations are much higher, but typically less than 20 mg/m
3
(IEH 1998).
The cumulative distributions of the indoor, 48-hour and 1-hour personal exposure concentrations of carbon monoxide in
European studies are presented in Figure 3.1, Figure 3.2, Figure 3.3 and Figure 3.4.
The INDEX project Final report
53
0
20
40
60
80
100
012345
Indoor concentration (mg/m
3
)
Cumulative frequency (%)
Hel
Mil
Fre
Figure 3.1. Cumulative frequency distributions of indoor air concentrations of carbon monoxide in Helsinki (Hel, n=
258785) for non-ETS population (Scotto di Marco et al 2003), in the French Survey (Fre n= 550666) (Kirchner 2004)
and in Milan (Mil, n= 4822) (Bruinen de Bruin et al 2004) for the whole population (sampling periods were 1-min, 15-
min, and 5-min in Helsinki, Milan and France, respectively).
0
20
40
60
80
100
0.00.51.01.52.0
Indoor concentration (mg/m
3
)
Cumulative frequency (%)
CO 14-day
Figure 3.2. Cumulative frequency distribution of 14-day indoor air concentrations of carbon monoxide in the kitchens in
England (GM= 0.47 mg/m3, max 4.45 mg/m3, n=830, Raw et al 2002).
The INDEX project Final report
54
0
20
40
60
80
100
0123456
Exposure (mg/m
3
)
Cumulative frequency (%)
Ath
Bas
Hel
Mil
Pra
Figure 3.3. Cumulative frequency distributions of 48-hour exposures to carbon monoxide in Athens (Ath), Basel (Bas),
and Prague (Pra) (Georgoulis et al 2002), Helsinki (Hel) (EXPOLIS 2002), and Milan (Mil) (Bruinen de Bruin et al
2004).
0
20
40
60
80
100
0 1020304050
Exposure (mg/m
3
)
Cumulative frequency (%)
Ath
Bas
Hel
Mil
Oxf
Pra
Figure 3.4. Cumulative frequency distributions of 1-hour exposure concentrations of carbon monoxide in Athens (Ath,
n=44), Basel (Bas, n=50), Helsinki (Hel, n=195), Milan (Mil, n=45) Oxford (Oxf, n=26) and Prague (Pra, n=23)
(Hanninen et al 2004).
Emission sources
The most common cause of high carboxyhaemoglobin concentrations in man is the smoking of tobacco and the
inhalation of the products by the smoker. Faulty domestic cooking and heating appliances, inadequately vented to
outside air, may cause high indoor concentrations of CO (WHO 1979). Also gas stoves, water heaters, and exhaust from
vehicles in attached garages might be important indoor sources (Alm 1999).
The most important source of carbon monoxide in ambient air is the exhaust of gasoline-powered motor vehicles. The
emission rate depends on the type of vehicle, its speed, and its mode of operation. Other common ambient sources
include heat and power generators, especially when using coal, industrial processes such as the carbonisation of fuel,
and the incineration of refuse (WHO 2000).
The INDEX project Final report
55
Based on the EXPOLIS results average residential indoor CO concentrations in Milan were the lowest when no special
indoor sources were present and the highest if gas cooking and environmental tobacco smoke (ETS) were present. The
highest short-time peak concentration was found during gas cooking, 7.4 mg/m
3
. Average 15-min ambient CO
concentration, 2.6 mg/m
3,
was higher than the respective indoor concentration when no indoor sources were present, but
lower if gas cooking or ETS was present. The highest in-transit concentration was measured in cars/taxis being 6.5
mg/m
3
(Bruinen de Bruin et al 2003). Average 1-hour exposures to CO were higher, 8.4 mg/m
3
, than the respective
ambient concentrations, 5.7 mg/m
3
. Instead, average 8-hour and 48-hour ambient concentrations were at the same level
than the respective exposures, being 3.8 and 2.4 mg/m
3
, respectively.
Short time carbon monoxide concentrations related to some typical indoor sources such as tobacco smoke, gas cooking
and commuting in five European cities are presented in Figure 3.5. Much higher concentrations were found in a Finnish
study determining personal exposures of preschool children in Helsinki. The highest exposures to carbon monoxide
were as high as 80 mg/m
3
, 69 mg/m
3
and 28 mg/m
3
for 15-min, 1-hour and 8-hour averages, respectively. Elevated
exposures were related to gas stoves, mothers’ smoking and living in high rise buildings (Alm et al 2000).
Brown et al (2002) studied emission rates and concentrations caused by unflued gas heaters in a room chamber with a
volume of 32.4 m
3
. In normal conditions carbon monoxide concentrations ranged from <1.15 mg/m
3
to 4.6 mg/m
3
. But
when the gas supply was restricted due to the backpressure of 0.02 kPa, concentration of 19.5 mg/m
3
was measured.
Even higher concentration, 34.4 mg/m
3
, was determined when the researchers caused a slight misalignment of the
burner. Because fatal concentrations may occur during malfunction of burning appliances, estimated concentrations
caused by burning different fuels in a stove are presented in Table 3.1. Despite of the fact that the assumptions are valid
for houses common in developing countries, these estimates may help to assess the concentrations that may be caused
by burning these fuels in stoves also in developed countries.
Typical sources and microenvironments where high concentrations of carbon monoxide may occur are reviewed in
Table 3.2 based on the recently published studies. The highest maximum values ranging 121 – 182 mg/m
3
were
measured in homes when using a gas grill attached to the gas stove (IEH 1998), in the underground parking facilities (El
Fadel et al 2001), and in a home with a faulty boiler (Ross 1996). Elevated concentrations were also found in other
microenvironments such as motor vehicles, indoor ice arenas, bars and restaurants. Carbon monoxide concentrations in
public buildings studied before 1985 are summarised in Table 3.3.
Lately attention is paid to incense burning in homes and other public buildings including stores and shopping malls.
Jetter et al (2002) reported emission rates of 23 different types of incense such as incense rope, cones, sticks, rocks,
powder etc. that are typically used indoors. The measured emission rates of carbon monoxide ranged 144 - 531 mg/h.
The authors estimated a peak concentration of 9.6 mg/m
3
caused by incense burning and, therefore concluded that
carbon monoxide concentrations could exceed the US EPA’s National Ambient Air Quality Standard (NAAQS) 10
mg/m
3
for an 8-hour average depending on the room volume, ventilation rate and the amount of incense burned. Lee
and Wang (2004) reported similar results when studying emissions of incense burning in chamber (18 m
3
) tests. They
measured maximum carbon monoxide concentrations up to 44 mg/m
3
during burning and concluded that incense
burning is an important indoor air pollution source in addition to carbon monoxide also to fine particles and VOCs.
Especially, incense burning might be a significant contributor to population exposure in such cultures, where incense is
burned frequently, for example in religious rituals.
The INDEX project Final report
56
0
1
2
3
4
5
ETS non-ETS Gas
cooking
No gas
cooking
Car Bus-
tram
Metro-
train
Concentration (mg/m
3
)
Ath
Bas
Hel
Mil
Pra
Figure 3.5. Short time (geometric means of the 15-min periods) exposures to carbon monoxide stratified by
environmental tobacco smoke (ETS) and gas cooking, and in-vehicle concentrations in European cities Athens (Ath),
Basel (Bas), Helsinki (Hel), Milan (Mil) and Prague (Pra) (Georgoulis et al 2002).
Table 3.1. Estimated indoor CO concentrations resulting from burning 1 h of fuel in stove without flues using
assumptions relevant in developing countries i.e. fuel consumption equal to the energy used for burning 1.7 kg of fuel
wood, ventilation rate of 15 h-1, and room volume of 40 m3 (adopted from Zhang et al 1999).
Fuel
Avgerage during burning maximum
Charbiquette 562 603
Charcoal 528 566
Brush wood 511 548
Coal 178 191
Fuel wood 150 161
Kerosene 9.4 10
LPG 4.5 4.8
Biogas 1.6 1.7
Natural gas 0.083 0.089
CO concentration (mg/m
3
)
The INDEX project Final report
57
Table 3.2. Short time CO concentrations related to specific microenvironments or emission sources.
Environment/
emission source
Location Averaging time Reference
A
MGMmax
Gas stove with pilot light UK peak when using a grill 10-182 IEH 1998
Underground parking Beirut, Lebanon 30-min 80 26 - 140 El Fadel et al 2001
facilities
Home UK 1-min faulty boiler 121 Ross 1996
gas cooking 6 - 49
all-electric homes 3.5 - 4
Undergrounds, car parks, several hours >115 60 - 115 WHO 2000
enclosed ice arenas, etc.
homes with gas appliances
Gas appliances The Netherlands max 1-min kitchen 5-108 Lebret et al 1987
max 1-hour kitchen 3-56
Exposure Helsinki, Finland max 15-min pre-school 80 Alm et al 2000
max 1-hour children 69
max 8-hour 28
Incense burning peak chamber tests 44 Lee and Wang 2004
Bars, restaurants Italy 12 - 23 35 Maroni et al 1995
Indoor ice arenas Finland max 1-hour 20-33 Pennanen et al 1997
Exposure Milan, Italy max 15-min 12 11 25 Bruinen de Bruin 2002
max 1-hour 8.4 8.4 20
max 8-hour 3.8 3.8 9.3
Unflued gas heaters room chamber test 1-min normal cond. <1.2 - 4.6 Brown et al 2002
gas supply restricted 20
misaligned burner 34
Shops and bars Genoa, Italy 8-hour shops 15 Valerio et al 1997
bars 18
Indoor ice skating rink Hong Kong, China 15-min gasoline-fueled 8-16 Guo et al 2004
15-min propane-fueled 5.7
Exposure Helsinki, Finland max 15-min ETS 9.0 5.7 Scotto di Marco et al 2003
non-ETS 7.1 5.3
max 1-hour ETS 5.7 3.7
non-ETS 4.3 3.3
max 8-hour ETS 2.6 2.1
non-ETS 2.0 1.8
Concert hall Switzerland event (5.5 hours) 3.5 5.2 Junker et al 2000
Gas appliances NY, USA RTI 1990
3-day gas stove 2.6
kerosene heater 3.9
Public buildings Athens, Greece 1-hour Office 3.7 Chaloulakou et al 2003
AM = arithmetic mean, GM = geometric mean, max = maximum value
Concentration (mg/m
3
)
The INDEX project Final report
58
Table 3.3. Indoor air concentrations of carbon monoxide in selected indoor microenvironments (adopted from Akland et
al 1985).
Indoor environment
n ppm std
mg/m
3
Public garage 116 13.46 18.14 15.41
Service station/motor vechile repair facility 125 9.17 9.33 10.50
Repair shop 55 5.64 7.67 6.46
Shopping mall 58 4.90 6.50 5.61
Residential garage 66 4.35 7.06 4.98
Restaurant 524 3.71 4.35 4.25
Office 2287 3.59 4.18 4.11
Sport arena, concert hall 100 3.37 4.76 3.86
Store 734 3.23 5.56 3.70
Health care facility 351 2.22 4.25 2.54
Other public buildings 115 2.15 3.26 2.46
Residence 21543 2.04 4.06 2.34
School 426 1.64 2.76 1.88
Church 179 1.56 3.35 1.79
Mean concentration
The INDEX project Final report
59
4. Toxicokinetics
Endogenous sources of carbon monoxide:
In addition to inhaled carbon monoxide, humans are also exposed to small amounts of carbon monoxide produced
endogenously, conducing in the healthy population to a baseline carboxyhaemoglobin concentration range of 0.4-0.7%.
Sources of carbon monoxide in the body include the endogeneous oxidation of halomethanes (e.g. dichloromethane),
phenols, flavonoids, as well as the lipid peroxidation of membrane lipids (Rodgers et al., 1994).
Carbon monoxide is a natural degradation product of haemoproteins, mainly haemoglobin* but also myoglobin,
cytochromes, peroxidases and catalase. Approximately 0.4 ml/h CO is formed by haemoglobin catabolism, and about
0.1 ml/h originates from non-haemoglobin sources (Coburn et al., 1964). In both males and females, week-to-week
variations in carbon monoxide production are greater than day-to-day or within-day variations. In females,
carboxyhaemoglobin levels fluctuate with the menstrual cycle; the mean rate of carbon monoxide production in the
premenstrual, progesterone phase almost doubles (Delivoria-Papadopoulos et al., 1970; Lynch & Moede, 1972).
Neonates and pregnants also showed a significant increase in endogenous carbon monoxide production related to
increased breakdown of red blood cells.
Any disturbance leading to increased destruction of red blood cells and accelerated breakdown of other haemoproteins
would lead to increased production of carbon monoxide. Haematomas, intravascular haemolysis of red blood cells,
blood transfusion and ineffective erythropoiesis will all elevate the carbon monoxide concentration in the blood.
Degradation of red blood cells under pathological conditions such as anaemias (haemolytic, sideroblastic, sickle cell),
thalassaemia, Gilbert’s syndrome with haemolysis and other haematological diseases will also accelerate carbon
monoxide production (Berk et al., 1974; Solanki et al., 1988). In patients with haemolytic anaemia, the carbon
monoxide production rate was 2–8 times higher and blood carboxyhaemoglobin concentration was 2–3 times higher
than in normals (Coburn et al., 1966). As a result, baseline carboxyhaemoglobin levels can be as high as 4%. In subjects
with haemoglobin Zürich, where affinity for carbon monoxide is 65 times that of normal haemoglobin,
carboxyhaemoglobin levels range from 4% to 7%.
Increased carbon monoxide production rates have been reported after administration of phenobarbital, progesterone
(Delivoria-Papadopoulos et al., 1970) and diphenylhydantoin (Coburn, 1970).
* In the process of natural degradation of haemoglobin to bile pigments, in concert with the microsomal reduced nicotinamide
adenine dinucleotide phosphate (NADPH) cytochrome P-450 reductase, two haem oxygenase isoenzymes, HO-1 and HO-2, catalyse
the oxidative breakdown of the alpha-methene bridge of the tetrapyrrol ring of haem, leading to the formation of biliverdin and
carbon monoxide. The production of haem oxygenase isoenzyme HO-1 is increased by any kind of stress, including heat, light,
sound, odors, infection, physical trauma and mental or psychological stress. Chronic stress in any of these pathways thus results in
chronic destruction of haem and chronic low-level CO poisoning. Stress-induced HO-1 activity and the relatively constant activity of
isozyme HO-2, that does not respond to stress, together account for about 75% of the human body's CO production.
Absorption
After reaching the lungs, inhaled carbon monoxide diffuses rapidly across the alveolar and capillary membranes. It also
readily crosses the placental membranes. Carbon monoxide binds reversibly to one of the haem proteins. Approximately
80–90% of the absorbed carbon monoxide binds with haemoglobin, which causes a reduction in the oxygen-carrying
capacity of the blood. The affinity of haemoglobin for carbon monoxide is 200–250 times that for oxygen, while the
relative affinities of other haem proteins (e.g. myoglobin), cytochrome oxidase and cytochrome P-450 for carbon
monoxide are much lower (U.S.EPA, 1991; ACGIH, 1991ab).
When in equilibrium with ambient air, the carboxyhaemoglobin (COHb) content of the blood will depend mainly on the
concentrations of inspired carbon monoxide and oxygen. If equilibrium has not been achieved, the COHb concentration
will also depend on the duration of exposure, pulmonary ventilation and the COHb originally present before inhalation
of the contaminated air.
The absorption and elimination of carbon monoxide have been described in various mathematical models (U.S.EPA,
1991; WHO, 1979). The most important of these models is the Coburn-Foster-Kane exponential equation, which takes
into account all known physiological variables affecting carbon monoxide uptake (Coburn et al., 1965). The most
important variables determining the COHb level are: baseline COHb level, blood haemoglobin level, minute ventilation,
diffusion capacity of the lungs, and mean oxygen tension in pulmonary capillaries. During an exposure to a fixed
concentration of carbon monoxide, the COHb concentration increases rapidly at the onset of exposure, starts to level off
The INDEX project Final report
60
after 3 hours, and reaches a steady state after 6–8 hours (see Figure 4.1). At steady state, the carbon monoxide
concentrations in alveolar breath and ambient air become practically equal (Peterson, 1970).
Figure 4.1: Log-log plot of carbon monoxide uptake by humans from
low ambient CO concentrations as computed from the Coburn-Forster-Kane equation.
Abbreviations: Pco2 = mean partial pressure of O2 in lung capillaries, VA = alveolar ventilation rate, Vb = blood volume, M = equilibrium constant,
DL = diffusing capacity of lungs, [COHb]0 = value prior to CO exposure, Vco = rate of endogenous CO production. (From: Peterson and Stewart,
1975).
Using an adaptation of the Coburn equation the average 1 and 8 hr ambient concentration required to achieve a COHb
level of 2.5% (the target COHb concentration served as a basis for the U.S. air quality standard for CO) for children
with different baseline levels of COHb was calculated. As shown in Figure 4.2, as baseline COHb concentration
increased, the amount of inhaled CO required to raise the blood level to 2.5% was decreased.
The INDEX project Final report
61
Figure 4.2: Increasing baseline COHb will reduce the time-weighted average
CO concentration required to reach 2.5% COHb after a given exposure. (from OEHHA, 2000)
Elimination
Carbon monoxide is eliminated unchanged via the lungs. The decline in COHb concentration depends on the rate of
carbon monoxide release from haem proteins, alveolar ventilation, oxygen concentration in inhaled air, duration of
carbon monoxide exposure, and the level of COHb saturation. The formation of COHb is a reversible process, but
because of the tight binding of carbon monoxide to haemoglobin, the elimination half-life while breathing room air is
2–6.5 hours depending on the initial COHb level. The elimination half-life of COHb is much longer in the fetus than in
the pregnant mother (U.S.EPA, 1991; ACGIH, 1991ab).
5. Health effects
Effects of short-term exposure
CO affects health by interfering with the systemic transport of oxygen to tissues (especially the heart and other muscles
and brain tissue) (Costa and Amdur, 1996). The resulting impairment of O
2
delivery cause tissue hypoxia and interferes
with cellular respiration. Direct intracellular uptake of CO could permit interactions with haemoproteins such as
myoglobin, cytochrome oxidase and cytochrome P-450, and therefore interfere with electron transport processes and
energy production at the cellular level (Brown and Piantidosi, 1992). Thus, in addition to observed physiological effects
and cardiovascular effects, CO can modify electron transport in nerve cells resulting in behavioral, neurological and
developmental toxicological consequences, and may itself play a role in neurotransmission.
The health effects associated with inhaled CO vary with its concentration and duration of exposure. Effects range from
subtle cardiovascular and neurobehavioral effects at low concentrations to unconsciousness and death after prolonged
exposures or after acute exposures to high concentrations of CO. First signs and symptoms on healthy individuals, such
as decreases in work capacity and decrements of neurobehavioral functions start at [COHb] of 5% (WHO, 1999;
U.S.EPA, 2000), whereas first signs of CO poisoning appear at [COHb] concentrations of 10% (see Table 5.1).
However, the variability within the human population must be considered high. A [COHb] of about 15 % only leads to
slight symptoms, such as headache, in healthy adults (Stewart et al., 1970; WHO, 1999). In contrast, the same [COHb]
can cause long-lasting defects in the cognitive development and behavioral alterations in children (Klees et al., 1985),
or even contribute to death from myocardial infarction in individuals with coronary artery disease (Grace and Platt,
1981; Balraj, 1984).
The INDEX project Final report
62
Table 5.1: Carboxyhaemoglobin levels resulting from steady-state exposure to increasing concentrations of
CO in ambient air and associated symptoms in healthy adult humans and susceptible (adapted from
U.S.EPA, 2001; Winter and Miller, 1976; Ellenhorn and Barceloux, 1988)
[CO] in atmosphere [COHb] Signs and symptoms
ppm mg/m
3
% Healthy adults Susceptible subpopulations
0 0 0.4 – 0.7 Physiologic background concentration
10 11.5 2 Asymptomatic
17 19.5 2.9
during physical exertion reduced
time to onset of angina and
electrocardiogram signs of
myocardial ischaemia in
sunjects with coronary artery
disease
5 - 6
Decreases in work capacity and
decrements of neurobehavioral function
Increase in cardiac arrythmias in
subjects with coronary artery
disease
42 48 7 Headache, nausea in children
- - 3 - 8 Background concentration in smokers
70 80 10
No appreciable effect, except shortness
of breath on vigorous exertion; possible
tightness across the forehead; dilation of
cutaneous blood vessels.
13
Cognitive development deficits
in children
15
Myocardial infarction in subjects
with coronary artery disease
120 137 20
Shortness of breath on moderate
exertion; occasional headache with
throbbing in temples
25 syncopes in children - stillbirths
220 252 30
Decided headache; irritable; easily
fatigued; judgment disturbed; possible
dizziness; dimness of vision
350 - 520 401 – 595 40 – 50
Headache, confusion; collapse; fainting
on exertion
800 - 1220 916 - 1400 60 – 70
Unconsciousness; intermittent
convulsion; respiratory failure, death if
exposure is long continued
1950 2230 80 Rapidly fatal
At CO levels typically encountered in indoor and outdoor environments, health effects are most likely to occur in
individuals who are physiologically stressed, either by exercise or by medical conditions that can make them more
susceptible to low levels of CO. Subpopulations at increased risk of adverse effects are :
1. Individuals with cardiovascular diseases: COHb levels of 2-6% may impair the delivery of oxygen to the
myocardium causing hypoxia and increasing coronary blood flow demand by nearly 30%. When myocardial
oxygen demands are increased, as in exercise, the hypoxic effects of CO may exceed the limited coronary
reserve producing adverse health effects including earlier onset of myocardial ischaemia, reduced exercise
tolerance in persons with stable angina pectoris, increased number and complexity of arrhythmias, and
increased hospital admissions for congestive heart failure.
2. Fetuses are more susceptible to CO exposure for several reasons: CO crosses the placenta; fetal Hb has greater
affinity for CO than maternal Hb; the half-life of COHb in fetal blood is three times longer than that of
maternal blood, and the fetus has high rate of oxygen consumption and lower oxygen tension in the blood than
adults. Also, maternal smoking during pregnancy exposes the fetus to greater than normal concentrations of
CO leading to a decrease in birth weight.
3. Children develop acute neurotoxic effects (e.g. headaches, nausea), long-lasting neurotoxic effects (e.g.
memory deficits) and impaired ability to escape (i.e. syncopes) at lower [COHb] than adults. Children have
greater activity levels and smaller body masses than adults and should therefore experience higher levels of
CO uptake than will adults for the same average exposure concentration.
4. Pregnant women have increased alveolar ventilation, increasing the rate of CO uptake from inspired air. Also,
a pregnant woman produces nearly twice as much endogenous CO.
The INDEX project Final report
63
5. Individuals with chronic obstructive pulmonary disease such as chronic bronchitis, emphysema and chronic
obstructive pulmonary disease are more susceptible to CO effects, since their lungs are less efficient at
oxygenating the blood.
6. Individuals with reduced blood haemoglobin concentrations, or with abnormal haemoglobin, will have reduced
O
2
carrying capacity in blood. In addition, disease processes that result in increased destruction of red blood
cells (haemolysis) and accelerated breakdown of haemoproteins accelerate endogenous production of CO,
resulting in higher COHb concentrations than in normal individuals. For example, patients with haemolytic
anemia have COHb concentrations 2 to 3 times those seen in normal individuals.
7. Certain occupation groups are at risk from ambient CO exposure including those who work on city streets
(street repairmen, street cleaners, street vendors, deliverymen, and garage attendants, taxi and bus drivers).
Individuals who work in industrial processes including those exposed to other chemical substances (e.g.
methylene chloride) that increase endogenous CO formation.
8. Individuals who have not adapted to high altitude and are exposed to a combination of high altitude and CO.
Cardiovascular effects
Numerous controlled human studies have been conducted in healthy subjects and in patients with ischaemic heart
disease in order to characterize the effects of low-level carbon monoxide exposures on the cardiorespiratory responses
to exercise. In these experiments, the subjects have typically been exposed to clean air and carbon monoxide in a
chamber or through a face mask. After the exposure, which has usually been conducted at rest to achieve a
predetermined COHb concentration in the blood, the subjects have engaged in an exercise test on a treadmill or cycle
ergometer until exhaustion (healthy subjects) or the appearance of angina pectoris or electrocardiographic signs of
cardiac ischaemia.
In apparently healthy subjects, the maximal exercise time and the maximal oxygen consumption have decreased at
COHb levels as low as 5%. The regression between the percentage decrease in maximal oxygen consumption and the
percentage increase in COHb concentration appears to be linear, with approximately a one percentage point fall in
oxygen consumption per one percentage point rise in COHb level above 4% (U.S.EPA, 1991; Bascom et al, 1996).
Patients with cardiovascular disease, especially ischaemic heart disease, are expected to be particularly sensitive to
carbon monoxide. Atherosclerotic narrowing of the coronary arteries and impaired dilatation mechanisms restrict blood
flow to the myocardium and prevent physiological compensation for lowered oxygen delivery caused by elevated levels
of COHb. In exercise, these subjects experience myocardial ischaemia, which can impair myocardial contractility,
affect cardiac rate and rhythm, and cause angina pectoris (U.S.EPA, 1991; Bascom et al, 1996).
The early studies of Aronow et al. (1972), Aronow & Isbell (1973) and Anderson et al. (1973) have suggested that low-
level carbon monoxide exposures resulting in COHb levels of 2.5–3.0% shorten the time to onset of exercise-induced
chest pain in patients with angina pectoris. Although the validity of the studies of Aronow and colleagues has been
questioned by the US Environmental Protection Agency, subsequent studies by other investigators have actually given
similar results (U.S.EPA, 1991; Bascom et al, 1996).
The design and results of the five most important clinical studies (Kleinman et al., 1989; Allred et al.,1989; Sheps et al.,
1987; Anderson et al., 1973; Adams et al., 1988) conducted in patients with ischaemic heart disease are summarized in
Table 5.2. Despite the obvious differences between these studies, they all show a significant shortening in the time to
onset of angina at mean post-exposure COHb levels of 2.9–5.9% (post-exercise COHb levels in Table 5.2 are somewhat
lower), which represent mean incremental increases of 1.5–4.4% COHb from the pre-exposure baseline levels
(U.S.EPA, 1991).
The potential arrhythmogenic effects associated with low-level carbon monoxide exposures have not been fully
resolved at COHb levels of 5% (U.S.EPA, 1991; ACGIH, 1991b). Hinderliter et al. (1989) reported no effects at 3.5%
and 4.9% COHb levels (post-exercise concentrations) on resting and exercise-induced arrhythmias in ten patients with
coronary artery disease and no baseline ectopia. In contrast, Sheps et al. (1990) showed in 41 nonsmoking patients with
documented coronary artery disease and various levels of baseline ectopia that the frequencies of both single and
multiple ventricular depolarizations increased significantly at a mean post-exercise COHb level of 5.0% but not at
3.5%. Dahms et al. (1993) found no additional effect of either 3% or 5% COHb over the exercise-induced increases in
single or multiple ectopic beats experienced by patients with myocardial ischaemia and baseline ectopia.
The INDEX project Final report
64
Table 5.2: A summary of results of the five most important double-blind clinical studies on the effects of low-level
carbon monoxide exposures on patients with documented ischaemic heart disease and exercise-induced angina
Exposure Reference
CO
(mg/m
3
)
a
COHb
(%)
b
Exposure duration
and activity
Subject
characteristics
Effects of CO exposure (symptoms,
ECG changes, etc.)
Anderson et
al., 1973
0
57
115
1.3
2.9
4.5
4-hour exposure at
rest, post-exposure
exercise on a
treadmill
10 males, mean age
49.9 years (5 smokers, 5
nonsmokers), repro-
ducible angina
Time to onset of angina shortened at
COHb 2.9% and 4.5% (P < 0.005) and
duration of angina prolonged at
COHb 4.5% (P < 0.01).
Deeper ST-segment depressions with CO
in 5 subjects.
Kleinman et
al., 1989
0
115
1.4
2.8
1-hour exposure at
rest, post-exposure
incre-mental exercise
on a cycle ergometer
24 males, mean age
58.8 years nonsmokers
for at least 6 months),
reproducible angina
Time to onset of angina shortened by
5.9% (P = 0.046), no significant effect on
duration of angina, oxygen uptake
at angina reduced by 2.2% (P = 0.04).
Time to 0.1 mV ST-segment depression
shortened by 19.1% (P = 0.044) in 8
subjects.
Allred et al.,
1989
0
134
290
0.6
2.0
3.9
50- to 70-minute
exposure at rest, pre-
and post-exposure
incremental exercise
on a treadmill
63 males, mean age 62
years (nonsmokers),
reproducible angina
Time to onset of angina shortened by
4.2% (P = 0.054) at COHb 2.0% and by
7.1% (P = 0.004) at COHb 3.9%.
Time to threshold ischaemic ST-segment
changes shortened by 5.1% (P = 0.02) at
COHb 2.0% and by 12.1% (P < 0.0001) at
COHb 3.9%.
Significant dose relationships in the
changes of both the onset of angina (P =
0.02) and the onset of ST-segment
changes (P < 0.0001).
Sheps et al.,
1987
0
115
1.5
3.6
1-hour exposure at
rest, post-exposure
incremental exercise
on a cycle ergometer
25 males and 5 females,
mean age 58.2 years
(nonsmokers for at least
2 months), ischaemia in
a screening test
No significant changes in time to onset of
angina, duration of angina, maximal
exercise time, maximal ST-segment
depression, time to significant ST-
segment depression, or maximal left
ventricular ejection fraction.
3 subjects experienced angina only on CO
exposure, actuarial analysis including
these subjects showed shortening in time
to onset of angina in the study group (P <
0.05).
a
Carbon monoxide, 1 mg/m
3
= 0.873 ppm.
b
Carboxyhaemoglobin concentrations are from venous blood samples taken immediately after exercise; in the study of Anderson et al.
(40) samples were taken only immediately after carbon monoxide exposure.
According to some epidemiological and clinical data, carbon monoxide from recent smoking and environmental or
occupational exposures may contribute to cardiovascular mortality and the early course of myocardial infarction
(U.S.EPA, 1991). It is not known whether this contribution is due to arrhythmogenic effects or to some longer-term
effects, as suggested by some authors. In patients with severe ischaemic heart disease, carbon monoxide poisonings
have been lethal at COHb levels of 10–30%, while usual COHb levels in lethal poisonings are around 50–60%
(U.S.EPA, 1991).
A number of recent epidemiological studies reported associations between levels of ambient air pollutants (CO, PM,
O3, NOx, SO2) and hospital admissions for cardiovascular diseases (Atkinson et al., 1999; Burnett et al., 1997; Morris
et al., 1995; Linn et al., 2000; Le Tertre et al., 2002; Mann et al., 2002; Yang et al., 2004). In all the studies cited a
positive association was found between CO ambient concentrations and the daily number of cardiovascular disease
hospitalizations. at the local level (Litovitz et al., 1992; Mathieu et al., 1996).
Often, individuals suffering from CO poisoning are unaware of their exposure because symptoms are similar to those
associated with viral illness or clinical depression (Raub et al., 2000). This may result in a significant number of
misdiagnoses by medical professionals (Grace and Platt, 1981; Fisher and Rubin, 1982; Barret et al., 1985; Dolan et al.,
1987; Kirkpatrick, 1987; Heckerling et al., 1987, 1988, 1990). Although the precise number of individuals who suffer
from CO poisoning is not known, it is certainly much larger than that indicated by mortality figures. Schaplowsky et al.
(1974) estimated that more than 10 000 people per year in the United States required medical attention or missed at
least 1 day of work in the early 1970s because of sublethal exposures to CO. A recent study (Hampson, 1998) estimated
over 40 000 emergency department visits annually for recognized acute CO poisoning in the United States.
The INDEX project Final report
65
Developmental effects
The pregnant mother, the fetus in utero and the newborn infant are at high risk of adverse health effects from
atmospheric carbon monoxide exposures. During pregnancy, the endogenous production of carbon monoxide can be
elevated as much as 3-fold, the concentration of maternal haemoglobin is often reduced, and the mothers have
physiological hyperventilation. As a result of these changes, maternal COHb levels are usually about 20% higher than
the nonpregnant values. Carbon monoxide diffuses readily across the placental membranes, and the carbon-monoxide-
binding affinity of fetal haemoglobin is higher than that of adult haemoglobin. Moreover, carbon monoxide is cleared
much more slowly from fetal blood than from maternal blood. At steady state, fetal COHb levels are up to 10–15%
higher than maternal COHb levels (U.S.EPA, 1991; Longo, 1977).
There are theoretical reasons and supporting laboratory animal data to suggest that the fetus and the developing organs
are especially vulnerable to carbon monoxide. The developing brain seems to have the highest sensitivity of all organs.
There is a well established and probably causal relationship between maternal smoking and low birth weight at fetal
COHb levels of 2–10%. In addition, maternal smoking seems to be associated with a dose-dependent increase in
perinatal deaths and with behavioural effects in infants and young children. Carbon monoxide is probably one of the
most important etiological factors for these effects, although there are numerous other toxic pollutants in tobacco smoke
(Longo, 1977).
A case-control study of the association between low birthweight infants and maternal CO exposures in approximately
1000 cases in Denver (Alderman et al., 1987) failed to detect a relationship between CO exposure (estimated form
fixed-site outdoor monitoring data) during the last 3 months of pregnancy and lower birth weights. Mean CO levels
ranged from 0.6 to 4.1 mg/m
3
(0.5 to 3.6 ppm) at 8 monitoring locations in metropolitan Denver. The 5th and 95th
percentile concentrations at the site with the highest (4.1 mg/m
3
) mean were 1.8 and 5.5 mg/m
3
(1.6 and 4.8 ppm),
respectively. The odds ratio at the highest concentration site was 1.1 and the 95% confidence interval was 0.8-1.6. This
study did not directly account for unmeasured sources of CO exposure, such as smoking, emissions from gas appliances
and exposures to vehicular exhaust, which are limitations of the study design.
A more extensive study of a cohort of 125573 children born to women living in the Los Angeles area (1989-1993)
found that exposure to ambient concentrations > 6.3 mg/m
3
(3 mo average) during the last trimester of pregnancy was
associated with a significantly increased risk of low birthweight (odds ratio = 1.22; confidence interval =1.03-1.44)
after adjustment for potential confounders (Ritz and Yu, 1999). Fetotoxicity has been demonstrated in laboratory animal
studies. Altered brain neurochemical development and growth retardation have been demonstrated in rats exposed to
CO in utero (Storm and Fechter, 1985; Leichter, 1993).
Neurological and neurobehavioural effects
Central nervous system (CNS) effects in individuals suffering acute CO poisoning cover a wide range, depending on
severity of exposure: headache, dizziness, weakness, nausea, vomiting, disorientation, confusion, collapse, and coma.
At low concentrations, CNS effects include reduction in visual perception, manual dexterity, learning, driving
performance, and attention level. Earlier work is frequently cited to justify the statement that CO exposure sufficient to
produce COHb levels of ca. 5% would be sufficient to produce visual sensitivity reduction and various neurobehavioral
performance deficits. In a recent literature re-evaluation, however, the best estimate was that [COHb] would have to rise
to 15–20% before a 10% reduction in any behavioral or visual measurement could be observed (Raub and Benignus,
2002). This conclusion was based on (1) critical review of the literature on behavioral and sensory effects, (2) review
and interpretation of the physiological effects of COHb on the CNS, (3) extrapolation from the effects of hypoxic
hypoxia to the effects of CO hypoxia, and (4) extrapolation from rat behavioral effects of CO to humans.
Summary of short-term exposure effect levels
NOAEL
% COHb
LOAEL
% COHb
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
1.1-1.3 2 aggravation of angina and other cardiovascular
diseases
Subjects with angina
pectoris, 1h
Aronow, 1981
OEHHA 1999
REL: 23
2.5
- acute ischaemic heart attacks (in documented
or latent coronary artery disease)
- untoward hypoxic effects in fetuses of
nonsmoking pregnants
- Subjects with
ischaemic heart
disease
- Fetuses
WHO 2000
100 (15m)
60 (30m)
30 (1h)
10 (8h)
2.7 aggravation of angina pectoris, and other
symptoms of myocardial ischaemia
Subjects with angina
pectoris
Aronow and Isbell,
1973; Aronow et al.,
1972; Anderson et al.,
1973
U.S.EPA 1994
10.3 (8h)
35 (1h)
2.9
a
2
b
cardiorespiratory effects in individuals with
ischaemic heart disease.
Subjects with
ischaemic heart
disease performing
Kleinman et al., 1998;
Allred et al.,1989;
Sheps et al., 1987;
Health Canada
2003
MDL(1h/8h)=15/6
The INDEX project Final report
66
light work Kleinman et al., 1989;
Adams et al., 1988
MAL(1h/8h)=35/1
5
MTC(8h)=20
Health Canada - Recommended National Ambient Air Quality Objectives - MDL(1h/8h): Maximum desirable level (1and 8 hour time
weighted average); MAL: Maximum acceptable level; MTC: Maximum tolerable level
a
as measured by CO-Oximeter -
b
as measured by gas chromatography
OEHHA (1999)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Because angina is a severe effect, there is no level protective against mild adverse effects.
Derivation of Acute Reference Exposure Level (
protective against severe adverse effects, for a 1-hour
exposure)
Study Aronow, 1981
Study population humans
Exposure method inhalation
Critical effects aggravation of angina and other cardiovascular diseases
LOAEL 2% carboxyhaemoglobin in blood
NOAEL 1.1%-1.3% carboxyhaemoglobin in blood (corresponds to 20 ppm
CO,
calculated toxicokinetically)
Exposure duration 1 hour
LOAEL uncertainty factor 1
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 1
Cumulative uncertainty factor 1
Reference Exposure Level 20 ppm (23 mg/m³, 23,000 mg/m³)
WHO Guidelines (2001)
In controlled human studies involving patients with documented coronary artery disease, mean postexposure COHb
levels of 2.9–5.9% (corresponding to postexercise COHb levels of 2.0–5.2%) have been associated with a significant
shortening in the time to onset of angina, with increased electrocardiographic changes and with impaired left ventricular
function during exercise (Anderson et al., 1973; Kleinman et al., 1989; Allred et al., 1989; Sheps et al., 1987; Adams et
al., 1988). In addition, ventricular arrhythmias may be increased significantly at the higher range of mean postexercise
COHb levels (Sheps et al., 1990; Stern et al., 1988). Epidemiological and clinical data indicate that carbon monoxide
from recent smoking and environmental or occupational exposures may contribute to cardiovascular mortality and the
early course of myocardial infarction (U.S.EPA, 1991). According to one study there has been a 35% excess risk of
death from arteriosclerotic heart disease among smoking and nonsmoking tunnel officers, in whom the long-term mean
COHb levels were generally less than 5% (Stern et al., 1988). Current data from epidemiological studies and
experimental animal studies indicate that common environmental exposures to carbon monoxide do not have
atherogenic effects on humans (U.S.EPA, 1991; Smith and Steichen, 1993).
During pregnancy, endogenous production of carbon monoxide is increased so that maternal COHb levels are usually
about 20% higher than the non-pregnant values. At steady state, fetal COHb levels are up to 10–15% higher than
maternal COHb levels U.S.EPA, 1991; Longo, 1977). There is a well established and probably causal relationship
between maternal smoking and low birth weight at fetal COHb levels of 2–10%. In addition, maternal smoking seems
to be associated with a dose-dependent increase in perinatal deaths and with behavioural effects in infants and young
children (Longo, 1977).
In order to protect nonsmoking, middle-aged and elderly population groups with documented or latent coronary artery
disease from acute ischaemic heart attacks, and to protect the fetuses of nonsmoking pregnant women from untoward
hypoxic effects, a COHb level of 2.5% should not be exceeded. The following guidelines are based on the Coburn-
Foster-Kane exponential equation, which takes into account all the known physiological variables affecting carbon
monoxide uptake (Coburn et al., 1965). The following guideline values (ppm values rounded) and periods of time-
weighted average exposures have been determined in such a way that the COHb level of 2.5% is not exceeded, even
when a normal subject engages in light or moderate exercise:
The INDEX project Final report
67
100 mg/m
3
(90 ppm) for 15 minutes
60 mg/m
3
(50 ppm) for 30 minutes
30 mg/m
3
(25 ppm) for 1 hour
10 mg/m
3
(10 ppm) for 8 hours.
U.S.EPA (1994)
The National Ambient Air Quality Standards (NAAQS) for CO were promulgated by the Environmental Protection
Agency (EPA) in 1971 at levels of 9 ppm (10 mg/m3) for an 8 h average and 35 ppm (40 mg/m3) for a 1 h average, not
to be exceeded more than once per year. (Primary and secondary standards were established at identical levels). The
1970 CO criteria document (National Air Pollution Control Administration, 1970) cited as the standard's scientific basis
a study which indicated that subjects exposed to low levels of CO, resulting in COHb concentrations of 2 to 3% of
saturation exhibited neurobehavioral effects (Beard and Wertheim, 1967). A revised CO criteria document (U.S.
Environmental Protection Agency, 1979) concluded that it was unlikely that significant, and repeatable,
neurobehavioral effects occurred at COHb concentrations below 5%. However, reports that aggravation of angina
pectoris, and other symptoms of myocardial ischaemia, occurred in men with chronic cardiovascular disease, exposed to
low levels of CO resulting in COHb concentrations of about 2.7% (Aronow and Isbell, 1973; Aronow et al., 1972;
Anderson et al., 1973), lead EPA to retain the 8 h 9 ppm primary standard level and to reduce the 1 h primary standard
from 35 to 25 ppm. (EPA also revoked the secondary CO standards because no adverse welfare effects had been
reported at nearambient levels). Later, concerns regarding the validity of data on which the proposed reduction in the 1
h standard was based caused EPA to decide to retain the 1 h 35 ppm standard.
Health Canada (2003)
The choice of National Ambient Air Quality Objectives for carbon monoxide is based upon the clinical significance of
health effects of concern, the number of people requiring protection, and the relationship between COHb levels,
exposure, and ambient carbon monoxide levels.
COHb levels are a biomarker for the toxicity of ambient-level exposures to carbon monoxide and are used as an
indicator of carbon monoxide exposure. Although more research is needed to evaluate the predictive capabilities of the
CFK model in individuals exposed to low concentrations of carbon monoxide and its applicability to sensitive
subpopulation (U.S. EPA, 1991), it is the best model available at present, and it will be used here to estimate
appropriate National Ambient Air Quality Objectives for carbon monoxide. However, it must be remembered that
models provide estimates based upon small numbers of representative measurements.
Table 5.3: Recommended National Ambient Air Quality Objectives, ppm (mg/m
3
)
a
Averaging
Times
Maximum Desirable
Level
Maximum Acceptable
Level
Maximum Tolerable
Level
1 hour 13 (15) 30 (35) n/a
8 hoursb 5 (b) 13 (15) 17.4 (20)
a 1 ppm = 1.146 mg CO/m
3
b rolling average
The maximum desirable levels are based on the carbon monoxide concentration that will result in a
carboxyhaemoglobin (COHb) blood level of less than 1%, or the upper end of the range of baseline COHb levels
resulting from endogenous production. Based on the Coburn-Foster-Kane (CFK) equation, a 1-hour exposure of 13 ppm
or an 8-hour exposure of 5 ppm would lead to less than 1 % COHB.
The recommended maximum acceptable levels for carbon monoxide are 30 ppm averaged over 1-hour and 13 ppm as
an 8-hour rolling average. Results from five recent studies in 3 laboratories were consistent in finding adverse effects of
COHb levels ranging from 2.9% to 6% (as measured by CO-Oximeter) or as low as 2% (as measured by gas
chromatography) on exercise induced angina and on ECG (electrocardiogram) values. CO levels averaging 13 ppm over
8 hours or 30 ppm over 1 hour resulted in COHb levels at or below 2% for adults performing light work (ventilation
rate of 18L/m).
Therefore, the maximum acceptable levels are based on the maintenance of COHb levels of less than 2%, thereby
providing a small margin of safety. At levels above these concentrations, action would be required to decrease the
probability or severity of effects in sensitive populations. Estimating COHb levels using pNEM indicates that less than
1% of the Toronto study area population will experience COHb levels greater than 2.0% if the ambient air quality is less
than or equal to 13 ppm (15 mg/m3) measured over 8 hours.
The INDEX project Final report
68
The recommended maximum tolerable level for an 8 hour exposure could be based on the lowest observed adverse
effect level (LOAEL) of 2.9% COHb observed in the same experiments cited previously. This level would be
approximately 21 ppm. However, the current tolerable level of 17 ppm is sufficiently close to this value to be retained
as the objective.
The recommended maximum tolerable level of 17 ppm averaged over 8 hours will result in a COHb level of about 2.5%
as projected by the CFK model. This is still below the COHb levels believed to result in cardiorespiratory effects in the
general population. Moreover, it is considered to be slightly more protective in accounting for non-standard conditions
and people at the high end of the distribution curve for parameters used in the CFK equation. However, owing to a
diminishing margin of safety within the tolerable range, action is recommended without delay when air quality exceeds
the highest concentration of this range to protect the health of sensitive subgroups.
The averaging times chosen for the maximum desirable, acceptable, and tolerable ranges of carbon monoxide in
ambient air are 1 and 8 hours. The latter averaging time approximates the length of time during which people may be
exposed to carbon monoxide continuously in a particular location (e.g., work, sleep). More importantly, most
individuals approach equilibrium levels of COHb in the blood after about 8–12 hours of exposure to (Anderson et al.,
1973). Owing to the possibility of missing some events (high levels of carbon monoxide exposure) using a continuous
averaging time, rolling averages are recommended for the calculation of the 8-hour averages. The 1-hour averaging
period is intended to be protective for effects that might occur following short exposures to high concentrations of
carbon monoxide.
Effects of long-term exposure
There is not enough reliable information on effects of chronic exposures to low concentrations from either controlled
human studies, ambient population-exposure studies, or from occupational studies. For example, current models cannot
confidently predict whether reduction in pollution will decrease monthly rates of hospital admissions or mortality, even
if they imply a reduction of admissions on days with low pollution. This public-health-related issue cannot be addressed
by daily time series analysis, using only admission or mortality counts. In future studies, investigators also could
consider time-averaged health effects over, say, 1 or 3 months, in relation to pollution exposure metrics for the
corresponding periods (Raub and Benignus, 2002).
Chronic exposures to low CO concentrations may not pose as much a problem as high, acute exposure due to
physiological compensation, tolerance, or adaptation. Smokers show an adaptive response to elevated COHb levels, as
evidenced by increased red blood cell volumes (through increased haemopoiesis) or reduced plasma volumes (Raub and
Benignus, 2002). The major source of total exposure to CO for smokers comes from active tobacco smoking. Baseline
COHb concentrations in smokers average 4% (compared to <2% for non-smokers), with a usual range of 3–8%for one
to two pack-per-day smokers, reflecting absorption of CO from inhaled smoke. COHb levels as high as 15% have been
reported for chain smokers.
The only experimental evidence for short- or long-term compensation to increased COHb levels in the blood from
exogenous sources other than smoking is indirect (Raub and Benignus, 2002). Experimental animal data indicate that
incremental increases in COHb produce physiological responses that tend to offset the deleterious effects of CO
exposure on oxygen delivery to the tissues.
Experimental human data indicate that compensatory cardiovascular responses to submaximal upper and lowerbody
exercise (e.g. increased heart rate, cardiac contractility, cardiac output) occur after CO exposures. These changes were
highly significant for exposures attaining 20% COHb. Other compensatory responses are increased coronary blood
flow, cerebral blood flow, Hb, and oxygen consumption in muscle.
Cardiovascular effects
Kristensen (1989) examined the relationship between cardiovascular diseases and chronic occupational exposures, and
concluded that CO exposure increases the acute risk of cardiovascular disease, at least transiently. Stern et al. (1988)
investigated the effects of occupational carbon monoxide exposures on deaths from arteriosclerotic heart disease among
5529 New York City bridge and tunnel officers in the period 1952–1981. Among the more exposed tunnel officers there
was a 35% excess risk compared with the New York City population, whereas among the less exposed bridge officers
the risk was not elevated. The elevated risk among the tunnel officers declined significantly within five years after
cessation of the occupational exposure, and there has also been a significant decline since 1970, when the introduction
of new ventilation systems lowered the carbon monoxide levels in tunnels and tunnel booths. The 24-hour average
carbon monoxide concentrations inside the tunnels were around 57 mg/m
3
(50 ppm) in 1961 and 46 mg/m
3
(40 ppm) in
1968. During rush hour traffic in 1968, carbon monoxide concentrations in tunnel toll booths were as high as 74–189
mg/m
3
(65–165 ppm) and in 1970 the mean concentration over 38 days was 72 mg/m
3
(63 ppm). However, the mean
The INDEX project Final report
69
COHb levels measured among smoking and nonsmoking tunnel officers in 1970 and 1981 were generally lower than 5
%.
Hansen (1989) reported the results of a 10-year follow-up study on mortality among 583 Danish male automobile
mechanics between 15 and 74 years of age. The number of deaths expected for the automobile mechanics was
compared with those for a similar group of Danish men employed as carpenters, electricians and other skilled workers
free from occupational exposure to automobile exhaust, petrochemical products, asbestos and paint pigments. The
number of deaths observed among the automobile mechanics exceeded the expected number by 21%. Although the
increased mortality was not confined to any single cause of death, the author reported a remarkable excess of deaths
attributed to ischaemic heart disease where the standardized mortality ratio was 121 and the 95% confidence interval
was 102–145. The only other significant category of death was that due to external causes (SMR = 131; 95% CI = 113–
153). No significant differences were found among the automobile mechanics for other diseases except for an increase
in pancreatic cancer (SMR = 219; 95% CI = 128–351). Exposure to carbon monoxide and polycyclic aromatic
hydrocarbons through the inhalation of automobile exhaust and the handling of solvents and oils may have accounted
for the difference in ischaemic heart disease deaths between the automobile mechanics and the comparison group;
however, other occupational exposures or other lifestyle factors, as indicated above, may also have contributed to the
findings.
The haemodynamic responses to CO have been reviewed by Penney (1988). Chronic CO exposures, at levels sufficient
to raise COHb concentrations to greater than 10% can produce increased numbers of red blood cells (polycythemia),
increased blood volume, and increased heart size (cardiomegaly). In addition, heart rate, stroke volume, and systolic
blood pressure may be increased. Some of these effects have been seen in smokers. Penney and Howley (1991) report
that CO can enhance atherosclerosis in individuals with elevated serum cholesterol.
Current data from epidemiological studies and laboratory animal studies do not suggest that common environmental
exposures to carbon monoxide have atherogenic effects on humans (U.S.EPA, 1991; Smith and Steichen, 1993).
Effects on pregnancy outcomes
A case-control study of the association between low birthweight infants and maternal CO exposures in approximately
1000 cases in Denver (Alderman et al., 1987) failed to detect a relationship between CO exposure (estimated form
fixed-site outdoor monitoring data) during the last 3 months of pregnancy and lower birth weights. Mean CO levels
ranged from 0.5 to 3.6 ppm at 8 monitoring locations in metropolitan Denver. The 5th and 95th percentile
concentrations at the site with the highest (3.6 ppm) mean were 1.6 and 4.8 ppm, respectively. The odds ratio at the
highest concentration site was 1.1 and the 95% confidence interval was 0.8-1.6). This study did not directly account for
unmeasured sources of CO exposure, such as smoking, emissions from gas appliances and exposures to vehicular
exhaust, which are limitations of the study design. A more extensive study of a cohort of 125,573 children born to
women living in the Los Angeles area (1989-1993) found that exposure to ambient concentrations > 5.5 ppm (3 mo
average) during the last trimester of pregnancy was associated with a significantly increased risk of low birthweight
(odds ratio = 1.22; confidence interval =1.03-1.44) after adjustment for potential confounders (Ritz and Yu, 1999).
Fetotoxicity has been demonstrated in laboratory animal studies. Altered brain neurochemical development and growth
retardation have been demonstrated in rats exposed to CO in utero (Storm and Fechter, 1985; Leichter, 1993).
Effects on lung function
Individuals exposed to relatively high concentrations of CO in both indoor and outdoor environments may have lung
function decreases. In most cases, however, causality is difficult to establish because, in addition to CO, these
individuals were also exposed to high concentrations of other combustion products, many of which are respiratory
system irritants.
In the study of tunnel and bridge officers, described earlier, lung functions, forced vital capacity (FVC) and forced
expiration volume in 1 s (FEV1.0), were slightly reduced in tunnel vs. bridge officers (Evans et al., 1988). Exposures of
adults to typical ambient concentrations of CO, both outdoors and indoors, have not been significantly associated with
lung function decrements (Lebowitz et al., 1987).
Pollutants related to automotive traffic, especially CO and nitrogen oxides, were associated with the prevalence of
asthma in middleschool Taiwanese students (Guo et al., 1999; Lin et al., 1999). Physician consultations in London for
lower respiratory diseases were significantly correlated with NO2 and CO levels in children, but not in adults (Hajat et
al., 1999). Exposure of children with mild asthma to environmental tobacco smoke resulted in pulmonary function
decrements, i.e. reduced FEV1.0 (Magnusen et al., 1993).
The INDEX project Final report
70
Carcinogenic and genotoxic effects
No studies documenting carcinogenic or genotoxic effects of CO in humans were located in the available literature.
Summary of Carbon Monoxide Dose Response Assessment
________________________________________________________________________________________________
Exposure other than inhalation: Endogenous production of CO due to the catabolism of haemoproteins and
membrane lipids results in 0.4-0.7% COHb (healthy subjects), increased to 0.7-2.5% during pregnancy. Increased
endogenous production of CO are associated with a series of haematological diseases (e.g.haemolytica anemia) and to
the endogenous oxidation of specific pollutants (e.g. halomethanes).
Toxicokinetics: ~80-90% of absorbed dose binds with haemoglobin (COHb; causing a reduction in the oxygen-
carrying capacity of the blood). CO affinity for haemoglobin 200-250 times that for O
2
. CO is eliminated unchanged via
the lungs, with t
½
(COHb) of 2-6.5 hours (while breathing clean air), depending on the initial COHb level. t
½
(COHb)
much higher in the fetus than in the pregnant mother.
Health effect levels of short- and long-term exposure
NOAEL
% COHb
LOAEL
% COHb
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
SHORT-TERM EXPOSURE
1.1-1.3 2 aggravation of angina and other cardiovascular
diseases
Subjects with angina
pectoris, 1h
Aronow, 1981
OEHHA 1999
REL: 23
2.5
- acute ischaemic heart attacks (in documented
or latent coronary artery disease)
- untoward hypoxic effects in fetuses of
nonsmoking pregnants
- Subjects with
ischaemic heart
disease
- Fetuses
WHO 2000
100 (15m-GV)
60 (30m-GV)
30 (1h-GV)
10 (8h-GV)
2.7 aggravation of angina pectoris, and other
symptoms of myocardial ischaemia
Subjects with angina
pectoris
Aronow and Isbell,
1973; Aronow et al.,
1972; Anderson et al.,
1973
U.S.EPA 1994
10.3 (8h)
35 (1h)
2.9 cardiorespiratory effects in individuals with
ischaemic heart disease.
Subjects with
ischaemic heart
disease
Kleinman et al., 1998;
Allred et al.,1989;
Sheps et al., 1987;
Kleinman et al., 1989;
Adams et al., 1988
Health Canada
2003
MDL(1h/8h)=15/6
MAL(1h/8h)=35/1
5
MTC(8h)=20
LONG-TERM EXPOSURE (not found in available literature)
Carcinogenicity: No evidence
Genotoxicity: No evidence
Odour threshold: practically odorless
Susceptible population: Human characteristics increasing risk:
- Individuals with Ischaemic heart disease or coronary artery disease
- Individuals having reduced blood haemoglobin concentrations (e.g.haemolytic anemia) or with abnormal
haemoglobin
- Individuals with chronic bronchitis, emphysema and chronic obstructive pulmonary disease (COPD), children with
other lung inflammatory problems (e.g.cystic fibrosis)
- Pregnants and fetuses
- Children: physiologically, children have larger metabolic demands and consequently greater oxygen uptake demands
than do adults, on a per unit mass basis.
________________________________________________________________________________________________
The INDEX project Final report
71
6. Risk Characterization
Health hazard evaluation
Carbon monoxide (CO) is a tasteless, non-irritating, odorless and colorless toxic gas which can cause acute and lethal
poisonings with very few and late occurring warning signs. Until very severe symptoms occur (inability to walk) none
or only nonspecific symptoms are noted in healthy humans. As a consequence, a large number of deaths occur annually
in fires, workplaces and in indoor environments. In the latter instance, poisonings occur because of the incorrect use of
combustion devices or due to faulty unvented gas appliances. The associated risk, the evaluation of its impact on the
European population as well as the related risk management strategies will be outlined in the following chapter (Risk
Management).
The present risk characterization adresses CO exposures experienced by people during their normal daily activities,
while changing from one microenvironment to another, the significance of related health effects, as a function of
individual susceptibility, and the relationship between carboxyhaemoglobin levels (COHb; the biomarker for CO-
exposure and -toxicity), exposure, and indoor carbon monoxide levels.
Currently available evidence suggests that individuals with heart disease (including stable exercise-induced angina,
coronary artery disease, and ischaemic heart disease) represent the population at greatest risk from exposure to indoor
carbon monoxide levels. In addition, population groups with either increased probability or increased severity of health
effects include fetuses, pregnant women, and young infants, individuals with anemia or respiratory disease, the elderly,
children, and persons with peripheral vascular disease and chronic obstructive lung disease.
Results from five clinical studies in 3 laboratories were consistent in finding adverse effects of COHb levels as low as
2.9% (as measured by CO-Oximeter) on exercise induced angina and on electrocardiogram values.
Providing a small margin of safety, a COHb level of 2 % will be here considered as the starting point of the present risk
characterization, and CO exposures considered acceptable
based on the maintenance of COHb levels of less than 2%.
These levels are considered protective for the subpopulation sensitive to cardiovascular effects and are expected to
provide an adequate margin of safety for the other susceptible subpopulations described above and for the general
population with respect to short- and long-term toxicity effects. Additionally, CO exposures are here considered
desirable
when they result in COHb levels of less than 1%, representing the upper end of the range of baseline COHb
levels resulting from endogenous production in normal, non-smoking individuals. This level was chosen taking into
consideration uncertainties in the available data and using the most conservative assumptions.
The CO concentrations in inhaled air associated with these threshold COHb levels (see Table 6.1) were estimated using
the Coburn-Foster-Kane (CFK) model, considering adults performing light work at a ventilation rate of 18L/min. The
CFK model still presents the best model available, although more research is needed to evaluate the predictive
capabilities in individuals exposed to low CO concentrations and its applicability to sensitive subpopulation. The
averaging times chosen for deriving the CO air concentrations are 1 and 8 hours, the latter approximating the length of
time required by most individuals to approach equilibrium levels of COHb in the blood (about 8–12 hours of exposure),
or the time during which people may be continuously exposed to carbon monoxide in a particular location. The 1-hour
averaging period is intended to evidence short-term exposures related to personal activities generating increased CO
levels.
Table 6.1: Carbon monoxide average concentrations in inhaled air derived (CFK-model) from
selected [COHb], considering 1-h and 8-h exposure duration
COHb levels considered [COHb] % [CO] in air mg/m
3
1 h–avg. 8h–avg.
acceptable 2 35 15
desirable 1 15 6
The INDEX project Final report
72
Percentage of population exposed beyond given threshold CO levels
Table 6.1
acceptable desirable
Available exposure studies 1-h averaging period
Description (Study, Year) N 35 mg/m
3
15 mg/m
3
1-h personal exposure (indoors)
Athens (Expolis, 96-98) 44 10 % 30 %
Basel (Expolis, 96-98) 50 < < 10 %
Helsinki (Expolis, 96-98) 195 < 5 %
Milan (Expolis, 96-98) 45 < < 10 %
Oxford (Expolis, 98-00) 26 < 5 %
Prague (Expolis, 96-98) 23 < 10 %
8-h averaging period
15 mg/m
3
6 mg/m
3
48-h personal through the day
exposure
Athens (Expolis, 96-98) 44 < <
Basel (Expolis, 96-98) 50 < <
Helsinki (Expolis, 96-98) 195 < <
Milan (Expolis, 96-98) 45 < <
Prague (Expolis, 96-98) 26 < <
2-week residential exposure
(kitchen and bedroom)
England (BRE, 97-99) 830 < <
< out of the evaluation range (i.e. <5% of the environments
investigated)
The studies
Expolis study: Within the multi-centre European EXPOLIS study, personal exposure to CO, measured every minute for 48 h, of 401
randomly selected study participants (25–55 yr old; living and working within the metropolitan areas; mainly non-smokers) was
monitored in Athens, Basel, Helsinki, Milan and Prague. 1-h average personal exposures were computed in order to assess the
influence of specifc sources. Each participant also completed a time-microenvironment-activity diary and an extended questionnaire.
In addition, for the same time period, ambient levels of CO from fixed site stations were collected. All measurements were performed
between October 1996 and June 1998.
In Georgoulis et al. (2002) both the 48-h and 15-min averaged personal exposures were calculated and compared, the latter
corresponding to specific 15-min time periods when different activities (i.e. ‘‘under smoke (ETS) exposure’’, ‘‘during use of gas
appliances’’) were taking place.
Selection bias (Oglesby et al., 2000): In Basel, participants of direct monitoring as compared to non-participants were more likely to
live at streets with low traffic volume; although in Helsinki, traffic volume was neither significantly related to participation in direct
nor indirect monitoring, the point estimates indicate a tendency to decreased participation with increasing traffic intensity at home.)
BRE, UK: The Building Research Establishment (BRE) has conducted a national survey of air pollutants in 876 homes in England,
to increase knowledge of pollutant levels and the factors associated with high concentrations. CO levels were measured, once in each
home, using Dräger colorimetric diffusion tubes (passive samplers), as two-week time-weighted averages in the kitchen and main
bedroom of each home. All measurements were performed between October 1997 to February 1999.
Comments
Available measurements from the Expolis study are twofold:
- the 48-h data are ‘through-the-day’ personal average exposures experienced by the working population in European
metropolitan areas and, hence, include the contribution of outdoor exposure (commuting). A typical pattern of CO
levels as measured (1-min resolution) during a 48-h monitoring cycle is shown in Figure 6.1. As examplified by the
Figure, short-term exposures to higher CO levels only marginally affect a 48-h average outcome. The 48-h data are
used in the present risk characterization assuming that even 8-h extrapolations from the overall dataset would not be
affected by short-term peak exposures. As outlined in the study conclusions (Georgoulis et al., 2002), the 48-h
personal CO exposures were highly and consistently associated with urban ambient air concentrations, consistently
but less signifcantly associated with exposure to ETS and inconsistently and mostly insignifcantly associated with
exposure to the use of gas appliances. Time spent in commuting had an insignifcant effect on the 48-h average CO
exposure.
The INDEX project Final report
73
- the 1-h average personal exposure measurements were exclusively taken indoors. As concluded by the study
members, the presence of smoking or gas cooking in residences signifcantly increased the short-term CO exposures;
the difference with the 48-h averages were explained by the relatively short duration of many activities. The exposure
data are here considered indicative for common indoor-activity related short-term exposures experienced by the
general population living in (sub-)urban European areas.
Significant differences in both personal exposure and ambient levels (Georgoulis et al., 2002) were found within the
five cities, ranging from high values in Milan and Athens to low in Helsinki.
It is noteworthy that, for CO, personal and not microenvironmental samples were drawn in the Expolis study,
considering that people experience various CO exposures due to their daily activities and to unhomogeneous CO levels
within indoor envornments, and that seasonal effects on CO levels were taken into account, supposed to be largely due
to indoor sources.
The BRE study concluded (530 homes; 2-week averaged microenvironmental samples in kitchens and bedrooms) that
CO levels were very low for most of the time in the majority of homes in England. Identified factors influencing long-
term CO levels were: gas cooking, unflued heaters, smoking and outdoor air (in urban locations). Seasonal effects were
largely associated to indoor sources and extract fans were found to have little effect on CO levels in the kitchen.
Figure 6.1: Typical result of a 48-h monitoring cycle (1-min resoltution) obtained during the Expolis study
Doubts could be expressed on whether the selected exposure data could be considered as descriptive of the nowadays
situation, when considering the recent evolution of ambient CO levels. Current ambient air quality regulations, i.e. those
contributing to the reduction of carbon dioxide emissions (the Kyoto Protocol), the EU Limit Value for CO in ambient air
and equivalent national regulations in the EU, have shown to be effective with respect to a reduction of CO ambient levels,
as examplified in Figure 6.2 for a german metropolitan area, the trend expected to continue in the years to come.
The INDEX project Final report
74
Figure 6.2: CO levels in urban areas - trend in the Ruhr-Rhine region
EU limit value for carbon monoxide in ambient air – Directive 2000/69/EC
Limit value for the protection of human health:
Averaging period: Maximum daily 8-hour mean
Limit value: 10 mg/m
3
Margin of tolerance: 6 mg/m
3
on 13 December, reducing on 1 January 2003 and every 12 months thereafter by 2 mg/m
3
to reach 0 % by 1 January
2005
Date by which limit value is to be met: 1 January 2005
The maximum daily 8-hour mean concentration will be selected by examining 8-hour running averages, calculated from hourly data and updated each
hour. Each 8-hour average so calculated will be assigned to the day on which it ends. i.e. the first calculation period for any one day will be the period
from 17:00 on the previous day to 01:00 on that day; the last calculation period for any one day will be the period from 16:00 to 24:00 on that day.
Result
Available exposure data confim that Carbon Monoxide (CO) sources in EU-residences are contributing to short-term
rather than to long-term exposures. Personal exposure outcomes averaged over 1-hour were considered of moderate
concern even for the most susceptible subpopulations. Nevertheless, uncertainties resulting from the predictive
capabilities of the CFK-model in individuals exposed to low CO concentrations and its applicability to sensitive
subpopulations, suggest that about 10% of the general non-smoking population experience CO levels which could be
hazardous for individuals with heart diseases. Increased exposures could be expected for residences in the vicinity of
busy city streets.
In addidion, there is no evidence that long-term CO exposures in EU residences contribute to carboxyhaemoglobin
levels in blood higher than the baseline levels resulting from endogenous production in normal, non-smoking
individuals.
On the other hand and in contrast with all other chemicals assessed in the present report, carbon monoxide causes a
considerable number of deaths and acute poisonings in the general population (with complications and late sequelae).
Also, individuals suffering from CO poisoning are often unaware of their exposure because symptoms are similar to
those associated with viral illness or clinical depression. In indoor environments, these health risks are nearly
completely associated with the incorrect use of combustion devices or faulty unvented gas appliances.
The INDEX project Final report
75
Nitrogen dioxide
Synonyms: -
CAS Registry Number: 10102-44-0
Molecular Formula: NO
2
1. Compound identification
Nitrogen dioxide is a reddish-brown gas in colour, and a strong oxidant with a characteristic pungent odour. It is soluble
in water. A low vapour pressure of 900 mm Hg at 25
o
C indicates nitrogen dioxide will exist as a gas in the ambient
conditions. It is corrosive and highly oxidising gas. Typically less than 10% by volume of the total emissions of NOx
from combustion sources is in the form of NO
2
(WHO 1997). NO is rapidly oxidised to NO
2
by available oxidants,
particularly ozone, in ambient conditions. In indoor air, it is generally a much slower process. Photolysis of NO
2
by
sunlight is the most important source of ozone (Alm 1999).
Nitrogen dioxide is also an important atmospheric gas, in addition to health effects, also because it absorbs visible solar
radiation and, therefore contributes to atmospheric visibility, it could have a role in global climate change, it is with
nitric oxide (NO), a main regulator of the oxidising capacity of the free troposphere and ozone (O
3
) concentrations in
the troposphere (WHO 2000).
There is no clear evidence for a concentration-response relationship for NO
2
. Asthmatics and persons with chronic
obstructive pulmonary disease (COPD) are the most susceptible populations to acute changes in lung function, airway
responsiveness and other respiratory symptoms. (WHO 2000).
2. Physical and Chemical properties
Molecular weight (g/mol) 46.01
Melting point (°C) -11.2
Boiling point (°C) 21.2
Density (at 20 °C, 1 atm) 1.448
Relative density (air =1) 1.59
Solubility Soluble in sulphuric and nitric acid
Conversion factors at 20 °C and 760 mm Hg:
1 ppb = 1.912 µg/m
3
1 µg/m
3
= 0.523 ppb
Sources: Alm (1999), Verschueren 2001, HSDB (2003)
3. Indoor Air Exposure assessment
Emission sources
The most important indoor sources of NO
2
include gas appliances such as stoves, ovens, space and water heaters, and
unflued kerosene heaters. Especially, gas stoves with pilot light have been found strong indoor sources of NO
2
.
Typically, NO
2
concentrations in homes with gas stoves are 2-5 times higher than in homes with electric stoves. Indoor
concentrations of NO
2
are typically higher in winter than in summer, probably due to increased use of gas heating,
lower ventilation rates, and higher outdoor concentrations (Alm 1999).
The INDEX project Final report
76
The main ambient sources of nitrogen oxides emissions include intrusion of stratospheric nitrogen oxides, bacterial and
volcanic action, and lightning. Fossil fuel power stations, motor vehicles and domestic combustion appliances emit
nitric oxide (NO), which is a reactive compound forming rapidly NO
2
. Other contributions of nitrogen dioxide to the
atmosphere come from specific non-combustion industrial processes, such as the manufacture of nitric acid, the use of
explosives and welding. Ambient concentrations of NO
2
in urban areas peak during the morning and evening rush hours
due to increased emissions of NO from motor vehicles (COMEAP 1997, WHO 1997, WHO 2000).
Indoor air and exposure concentrations
Indoor concentrations vary widely depending on the presence of special indoor sources of NO
2
. Concentrations in
homes without NO
2
sources are typically lower than outdoor concentrations and in those cases indoor levels are driven
by outdoor sources. In the recently published ECRHS II study, carried out in 21 European cities annual ambient NO
2
concentrations ranged from 4.9 µg/m
3
in Reykjavik, Iceland to 72 µg/m
3
in Turin, Italy (Hazenkamph-von Arx et al
2004). The average winter/summer ratio showed considerably higher concentrations in winter than in summer, being
1.5. According to WHO (2000) annual mean ambient concentrations in urban areas throughout the world typically
range from 20 to 90 µg/m
3
. Typical indoor air concentrations in homes with gas cooking vary between 25 and 200
µg/m
3
over a period of several days. Maximum indoor 1-hour peaks may reach up to 2000 (COMEAP 1997, WHO
2000).
In Helsinki and Prague average indoor concentrations were lower than the respective outdoor concentrations that ranged
from 24 to 61 µg/m
3
(Kousa et al 2001). Mean indoor concentrations in Europe ranged from 13 µg/m
3
in UK to 43
µg/m
3
in Prague according to the results by Kousa et al (2001) and COMEAP (1997). Average personal exposures were
equal or slightly higher.
In an Italian population based study, the highest weekly indoor NO
2
concentrations were measured in a rural area of Po
Delta (Simoni et al 2002). Weekly mean indoor concentration in kitchen in winter was higher than the respective
concentration in summer, being 62 µg/m
3
and 38 µg/m
3
. The presence of gas-furnace heating was identified as the
determining factor for the elevated NO
2
concentrations.
In a Spanish study of 340 dwellings between 1996 and 1999 (Garcia-Algar et al 2003), average annual indoor
concentrations of NO
2
did not vary significantly, ranging only from 12.5 to 14.7 µg/m
3
. Respective outdoor air
concentrations were slightly higher in 1996 and 1998 and slightly lower in 1997 and 1999, thus typical indoor-outdoor
ratios were close to 1. The principal indoor sources of NO
2
in Spanish homes were the use of gas cooker, the absence of
an extractor fan when cooking, the absence of central heating and cigarette smoking.
Levy et al (1998) studied NO
2
concentrations in homes in 18 cities in 15 countries reporting 2-day means ranging from
10 µg/m
3
to 81 µg/m
3
and personal exposures from 21 µg/m
3
to 97 µg/m
3
. Use of a gas stove was found the dominant
activity influencing indoor concentrations with an increase in indoor-outdoor ratios from 0.7 to 1.2 for participants
using gas stoves. Results showed also the importance of combustion space heaters to elevated NO
2
concentrations. On
the contrary, wood-burning appliances were not related to elevated NO
2
concentrations in a Canadian study carried out
in 49 houses (Levesque et al 2001).
Brown et al (2002) studied emissions of unflued gas heaters in room chamber (32.4 m
3
) tests. NO
2
concentrations in the
chamber ranged from 180 µg/m
3
to 530 µg/m
3
due to emissions from the gas heaters. Lee and Wang (2004) studied
nitrogen dioxide emissions from incense burning also in the chamber (18 m
3
) tests. They found that maximum nitrogen
dioxide concentrations peaked up to 91 µg/m
3
during incense burning.
Other studies showed indoor concentrations of NO
2
in homes without gas stoves ranging from 13 µg/m
3
to 40 µg/m
3
and with gas stoves from 25 µg/m
3
to 70 µg/m
3
in UK (COMEAP 1997). Determinants of NO
2
exposures were studied
with preschool children in Finland. Preschool located in down town, gas stoves and parent smoking at home were found
the most important factors of the increased NO
2
exposures for those children (Alm 1999).
Emission sources and microenvironments where high concentrations of NO
2
occur are reviewed in Table 3.1. Elevated
concentrations were typically related to gas cooking, gas heating, and incense burning. High NO
2
concentrations were
also found in some public buildings. The highest maximum values up to 7530 µg/m
3
were measured in indoor ice
arenas (Pennanen et al 1997). Also in kitchens with gas appliances high concentrations, 3808 µg/m
3
, were determined
(Lebret et al 1987).
Residential indoor air concentrations of NO
2
in European urban populations are presented in Figure 3.1 and Figure 3.2.
Respective exposure concentrations are presented in Figure 3.3.
The INDEX project Final report
77
0
20
40
60
80
100
0 20 40 60 80 100 120 140
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Hel
Bas
Oxf
Pra
PoD
Figure 3.1. Cumulative frequency distributions of indoor air concentrations of nitrogen dioxide in Helsinki (Hel, n=
175), Basel (Bas, n= 50), Prague (Pra, n= 33) (Kousa et al 2001), Oxford (Oxf, n=40) (Hanninen et al 2004, EXPOLIS
2002) and in Po Delta (PoD) (n = 112, AM = 62 µg/m
3
, std = 32 µg/m
3
) region in Northern Italy (Simoni et al 2002).
0
20
40
60
80
100
0 102030405060708090100
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
NO2 14-day
Figure 3.2. Cumulative frequency distribution of 14-day indoor air concentrations of nitrogen dioxide in the kitchens in
England (GM= 21.8 µg/m
3
, max 620 µg/m
3
, n=845, Coward et al 2002).
The INDEX project Final report
78
0
20
40
60
80
100
0 20406080100120140
Exposure (µg/m
3
)
Cumulative frequency (%)
Hel
Bas
Oxf
Pra
Figure 3.3. Cumulative frequency distributions 48-hour personal exposure concentrations of nitrogen dioxide in
Helsinki (Hel, n= 177), Basel (Bas, n= 50), Prague (Pra, n= 35) (Kousa et al 2001) and Oxford (Oxf, n=42) (Hanninen
et al 2004, EXPOLIS 2002).
Table 3.1. Short time NO
2
concentrations related to specific microenvironments or emission sources.
Environment/
emission source
Location Averaging time Reference
A
M GM median max
Indoor ice arenas Finland max 15-min 320 - 7530 Pennanen et al 1997
max 1-hour 270 - 7440
Gas appliances The Netherlands max 1-min kitchen 400 - 3808 Lebret et al 1987
max 1-hour kitchen 230 - 2055
max 24-hour kitchen 53 - 478
Gas stoves The Netherlands kitchen 2500 Noy et al 1990
Indoor ice arenas Finland 1-week 2 - 1838 Pennanen et al 1998
ice resurfacers propane 396
gasoline 283
elecrtic 25
Homes with gas appliances several hours 200 WHO 2000
1-hour 2000
Gas cooking UK 1-week gas stove 28 - 107 Ross 1996
max 1-hour 342 - 1585
1-week electric stove 23 - 26
max 1-hour 38 - 55
Unflued gas heaters room chamber tests 180 - 530 Brown et al 2002
Homes Sweden 24-hour urban dwellings 6.7 11 Sakai et al 2004
Japan 24-hour urban dwellings 98 369
Indoor ice ring Hong Kong, China 15-min gasoline-fueled 58 - 91 Guo et al 2004
15-min propane-fueled 242
Homes 15 countries 2-day indoor 10 - 81 Levy et al 1998
exposure 21 - 97
Incense burning Chamber tests during burning 17-91 Lee and Wang 2004
Exposure, schoolchildren Sweden 24-hour urban 13 Berglund et al 1994
rural 7
AM = arithmetic mean, GM = geometric mean, max = maximum value
Concentration (µg/m
3
)
The INDEX project Final report
79
4. Toxicokinetics
Absorption
Inhaled nitrogen dioxide (NO
2
) reaches easily the lower respiratory tract (bronchioli and alveoli of the lungs) and
dissolves in lung fluids, hydrating slowly to nitrous (HNO
2
) or nitric acid (HNO
3
). These acids dissociate into nitrite
(NO
2
-
) or nitrate (NO
3
-
) ions, which are rapidly taken up in the lung epithelium (Goldstein ate al., 1977; Postlethwait
and Bidani 1989). NO
2
acts as a strong oxidant and likely reacts with unsaturated lipids and functional proteins in cells
either within the lung epithelial lining fluid or in the epithelial cell membrane. This may result in loss of cell
permeability and control. Furthermore, the substance activates peroxide detoxification pathways (WHO97).
In Bauer et al. (1986) fifteen asthmatic subjects were exposed to 0.6 mg/m
3
NO
2
via mouthpiece for 20 minutes at rest,
followed by 10 minutes of exercise. Expired NO
2
concentrations were measured continuously. NO
2
deposition was
72±2% at rest, increasing to 87±1% with moderate exercise. These findings indicate that the NO
2
dose to the distal
airways and alveolar space, and therefore toxic effects in this region, would be substantially increased by exercise.
Mathematical modelling studies show that the deposition of nitrogen dioxide in the tissues of the lower respiratory tract
is predicted to be maximal at about the junction of the conducting airways and the gas exchange region of the lungs in
humans, rats, guinea pigs and rabbits. Although the actual tissue dose at a similar starting tracheal concentration differs
across species, the shape of the deposition curves is roughly equivalent. The region predicted to receive the maximal
dose is that where the typical nitrogen-dioxide-induced morphometric lesion is observed in several species of animals.
Using this mathematical model, it also can be predicted that, as tidal volume increases in humans (e.g. in exercise), the
dose to the tissue of the gas exchange region increases substantially more than the dose to the conducting airways
(Miller et al., 1982; Overton, 1984; Overton et al., 1987).
Distribution
After passing the lungs, nitrogen dioxide products are distributed to all parts of the body via the bloodstream. Goldstein
et al. (Goldstein ate al., 1977) exposed two female rhesus monkeys to radiolabelled nitrogen dioxide (
13
NO
2
) for 6 to 9
minutes through a facemask. The investigators found the radioactive tracer back in the circulation. They also reported
that the time-concentration relationship of
13
N in arterial blood correlated well with the time-concentration relationship
in the lungs. In addition, the investigators showed that the gas was distributed throughout the lungs.
Elimination
No human data have been found. Saul and Archer (1983) observed that nitrogen dioxide or its products (nitrite, nitrate)
were excreted as nitrate in the urine of Sprague-Dawley rats. Also, they showed that the amount of urinary nitrate
correlated linearly to nitrogen dioxide exposure levels. The urinary values returned to normal after four days.
5. Health effects
Nitrogen dioxide is thought to damage lungs in three ways: (1) it is converted to nitric and nitrous acids in the distal
airways, which directly damages certain structural and functional lung cells; (2) it initiates free radical generation,
which results in protein oxidation, lipid peroxidation, and cell membrane damage; and (3) it reduces resistance to
infection by altering macrophage and immune function.
Effects of short-term exposure
Numerous controlled clinical studies have examined lung function, airway responsiveness to pharmacological, physical
(e.g. cold air) or natural (i.e. allergens) bronchoconstrictors and the defence against viral and bacterial airway infections
in human subjects exposed to nitrogen dioxide.
Generally, concentrations in excess of 1880 µg/m
3
(1 ppm) are necessary during acute controlled exposures to induce
changes in pulmonary function in healthy adults (U.S.EPA, 1993; Berglund et al., 1993; Wagner, 1985). Because these
concentrations rarely occur in indoor air, concern about the effects of nitrogen dioxide has been focused on people with
The INDEX project Final report
80
pre-existing lung disease. There have been numerous studies of people with asthma, chronic obstructive pulmonary
disease, or chronic bronchitis showing that exposure to low levels of nitrogen dioxide can cause small decrements in
forced vital capacity and forced expiratory volume in 1 second (FEV
1
) or increases in airway resistance.
The lowest level of nitrogen dioxide exposure reported in more than one laboratory to show a direct effect on
pulmonary function in asthmatics was a 30-minute exposure, with intermittent exercise, to 560 µg/m
3
(0.3 ppm) (Roger
at al., 1990; Bauer et al., 1986). Although similar but statistically non significant trends have been observed in other
controlled human studies performed at lower concentrations (Bylin et al., 1985; Hazucha et al., 1983; Orehek et al.,
1976), the small size of the decrements and questions regarding the statistical significance of some of these results
together suggest that caution should be exercised in accepting these findings as demonstrating acute effects.
Orehek and colleagues (Orehek et al., 1976) were the first to report that relatively brief exposures of asthmatics to low-
level NO
2
(0.19 mg/m
3
) might enhance subsequent responsiveness to challenge with a broncho-constricting drug.
Although NO
2
alone caused an increase in airway resistance in only 3 of 20 asthmatics, bronchial responsiveness to
carbachol increased in 13 of these 20 subjects. However, this report was challenged because of the retrospective
separation of responding from non-responding subjects.
Hazucha and colleagues (Hazucha et al., 1983) failed to confirm these results in a study of 15 asthmatic subjects.
Although there were some differences in techniques and patient selection between the Orehek and Hazucha studies, it
seems likely that the findings of Orehek and coworkers reflect a retrospective stratification of subjects into “responder”
and “non-responder” groups that was not justified a priori.
Other investigators have also been unable to confirm effects of 0.19–0.38 mg/m
3
(0.1-0.2 ppm) NO
2
on lung function in
either asthmatic adolescents (Koenig et al., 1985; Koenig et al., 1988) or in mildly asthmatic adults (Koenig et al., 1985;
Bauer et al., 1986; Orehek et al., 1976; Bylin et al., 1985; Hazucha et al., 1983; Koenig et al., 1988; Kleinman et al.,
1983; Linn et al., 1986; Mohsenin & Gee, 1987; Morrow & Utell, 1989; Roger et al., 1990).
Kleinman and colleagues (Kleinman et al., 1983) evaluated the response of lightly exercising asthmatic subjects to
inhalation of 0.38 mg/m
3
NO
2
for 2 hours, during which resting minute ventilation was doubled. Although NO
2
did not
cause alterations in flow rates or airways resistance, approximately two-thirds of the subjects experienced increased
responsiveness to methacholine after inhalation of NO
2
compared with clean air, as assessed by specific airway
resistance.
In view of the inconclusive findings at 0.19 and 0.38 mg/m
3
NO
2
, Bauer and colleagues (Bauer et al., 1986) studied the
effects of mouthpiece exposure to 0.56 mg/m
3
NO
2
for 30 minutes (20 minutes at rest followed by 10 minutes of
exercise at approximately 40 L/min) in 15 asthmatics. At this level, NO
2
inhalation produced significant decrements in
forced expiratory flow rates after exercise, but not at rest. Furthermore, after airway function was allowed to return to
baseline during a 1-hour recovery period, isocapneic cold-air hyperventilation elicited increased airway responsiveness
in the asthmatics who had earlier been exposed to NO
2
.
Roger and coworkers (Roger et al., 1990), in a comprehensive, concentration response experiment, were unable to
confirm the results of a previous pilot study suggesting airway responses in asthmatic subjects. Twenty-one male
asthmatics exposed to NO
2
at 0.28, 0.38, and 1.13 mg/m
3
for 75 minutes did not experience significant effects on lung
function or airway responsiveness compared with air exposure. Bylin and coworkers (Bylin et al., 1985) found
significantly increased bronchial responsiveness to histamine challenge compared with sham exposure in 8 atopic
asthmatics exposed to 0.56 mg/m
3
NO
2
for 20 minutes. Five of 8 asthmatics demonstrated increased reactivity, while 3
subjects showed no change, as assessed by specific airway resistance. Mohsenin (Mohsenin, 1987) reported enhanced
responsiveness to methacholine in eight asthmatic subjects exposed to 0.94 mg/m
3
NO
2
at rest for 1 hour; airway
responsiveness was measured by partial expiratory flow rates at 40% vital capacity, which may have increased the
sensitivity for detecting small changes in airway responsiveness.
Strand et al. (Strand et al., 1996) found increased responsiveness to histamine among 19 asthmatic subjects 5 hours after
a 30 minute exposure to 0.49 mg/m
3
NO
2
, with intermittent exercise. The inconsistent results of these studies have not
been satisfactorily explained. It is evident that a wide range of responses occur among asthmatics exposed to NO
2
. This
variation may in part reflect differences in subjects and exposure protocols: mouthpiece vs. chamber, obstructed vs.
non-obstructed asthmatics, sedentary vs. exercise, and requirements for medication. Identification of factors that
predispose to NO
2
responsiveness requires further investigation.
These studies have typically involved volunteers with mild asthma; data are needed from more severely affected
asthmatics who may be more susceptible. Overall, there is little convincing evidence that short-term exposures to NO
2
at outdoor ambient concentrations significantly alter lung function or nonspecific airway responsiveness in most people
The INDEX project Final report
81
with mild asthma. However, outdoor levels influence indoor concentrations, which may reach peak levels that are
clinically important for some adults and children with asthma.
Effects on Allergen Responsiveness:
The potential for NO
2
exposure to enhance responsiveness to allergen challenge in asthmatics deserves special mention.
Several recent studies have reported that low-level exposures to NO
2
,both at rest and with exercise, enhance the
response to specific allergen challenge in mild asthmatics. Tunnicliffe et al. (Tunnicliffe et al., 1994) reported exposures
of 8 subjects with asthma to 400 ppb NO
2
for only 1 hour at rest, and found increased responsiveness to a fixed dose of
allergen, both during the early and late phases of the response. No significant effect was seen at 100 ppb, but the data
suggested an exposure-response relationship. Davies’ group from the U.K., in two reports (Devalia et al., 1994;
Rusznak et al., 1996), described an effect of exposure to the combination of 400 ppb NO
2
and 200 ppb SO
2
, but not
either pollutant alone, on subsequent allergen challenge in mild asthmatics.
Strand and colleagues (Strand et al., 1998) from Sweden demonstrated increases in both the early and late phase
responses to allergen following 4 daily repeated exposures to 260 ppb NO
2
for 30 minutes, at rest. Finally, Jenkins et al.
(Jenkins et al., 1999) exposed asthmatic subjects to NO
2
, ozone, and their combination using two different protocols
that varied time of exposure and gas concentration, but kept the total exposure constant. All three exposures of the high
concentration regimen (200 ppb ozone, 400 ppb NO
2
, and the combination for 3 hours), but not the low concentration
regimen, enhanced subsequent responsiveness to allergen.
Additional data from both animal exposure and in vitro exposure studies provide support for enhancement of allergen
responsiveness by NO
2
exposure. Gilmour (Gilmour, 1995) has reviewed the evidence in animal models. Of particular
interest is a rat model of house-dust-mite sensitivity in which a 3-hour exposure to 9.4 mg/m
3
NO
2
, after a priming
injection and pulmonary challenge with antigen, increased the specific immune response and immune-mediated
pulmonary inflammation. NO
2
exposure also enhanced lymphocyte proliferation responses to allergen in both the spleen
and mediastinal lymph nodes.
Schierhorn et al. (Schierhorn et al., 1999) observed increased histamine release by cultured human nasal mucosa from
surgical resections in response to exposure to NO
2
at 200 and 800 µg/m3 (106 and 424 ppb) for 24 hours. The
magnitude of the effect was more pronounced than for ozone.
These recent studies involving allergen challenge appear relatively consistent in demonstrating effects at concentrations
that occur indoors, and suggest that NO
2
may enhance both allergen sensitization and its associated inflammatory
response. Confirmation of these findings is needed from other centers. However, the rising incidence, prevalence, and
mortality from asthma make these observations particularly important and timely. Additional work is needed in
understand more completely the exposure-response characteristics, effects of exercise, relationship to severity of
asthma, role of asthma medications, and other clinical factors. Animal and in vitro studies are needed to establish the
precise mechanisms involved.
Chronic Obstructive Pulmonary Disease:
Few studies have examined responses to NO
2
in subjects with chronic obstructive pulmonary disease (COPD). In a
group of 22 subjects with moderate COPD, Linn and associates (Linn et al., 1985a) found no pulmonary effects of 1-
hour exposures to 0.94, 1.88, and 3.76 mg/m
3
NO
2
. In a study by Morrow and colleagues (Morrow et al., 1992), 20
subjects with COPD were exposed for 4 hours to 0.56 mg/m
3
NO
2
in an environmental chamber, with intermittent
exercise. Although progressive decrements in lung function occurred during the exposure, significant decreases were
not found for FVC until the end of the exposure. The decrement in lung volume occurred without changes in flow rates.
The difference in results between the Linn and Morrow studies may reflect the difference in duration of exposure. It is
worth noting that changes in lung function were typical of the “restrictive” pattern seen with ozone rather than the
obstructive changes described by some with NO
2
exposure in asthmatics.
Effects on Host Defense:
More recently, investigators have sought to evaluate the effects of nitrogen dioxide on measures other than on
pulmonary function. The key studies are summarized here. Analysis of lung lavage from healthy humans indicated that
high levels (5640–7520 µg/m
3
; 3–4 ppm) reduce the activity of alpha-1-protease inhibitor, a protein that acts to protect
the lung from the proteolytic enzyme elastase by inhibiting connective tissue damage. However, 2820 µg/m
3
(1.5 ppm)
had no such effect.
Goings et al. (Goings et al., 1989) exposed healthy volunteers to either 1.9-5.6 mg/m
3
NO
2
or to air for 2 hours per day
for 3 consecutive days. A live, genetically engineered influenza A vaccine virus was administered intranasally to all
subjects after exposure on day 2. Infection was determined by virus recovery from nasal washings, a 4-fold or greater
increase in antibody titer, or both. The findings of this study were inconclusive, in part because of limitations in sample
The INDEX project Final report
82
size. In addition, the attenuated, cold-adapted virus used in the study was incapable of infecting the lower respiratory
tract, where NO
2
may have its greatest impact on host defense.
Another approach has been to obtain lavaged cells from NO
2
-exposed individuals and examine their handling of
infectious virus in vitro. Several NO
2
exposure scenarios, including continuous low-level exposure or intermittent peak
exposures have been examined (Frampton et al., 1989). Alveolar macrophages obtained by BAL 3 1/2 hours after a 3-
hour continuous exposure to 1.1 mg/m
3
NO
2
tended to inactivate influenza in vitro less effectively than cells collected
after air exposure. The effect was observed in cells from 4 of the 9 subjects studied; alveolar macrophages from these 4
subjects increased release of interleukin-1 after exposure to NO
2
, whereas cells from the remaining 5 subjects decreased
release of interleukin-1 following exposure. However, in a subsequent study (Azadniv et al., 1998) involving 3.8 mg/m
3
NO
2
exposures for 6 hours with intermittent exercise, no effect on alveolar macrophage function or inactivation of
influenza virus was observed, either immediately or 18 hours after exposure.
Airway Inflammation:
Unlike ozone exposure, NO
2
exposure at near-ambient levels (i.e., less than 3.8 mg/m
3
) does not cause a significant
influx of polymorphonuclear leukocytes (PMN) into the airways and alveoli (Frampton et al., 1989). NO
2
appears to be
much less potent than ozone in eliciting a neutrophilic inflammatory response.
However, prolonged exposure to NO
2
at concentrations only slightly above peak levels occurring indoors can cause
mild airway inflammation. Healthy volunteers exposed to 3.8 mg/m
3
NO
2
for 6 hours with intermittent exercise
(Azadniv et al., 1998) showed a slight increase in the percentage of PMN obtained in bronchoalveolar lavage fluid 18
hours after exposure (air: 2.2±0.3%; NO
2
: 3.1±0.4%). In a separate group of subjects, no effects of this exposure
protocol were found on alveolar macrophage phenotype or expression of the adhesion molecule CD11b or receptors for
IgG when assessed immediately after exposure (Gavras et al., 1994). Blomberg et al. (Blomberg et al., 1997) reported
that 4-hour exposures to 3.8 mg/m
3
NO
2
resulted in an increase in interleukin-8 and PMN in the proximal airways of
healthy subjects, although no changes were seen in bronchial biopsies. This group also studied the effects of repeated 4-
hour exposures to 3.8 mg/m
3
NO
2
on 4 consecutive days, with BAL, bronchial biopsies, and BAL fluid antioxidant
levels assessed 1.5 hours after the last exposure (Blomberg et al., 1999). The bronchial wash fraction of BAL fluid
showed a two-fold increase in PMN and a 1.5-fold increase in myeloperoxidase, indicating persistent mild airway
inflammation with repeated NO
2
exposure. Interestingly, small but significant decrements in FVC and FEV1 were
observed after the first exposure, which returned to baseline following subsequent exposures.
There is evidence from both animal and human studies that exposure to NO
2
may alter lymphocyte subsets in the lung
and possibly in the blood. Lymphocytes, particularly cytotoxic T cells and NK cells, play a key role in host defense
against respiratory viruses by eliminating infected host cells. Richters and colleagues (Damji & Richters, 1989)
(Richters & Damji, 1988; Richters & Richters, 1989; Kuraitis & Richters, 1989) showed that mice exposed to NO
2
at
levels as low as 7.5 mg/m
3
for eight hours demonstrate reductions in populations of CD8+ (cytotoxic/suppressor)
lymphocytes in the spleen. In humans, Sandstrom et al. (Sandstrom et al., 1991) observed a significant, dose-related
increase in lymphocytes and mast cells recovered by BAL 24 hours after a 20-minute exposure to NO
2
at 4.23 – 10.3
mg/m
3
. Rubinstein et al. (Rubinstein et al., 1991) found that a series of 4 daily 2- hour exposures to 1.1 mg/m
3
NO
2
resulted in a small increase in NK cells recovered by BAL. In contrast, repeated exposures to 2.8 or 7.5 mg/m
3
NO
2
for
20 minutes every 2nd day on six occasions resulted in decreased CD16+56+ and CD19+ cells in BAL fluid, 24 hours
after the final exposure (Sandstrom et al., 1992b; Sandstrom et al., 1992a). No effects were seen on PMN or total
lymphocytes. Finally, Azadniv et al. (Azadniv et al., 1998) observed a small but significant reduction in CD8+ T
lymphocytes in peripheral blood, but not BAL, 18 hr following single 6 hour exposures to 3.8 mg/m
3
NO
2
.
Differing exposure protocols and small numbers of subjects among these studies may explain the varying and
conflicting findings. Furthermore, the clinical significance of transient, small changes in lymphocyte subsets is unclear.
However, even small changes in susceptibility to respiratory viruses resulting from exposure to NO
2
may have a
significant public health impact because of the large number of individuals exposed in the home, both to NO
2
and to
respiratory viruses. However, clinical studies provide little evidence for effects on lung function, airway inflammation,
or host defense impairment in healthy subjects at outdoor ambient exposure concentrations.
Induction of Emphysema:
Clinical emphysema in humans has been linked with deficient proteinase inhibitor activity in the lung, presumably via
inactivation by cigarette smoke. One mechanism by which chronic NO
2
exposure may result in structural lung injury is
through inactivation of lung proteinase inhibitors. Animal models involving prolonged exposure to relatively high levels
of NO
2
have found pathological changes of emphysema (Evans et al., 1976; Lafuma et al., 1987). Mohsenin and Gee
(Mohsenin and Gee, 1987) exposed healthy volunteers to 5.6 or 7.5 mg/m
3
NO
2
for 3 hours and observed a 45%
decrease in the functional activity of a1-proteinase inhibitor in BAL fluid. Supplementation with vitamins C and E prior
to exposure abrogated the effect of 7.5 mg/m
3
NO
2
on elastase inhibitory capacity of the alveolar lining fluid
The INDEX project Final report
83
(Mohsenin, 1991). In contrast, Johnson et al. (Johnson et al., 1990) found no effect of exposure for 3 hours to
continuous 2.8 mg/m
3
or intermittent peaks of 3.8 mg/m
3
NO
2
on either the concentration (immunoassay) or functional
activity of a1-proteinase inhibitor in BAL fluid. The absence of an effect in the Johnson study may reflect the lower
exposure levels used.
Frampton et al. (Frampton et al., 1989) observed a 47% increase in a2-macroglobulin, a metalloproteinase inhibitor
released by alveolar macrophages, in BAL fluid 3 and 1/2 hours following 3-hour exposures to 1.1 mg/m
3
NO
2
. This
protein may have local immunoregulatory effects as well as provide local protection against proteinases. Its increase
following NO
2
exposure suggests a protective response. However, no change in BAL fluid levels of a2-macroglobulin
was observed following similar exposures to 2.8 mg/m
3
of NO
2
(Frampton et al., 1989).
Summary of short-term exposure effect levels
NOAEL
mg/m
3
LOAEL
mg/m
3
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
0.47
increase in airway reactivity
sensitive humans
(asthmatics), 1h
Review: California Air
Resources Board
(CARB), 1992
OEHHA 1999
REL: 0.47
0.19 0.38-0.56 increased responsiveness to bronchoconstrictors Mild asthmatics, 0.5h WHO 2000
GV: 0.2 (1h)
0.96 possible detrimental respiratory effects in both
normal and asthmatic subjects
Short-term human
clinical studies
Health Canada
1995
ASTER: 0.48 (1h)
OEHHA (1999)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Derivation of Acute Reference Exposure Level (protective against mild adverse effects): 0.25 ppm (470 mg/m³)
California Ambient Air Quality Standard
Study California Air Resources Board (CARB), 1992
Study population sensitive humans (asthmatics)
Exposure method inhalation
Critical effects increase in airway reactivity
LOAEL
NOAEL 0.25 ppm
Exposure duration 1 hour
Extrapolated 1 hour concentration 0.25 ppm
LOAEL uncertainty factor 1
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 1
Cumulative uncertainty factor 1
Reference Exposure Level 0.25 ppm (0.47 mg/m³; 470 mg/m³)
WHO Guidelines (2001)
Despite the large number of acute controlled exposure studies on humans, several of which used multiple
concentrations, there is no evidence for a clearly defined concentration–response relationship for nitrogen dioxide
exposure. For acute exposures, only very high concentrations (1990 µg/m3; > 1000 ppb) affect healthy people.
Asthmatics and patients with chronic obstructive pulmonary disease are clearly more susceptible to acute changes in
lung function, airway responsiveness and respiratory symptoms. Given the small changes in lung function (< 5% drop
in FEV1 between air and nitrogen dioxide exposure) and changes in airway responsiveness reported in several studies,
375–565 µg/m3 (0.20 to 0.30 ppm) is a clear lowest-observed-effect level. A 50% margin of safety is proposed because
of the reported statistically significant increase in response to a bronchoconstrictor (increased airway responsiveness)
with exposure to 190 µg/m3 and a meta-analysis suggesting changes in airway responsiveness below 365 µg/m3. (The
significance of the response at 190 µg/m3 (100 ppb) has been questioned on the basis of an inappropriate statistical
analysis.)
On the basis of these human clinical data, a 1-hour guideline of 200 µg/m
3
is proposed. At double this recommended
guideline (400 µg/m3), there is evidence to suggest possible small effects in the pulmonary function of asthmatics.
Should the asthmatic be exposed either simultaneously or sequentially to nitrogen dioxide and an aeroallergen, the risk
The INDEX project Final report
84
of an exaggerated response to the allergen is increased. At 50% of the suggested guideline (100 µg/m3, 50 ppb), there
have been no studies of acute response in 1 hour.
Effects of long-term exposure
Studies with animals have clearly shown that several weeks to months of exposure to NO
2
concentrations of less than
1880 µg/m
3
(1 ppm) cause a plethora of effects, primarily in the lung but also in other organs such as the spleen, liver
and blood. Both reversible and irreversible lung effects have been observed. Structural changes range from a change in
cell types in the tracheobronchial and pulmonary regions (lowest reported level 640 µg/m
3
) to emphysema-like effects
(at concentrations much higher than ambient). Biochemical changes often reflect cellular alterations (lowest reported
levels for several studies 380–750 µg/m
3
(0.2–0.4 ppm) but isolated cases of lower effective concentrations). Nitrogen
dioxide levels as low as 940 µg/m
3
(0.5 ppm) also increase susceptibility to bacterial and viral infection of the lung.
Several epidemiological studies show significant relationships between ambient (urban) NO
2
levels and health effects,
including respiratory symptoms, episodes of respiratory illness, lung function, and even mortality. However, because
NO
2
shares sources with other pollutants, one of the difficulties in deriving quantitative estimates of health risks is in
separating the relative contributions of NO
2
from those of other major pollutants (ozone, sulfur dioxide, particulate
matter, etc.); hence nitrogen dioxide might best be considered as just one indicator of polluted ambient air, especially
where it is present in traffic-dominated urban air pollution mixes. Great care is therefore needed in interpreting
available outdoor epidemiology studies, so as not to ascribe an undue degree of confidence to reported health
associations with nitrogen dioxide concentrations as being specifically due to that compound as compared to the overall
ambient air mix.
Indoor studies have compared groups exposed to nitrogen dioxide emitted from the combustion of gas inside buildings
to groups in homes without such sources and hence with lower levels of nitrogen dioxide. The limited studies of adults
have tended to show no relationship between the use of gas for cooking and respiratory symptoms or lung functions.
Thus, the following discussion focuses on children.
Hasselblad et al. (1992) performed a meta-analysis of studies in homes with gas stoves that met several criteria. The
endpoint was the presence of lower respiratory symptoms and disease in children aged 5–12 years. Given the nature of
the studies evaluated, the goal was to estimate the odds ratio of an increase in nitrogen dioxide concentration of
28.3 µg/m
3
(15 ppb). Although two analytic models (fixed and hierarchical) were used, they gave about the same
results. The combined odds ratio was about 1.2, with confidence intervals ranging from about 1.1 to 1.3. Thus, the
analysis estimated an increased risk of approximately 20% for respiratory symptoms and disease for each increase of
28.3 µg/m
3
(2-week average), where average weekly bedroom concentrations were between 15 and 122 µg/m
3
(8 and
65 ppb) (Hasselblad et al., 1992). Several uncertainties exist in this analysis, and exposure measurement errors are
present in the studies.
A number of additional studies have been published since 1990, with continued mixed results. A prospective cohort
study of infants conducted in Albuquerque, New Mexico (Samet et al., 1993) attempted to address many of the issues of
previous studies related to sample size and exposure misclassification. Exposures to NO
2
and respiratory illnesses were
monitored prospectively from birth to 18 months of age in a cohort of 1205 infants living in homes with gas and electric
cooking stoves, without smoking. NO
2
exposures were estimated from serial measurements of bedroom NO
2
concentrations. Respiratory illnesses were quantified from reports of symptoms and illnesses from mothers and
validated by home visits. No consistent trends in incidence or duration of illness were observed by level of NO
2
exposure at the time of illness or during the prior month, or by type of stove. However, indoor NO
2
levels were very
low in this study.
To further illustrate the indoor epidemiological findings, the study of Neas et al. (1991) is described in further detail
here. It was selected because the individual symptom results are consistent with the magnitude of effects found in the
British studies and other analyses of the data from six United States cities. The authors evaluated 1286 white children
(7–11 years) from the larger six-city study. For the children selected there was complete covariate information and at
least one indoor measurement of nitrogen dioxide (Palmes passive diffusion tubes for 2 weeks during the heating season
and 2 weeks during the cooling season at three sites: kitchen, activity room and the child’s bedroom). Parents completed
a questionnaire on symptoms during the previous year. An increase in symptoms was estimated for an additional
exposure of 28.3 µg/m3 (15 ppb). A multiple logistic model was used. The odds ratios were as follows: shortness of
breath, 1.23 (95% confidence interval (CI), 0.93–1.61); persistent wheeze, 1.16 (CI, 0.89–1.52); chronic cough, 1.18
(CI, 0.87–1.60); chronic phlegm, 1.25 (CI, 0.94–1.66); and bronchitis, 1.05 (CI, 0.75–1.47).When the authors
performed a multiple logistic regression of the combined lower respiratory symptom measure (i.e. the presence of any
The INDEX project Final report
85
of the above-mentioned symptoms), they obtained an odds ratio of 1.40 (CI, 1.14–1.72). Table 5.2 shows the adjusted
odds ratios for combined lower respiratory symptoms for ordered nitrogen dioxide exposure categories. These findings
are consistent with a linear concentration–response relationship.
Table 5.2: Odds ratio and 95% confidence interval for the effect of nitrogen dioxide exposure
on the prevalence of lower respiratory symptoms in children (Neas et al., 1991)
Nitrogen dioxide concentration
(µg/m
3
)
Range Mean
Number of children
Odds ratio
95%Confidence
interval
0–9 7 263 1.00
9–19 14 360 1.06 0.71–1.58
19–37 27 317 1.36 0.89–2.08
38–147 58 346 1.65 1.03–2.63
More recent studies have utilized personal monitoring methods in an attempt to improve exposure classification.
Mukala et al. (Mukala et al., 1999) prospectively studied personal exposure to NO
2
for periods of 13 weeks among 163
preschool children in Helsinki, using individual passive diffusion monitors. Daily diaries of symptoms were kept by the
parents, and in a subset of 53 children, peak expiratory flow rates were measured in the morning and evening. Co-
variates considered in the model included allergy, education, smoking, stove type, and outdoor pollutant concentrations
(NO, NO
2
, O
3
, SO
2
, and total suspended particles). The median personal NO
2
exposure was 21.1 µg/m
3
(11 ppb), with a
maximum of 99 µg/m
3
(50 ppb). An increased risk of cough was associated with increasing NO
2
exposure (risk ratio =
1.52; 95% confidence interval 1.00-2.31). There were no significant effects on other respiratory symptoms or peak flow.
In Australia, where unvented natural gas cooking and heating are common, Pilotto et al. (Pilotto et al., 1997) queried
respiratory symptoms and school absences among 388 children from 6 to 11 years of age, and monitored indoor NO
2
levels at their schools, which were chosen for having either unvented gas heating or electric heating. Classroom
monitoring of NO
2
levels was conducted intermittently over several months. A significant increase in sore throat, colds,
and absences from school were found for children in environments with hourly peak levels of 150 µg/m
3
, compared
with background levels of 38 µg/m
3
. Exposure-response relationships were evident for each outcome. However, no
measurements of other pollutants, either indoor or outdoor, were provided. Caution must be used in interpreting the
findings from cross-sectional studies, because many factors other than pollutant levels may influence differences
between populations.
In the Latrobe Valley of Australia (Garrett et al., 1998), NO
2
levels were monitored in eighty homes, on 5 separate
occasions for 4 days each, and health questionnaires administered to the 148 children residing in those homes. 58 of the
children were asthmatic, although the diagnostic criteria were not provided. Children underwent allergy prick testing
and monitored their peak flow rates for a 2-week period in the winter and spring. The indoor median NO
2
concentration
was 11 µg/m
3
(6.0 ppb), with a maximum of 241 µg/m
3
(128 ppb). Respiratory symptoms were more common in
children exposed to a gas stove (odds ratio 2.3, CI 1.00-5.2), even after adjusting for NO
2
levels (odds ratio 2.2, CI 1.0-
4.8). Atopic children tended to have a greater risk than non-atopic children. NO
2
concentration was not a significant risk
factor for symptoms. The authors conclude that gas stoves may pose a risk apart from NO
2
. However, the relative
paucity of NO
2
monitoring data for each home may have provided insufficient statistical power to demonstrate an
association. More important weaknesses in the study are the inclusion of homes with cigarette smokers, and the failure
to monitor other pollutants, either inside or outside the home. These factors may have confounded the findings.
Jarvis et al. (1996) studied symptoms, lung function, and atopy in 15000 adults aged 20-44 years in Britain, as part of
the European Community Respiratory Health Survey. Women, but not men, who reported cooking with gas had an
increased risk for symptoms consistent with asthma, such as wheezing (odds ratio (OR) 2.07, CI 1.41- 3.05), waking
with shortness of breath (OR 2.32, CI 1.25-4.34), and “asthma attacks” (OR 2.60, CI 1.20-5.65). Lung function was
measured in a subset of subjects, and FEV1 was reduced 3.1% of predicted for women cooking with gas compared to
those using other means, after adjusting for age, smoking, and town of residence. Total and specific IgE levels were not
associated with gas stove use. There was no protective effect associated with use of an exhaust fan. The authors boldly
concluded from their estimate of the population attributable risk fraction that “the prevalence of wheeze with
breathlessness in young women would be reduced by between 8% and 48% if cooking with gas were abandoned.
Although studies such as this are limited by the potential for exposure misclassification and the influence of other
environmental and biological factors, the findings are consistent with women spending more time at cooking than men,
and with reports of increased responsiveness to allergen challenge following NO
2
exposure (see below).
The INDEX project Final report
86
Taken together, studies of the health effects of exposure to NO
2
indoors fail to make a convincing case for association
with respiratory illness in either children or adults. The findings of the Hasselblad meta-analysis (Hasselblad et al.,
1992) must be interpreted with caution because the 11 studies used in the analysis employed varying methodologies and
study populations. Small sample size, potential for misclassification, inclusion of smokers in many of the studies, and
failure to consider potential effects of outdoor pollution, or other indoor pollutants, may bias many of the studies. For
example, burning of natural gas in gas stoves emits ultrafine particles in addition to NO
2
, and the cooking process is
also a source of particles. It is possible that observed health effects associated with gas stove use may represent health
effects of particle exposure, or of particles combined with NO
2
. This may explain why Garrett et al. (Garrett et al.,
1998), found a significant relationship between respiratory symptoms in children and gas stove use, but not indoor NO
2
levels. The Samet et al. study of infants in Albuquerque (Samet et al., 1993) provides convincing evidence that indoor
NO
2
, at the very low concentrations found in that study, are not associated with respiratory illnesses in children under
18 months of age.
Recently reported time-series analyses of ambient air pollution effects from a European multi-city epidemiology project
(Katsouyanni et al., 1996) also found the following very mixed results.
- There was no association with any cause of death for nitrogen dioxide or ozone in Lyon, France, where daily nitrogen
dioxide concentrations averaged 70 µg/m
3
(maximum 324 µg/m
3
) and both sulfur dioxide and particulate matter were
significantly related to mortality from cardiovascular and respiratory causes (Zmirou et al., 1996).
- No relationship between daily nitrogen dioxide levels and daily mortality was found in Cologne, Germany, where the
all-year median for daily nitrogen dioxide was 45 µg/m
3
(maximum 176 µg/m
3
) and both sulfur dioxide and
particulate matter were significantly related to increased mortality risk (Spix and Wichmann, 1996).
- In Paris, there were no significant effects of photo-oxidant pollutants (nitrogen dioxide, ozone) on daily mortality
risk, but there was a significantly increased hospital asthma admission relative risk (RR) of 1.175 per 100 µg/m
3
increase in nitrogen dioxide concentration above the reference value. The mean 24-hour nitrogen dioxide
concentration averaged 45 µg/m
3
(99th centile 108 µg/m
3
) and the mean 1-hour maximum was 74 µg/m
3
(99th centile
203 µg/m
3
). Particulate matter and sulfur dioxide significantly increased daily mortality from respiratory causes and
hospital admissions for all respiratory disease (Dab et al., 1996).
- Negative (sometimes statistically significant) effects of nitrogen dioxide in decreasing respiratory hospital admissions
(RR = 0.86–0.89) were found in Amsterdam for adults (aged 15–64 years) and in chronic obstructive pulmonary
disease admissions for all ages (RR = 0.795–0.948), but there were some positive effects on respiratory admissions
for the elderly (65+ years) and on asthma admissions (RR = 0.902–1.062). The mean 24-hour nitrogen dioxide
concentration averaged 50 µg/m
3
(95th centile 84 µg/m
3
) and the mean 1-hour maximum was 75 µg/m
3
(95th centile
127 mg/m
3
). In addition, ozone showed nonsignificant positive effects on summer respiratory admissions for the
elderly (65+ years) but neither particulate matter nor sulfur dioxide showed any clear effects on hospital admissions
(Schouten et al., 1996).
- Rotterdam showed contrasting results, i.e. predominantly positive (albeit few significant at P < 0.05) effects of
nitrogen dioxide in terms of relative risk increases for all respiratory admissions for all age groups and increases
(mostly significant) for chronic obstructive pulmonary disease admissions for all ages. The mean 24-hour nitrogen
dioxide concentration averaged 54 µg/m
3
(95th centile 86 µg/m
3
) and the mean 1-hour maximum averaged 82 µg/m
3
(95th centile 138 µg/m
3
). Ozone and particulate matter (as black smoke) generally increased the risk of hospital
admissions, and sulfur dioxide showed mixed results (Schouten et al., 1996).
- Ambiguities regarding the effects of ambient nitrogen on hospital admissions for asthma were found in Helsinki
(daily mean in the range 34–44 µg/m
3
). Pönkä & Virtanen (1996) reported asthma admissions for children (aged 0–14
years) to be related to 8-hour ozone levels and for adults (15–64 years) to 24-hour sulfur dioxide levels, but provided
no results for nitrogen dioxide. This was in contrast to an earlier report (Pönkä, 1991) of significant associations
between hospital admissions for asthma and nitrogen dioxide, sulfur dioxide, ozone and total suspended particulates
based on different statistical modelling.
- In London, the effects of nitrogen dioxide and sulfur dioxide on daily mortality were significant but smaller and less
consistent than the effects of ozone and particulate matter (as black smoke) on respiratory, cardiovascular and all-
cause mortality, the effects being greater in the warm season (April–September) and independent of other pollutants
(Anderson et al., 1996).
The INDEX project Final report
87
Summary of long-term exposure effect levels
NOAEL
mg/m
3
LOAEL
mg/m
3
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
0.038-
0.056
excess of lower respiratory illness Children 5-12 y old,
annual avg conc. , also
basing on background
level of 0.015 mg/m³
taken from EHC 188 WHO 2000
GV: 0.04 (1y)
WHO Guidelines (2001)
Although there is no particular study or set of studies that clearly support selection of a specific numerical value for an
annual average guideline, the database nevertheless indicates a need to protect the public from chronic nitrogen dioxide
exposures. For example, indoor air studies with a strong nitrogen dioxide source, such as gas stoves, suggest that an
increment of about 30 µg/m3 (2-week average) is associated with a 20% increase in lower respiratory illness in children
aged 5–12 years. However, the affected children had a pattern of indoor exposure that included peak exposures higher
than those typically encountered outdoors. Thus the results cannot be readily extrapolated quantitatively to the outdoor
situation. Outdoor epidemiological studies have found qualitative evidence of ambient exposures being associated with
increased respiratory symptoms and lung function decreases in children (most clearly suggestive at annual average
concentrations of 50–75 µg/m3 or higher and consistent with findings from indoor studies), although they do not
provide clear exposure–response information for nitrogen dioxide. In these epidemiological studies, nitrogen dioxide
has appeared to be a good indicator of the pollutant mixture. Furthermore, animal toxicological studies show that
prolonged exposures can cause decreases in lung host defences and changes in lung structure. On these grounds, it is
proposed that a long-term guideline for nitrogen dioxide be established. Selecting a well-supported value based on the
studies reviewed has not been possible, but it has been noted that a prior review conducted for the Environmental
Health Criteria document on nitrogen oxides recommended an annual value of 40 µg/m3 (WHO, 1997). In the absence
of support for an alternative value, this figure is recognized as an air quality guideline.
Carcinogenic and mutagenic effects
To date, there are no reports that nitrogen dioxide causes malignant tumours or teratogenesis (U.S.EPA, 1993; Berglund
et al., 1993; Witschi, 1988). Limited genotoxicity studies have produced mixed results with in vitro and high-
concentration in vivo studies (e.g. 50 000 µg/m
3
, 27 ppm) (Victorin, 1994).
Interaction with other chemicals
Environmental exposures to NO
2
do not occur singly, but rather as a complex mixture of pollutants, and failure to
consider the presence of other pollutants may confuse interpretation of the observed effects. Recent data suggest
exposure to low concentrations of NO
2
at rest may enhance the response to allergen inhalation in subjects with asthma.
When considering mixtures of anthropogenic pollutants, It may be impossible to separate the effects of one component
from those of other components, particularly with the possibilities of synergistic or antagonistic interactions. In
considering the health effects of mixtures, potential causal pathways should be carefully delineated. For example, some
reports have suggested that HONO may contribute to the health effects attributed to indoor NO
2
(Spengler et al., 1990).
Efforts have been made to study effects of NO
2
-ozone mixtures on pulmonary function. These studies have generally
revealed no interactive effects; the observed pulmonary function decrements appear to reflect the ozone component of
the mixtures. Hazucha et al., (Hazucha et al., 1994) found that pre-exposure of healthy women to 1.1 mg/m
3
(0.6 ppm)
NO
2
for 2 hours enhanced the development of nonspecific airway responsiveness induced by a subsequent 2-hour
exposure to 0.6 mg/m
3
(0.3 ppm) ozone, with intermittent exercise.
Relatively high-level, prolonged (6 hours/day, up to 90 days) exposure to NO
2
(27 mg/m
3
) and ozone (1.6 mg/m
3
)
results in a syndrome of progressive pulmonary fibrosis in rats (Rajini et al., 1993), associated with a sustained increase
in procollagen gene expression in the central acini (Farman et al., 1999). This does not occur with either gas alone,
indicating a true synergistic effect. The relevance of this observation for human ambient exposures is not clear, given
the high exposure concentrations used in the study, and absence of evidence for alveolar fibrosis or restrictive lung
disease in epidemiological studies.
The INDEX project Final report
88
Bermudez et al. (Bermudez et al., 1999) examined DNA strand breaks in BAL cells from rats exposed to ozone (0.6
mg/m
3
), to NO
2
(2.3 mg/m
3
), and the combination. Ozone and the combination exposure increased DNA strand breaks
to a similar degree compared with air exposure, but NO
2
alone had no effect.
The effects of NO
2
exposure on SO
2
-induced bronchoconstriction have been examined, but with inconsistent results.
Jorres and Magnussen (Jorres & Magnussen, 1991) found an increase in airways responsiveness to SO
2
in asthmatic
subjects following exposure to 0.47 mg/m
3
NO
2
for 30 minutes, yet Rubinstein et al. (Rubinstein et al., 1990) found no
change in responsiveness to SO
2
inhalation following exposure of asthmatics to 0.56 mg/m
3
NO
2
for 30 minutes.
Overall, there are little definitive data suggesting that NO
2
interacts with other pollutants in causing human health
effects. However, human clinical studies have not systematically addressed the effects of pollutant combinations
containing NO
2
, in part because of the complexity of the experimental design and the difficulty in studying the most
susceptible subjects.
Odour perception
Source: Devos et al. 1990
Odour thresholds: 185 µg/m
3
Source: Berglund et al., 1993
Nitrogen dioxide has a stinging, suffocating odour. The odour threshold has been placed by various authors at between
94 µg/m
3
(0.05 ppm) and 410 µg/m
3
(0.22 ppm). Owing to adaptation, however, no odour was perceived during a
gradual (15-minute) increase in concentration from 0 to 51 mg/m
3
(0–27 ppm).
Source AIHA, 1989 (Odor thresholds for chemicals with established occupational health standards. American Industrial
Hygiene Association, 1989. p. 25, 70-71)
Minimum perceptible value: 207–263 µg/m
3
(0.11-0.14 ppm).
Source: Amoore and Hautala, 1983
Odour thresholds: 733 µg/m
3
(0.39 ppm)
The INDEX project Final report
89
Summary of Nitrogen dioxide Dose Response Assessment
________________________________________________________________________________________________
Exposure other than inhalation: Does not exist in water (food), reacting instantaneously to form nitric and nitrous
acids
Toxicokinetics: 70-90% absorbed in the respiratory tract (increased during exercise). Oral breathing increases the
delivery to the lower respiratory tract. Produces oxidation of either membrane lipids or proteins resulting in the loss of
cell permeability control.
Health effect levels of short- and long-term exposure
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
Short-term exposure
0.47
increase in airway reactivity
sensitive humans
(asthmatics), 1h
Review: California
Air Resources Board
(CARB), 1992
OEHHA 1999
REL: 0.47
0.19 0.38-0.56 increased responsiveness to
bronchoconstrictors
Mild asthmatics, 0.5h WHO 2000
GV: 0.2 (1h)
0.96 possible detrimental respiratory effects in both
normal and asthmatic subjects
Short-term human
clinical studies
Health Canada
1995
ASTER: 0.48
(1h)
Long-term exposure
0.038-
0.056
excess of lower respiratory illness Children 5-12 y old,
annual avg conc. ,
also basing on
background level of
0.015 mg/m³
taken from EHC 188 WHO 2000
GV: 0.04 (1y)
Carcinogenicity: No evidence
Genotoxicity: Limited evidence in vitro and in vivo at high concentrations (50 mg/m³)
Odour threshold: Range: 0.09-0.41 mg/m³ (Berglund), 0.185 mg/m³ (Devos), 0.2 mg/m³ (AIHA). Owing to
adaptation, however, no odour was perceived during a gradual (15-minute) increase in concentration from 0 to 51
mg/m
3
Susceptible population:
- Asthmatic individuals appear to be the most susceptible members of the population. Exposure of asthmatics to NO
2
could increase airway responsiveness to a variety of provocative mediators, including cholinergic and histaminergic
chemicals, SO
2
, and cold air.
- Individuals with chronic obstructive pulmonary disease (COPD: emphysema and chronic bronchitis).
- Children are at increased risk for respiratory illness; of concern because repeated lung infections in children can cause
lung damage later in life (greater lung surface area/body weight ratios and increased minute volumes/weight ratios;
increased exposure of children is finally expected due to higher levels of nitrogen dioxide found nearer to the
ground).
Remarks: High uncertainties in exposure-response relationships for both acute (<3-hour) and long-term exposure; high
uncertainties in establishing an appropriate margin of protection.
The INDEX project Final report
90
6. Risk characterization
Health hazard evaluation of short- and long-term exposure
[Adopted from WHO, 2000]
Despite the large number of acute controlled exposure studies on humans, several of which used multiple
concentrations, there is no evidence for a clearly defined concentration–response relationship for nitrogen dioxide
exposure. For acute exposures, only very high concentrations (2000 µg/m
3
) affect healthy people. Asthmatics and
patients with chronic obstructive pulmonary disease are clearly more susceptible to acute changes in lung function,
airway responsiveness and respiratory symptoms. Given the small changes in lung function (< 5% drop in FEV
1
between air and nitrogen dioxide exposure) and changes in airway responsiveness reported in several studies, 375-565
µg/m
3
(0.20 to 0.30 ppm) is a clear lowest-observed-effect level. A 50% margin of safety is proposed because of the
reported statistically significant increase in response to a bronchoconstrictor (increased airway responsiveness) with
exposure to 190 µg/m
3
and a meta-analysis suggesting changes in airway responsiveness below 0.36 mg/m
3
. (The
significance of the response at 190 µg/m
3
(0.1 ppm) has been questioned on the basis of an inappropriate statistical
analysis).
On the basis of these human clinical data, a 1-hour exposure limit of 200 µg/m
3
is proposed. At double this
recommended guideline (400 µg/m
3
), there is evidence to suggest possible small effects in the pulmonary function of
asthmatics. Should the asthmatic be exposed either simultaneously or sequentially to nitrogen dioxide and an
aeroallergen, the risk of an exaggerated response to the allergen is increased. At 50% of the suggested guideline
(100 µg/m
3
, 50 ppb), there have been no studies of acute response in 1 hour.
Although there is no particular study or set of studies that clearly support selection of a specific numerical value for an
annual average guideline, the database nevertheless indicates a need to protect the public from chronic nitrogen dioxide
exposures. For example, indoor air studies with a strong nitrogen dioxide source, such as gas stoves, suggest that an
increment of about 30 µg/m
3
(2-week average) is associated with a 20% increase in lower respiratory illness in children
aged 5–12 years. However, the affected children had a pattern of indoor exposure that included peak exposures higher
than those typically encountered outdoors. Thus the results cannot be readily extrapolated quantitatively to the outdoor
situation. Outdoor epidemiological studies have found qualitative evidence of ambient exposures being associated with
increased respiratory symptoms and lung function decreases in children (most clearly suggestive at annual average
concentrations of 50–75 µg/m
3
or higher and consistent with findings from indoor studies), although they do not
provide clear exposure–response information for nitrogen dioxide. In these epidemiological studies, nitrogen dioxide
has appeared to be a good indicator of the pollutant mixture. Furthermore, animal toxicological studies show that
prolonged exposures can cause decreases in lung host defences and changes in lung structure.
On these grounds, it is proposed that a long-term guideline for nitrogen dioxide be established. Selecting a well-
supported value based on the studies reviewed has not been possible, but it has been noted that a prior review conducted
for the Environmental Health Criteria document on nitrogen oxides recommended an annual value of 40 µg/m
3
. In the
absence of support for an alternative value, this figure is recognized as an air quality guideline.
Exposure data of relevance for the European population
Table 6.1: Short-term NO
2
peak levels related to gas appliances (adapted from Chapter 1.3)
Studies based on NO
2
sources µg/m
3
Gas appliances in kitchen, 1h-TWA (Lebret. 1987) 230 – 2055
Gas stoves in kitchen, 1h-TWA (Noy, 1990) 2500
Gas cooking, 1h-TWA (Ross, 1996) 342 – 1585
Homes with gas appliances, 1h-TWA (WHO, 2000) 2000
Unflued gas heaters, ?-TWA (Brown, 2002) 180 – 530
The INDEX project Final report
91
Table 6.2: Percentage of population exposed beyond given guideline values for NO
2
Available exposure data
Indoor concentrations (48h-PEM)
WHO recommended value
1-y average
German GV II
1-week average
Description (Study, Year) N 40 µg/m
3
* 60 µg/m
3
Helsinki (Expolis, 96-98) 175 10% (15 %) <
Basel (Expolis, 96-98) 50 15% (15 %) 5 %
Prague (Expolis, 96-98) 33 50% (50 %) 25 %
Oxford (Expolis, 98-00) 40 20% (20 %) 10 %
Northen Italy-TWA (Simoni, 2002) 112 75% 45 %
England 2-w TWA (BRE, 97-99) 796 25% 20 %
percentages put in parenthesis result from “through the day” 48h-personal exposure monitorings
* also EU annual limit value for the protection of human health (total of NO
2
and oxides of nitrogen) to be met the 01/01/2010 (see
margins of tolerance hereafter)
< out of the evaluation range (i.e. <5% of the environments investigated)
Guideline Value II (GV II): GV II is a health-related value based on current toxicological and epidemiological knowledge. If the concentration
corresponding to GV II is reached or exceeded immediate action must be taken because permanent stay in a room at this concentration level is likely
to represent a threat to health, especially for sensitive people. In this context, taking action means an immediate examination of the situation with
regard to a need for control measures. It may include the evacuation of the room in question. If by measurement GV II has been found to be exceeded,
the results should be checked by repetitive measurements carried out immediately under normal conditions of occupancy. If possible and deemed
meaning ful, biological monitoring of the occupants should be carried out in addition.
EU limit values for nitrogen dioxide NO
2
and oxides of nitrogen NO
X
– Directive 1999/30/EC
Averaging period Limit value
Margin of tolerance
Date by which limit
value is to be met
1. Hourly limit value
for the protection
of human health
1 hour 200 µg/m
3
NO
2
,
not to be exceeded
more than 18 times a
calendar year
50 % on the entry into force of this
Directive, reducing on 1 January 2001
and every 12 months thereafter by
equal annual percentages to reach 0%
by 1 January 2010
1 January 2010
2. Annual limit value
for the protection of
human health
Calendar year 40 µg/m
3
NO
2
50 % on the entry into force of this
Directive, reducing on 1 January 2001
and every 12 months thereafter by
equal annual percentages to reach 0%
by 1 January 2010
1 January 2010
3. Annual limit value
for the protection of
vegetation
Calendar year 30 µg/m
3
NO
2
None 19 July 2001
Alert threshold for nitrogen dioxide
400 µg/m3 measured over three consecutive hours at locations representative of air quality over at least 100 km 2 or an entire zone or agglomeration,
whichever is the smaller.
Minimum details to be made available to the public when the alert threshold for nitrogen dioxide is exceedeed
Details to be made available to the public should include at least:
- the date, hour and place of the occurrence and the reasons for the occurrence, where known;
- any forecasts of:
- changes in concentrations (improvement, stabilisation, or deterioration), together with the reasons for those changes,
- the geographical area concerned,
- the duration of the occurrence;
- the type of population potentially sensitive to the occurrence;
- the precautions to be taken by the sensitive population concerned
The INDEX project Final report
92
Result
Reported maximum nitrogen dioxide (NO
2
) levels associated with the use of gas appliances in homes (gas cooking and
heating) are in the range 180-2500 µg/m
3
. Exposure at these levels could generate effects in the pulmonary function of
asthmatics, considered to be the subjects most susceptible to acute NO
2
exposure, with the lower end of the range
approximating the WHO guideline (200 µg/m
3
, 1-hour average), established for the protection of asthmatic individuals
and the upper end starting to affect health in normal individuals.
For long-term exposures, increased respiratory symptoms and lung function decreases in children were shown to be the
most sensitive effect in the general population. Measured background levels in European homes indicate that a
remarkable portion of the population is exposed at NO
2
levels higher than current guideline values protecting from
respiratory effects in children. In up to 25% of the investigated residences (45% in an Italian study) NO
2
levels
exceeded the German indoor-related guideline value (GV II: 60 µg/m
3
, 1-week average), what would have resulted in
immediate action i.e. the examination of the situation with regard to a need for control measures.
On the other hand, safe levels in homes, i.e. < 40 µg/m
3
(following the WHO recommended annual value), are not
likely to be achievable everywhere (e.g. in areas with intense automotive traffic) given that ventilation alone may
introduce outdoor air containing such concentrations.
The INDEX project Final report
93
Benzene
Synonyms: Annulene, benzine, benzol, benzole, benzol coal naphtha, cyclohexatriene,
mineral naphtha, motor benzol, phenyl hydride, pyrobenzol, pyrobenzole
CAS Registry Number: 71-43-2
Molecular Formula: C
6
H
6
1. Compound identification
Benzene is a naturally occurring colourless liquid at room temperature and ambient pressure, and it has a characteristic
aromatic odour. It is widely used toxic, volatile, flammable organic solvent. It is used as an industrial solvent for
example in paints, varnishes, lacquer thinners and gasoline (1-4%) (IARC, 1989, Holmberg and Lundberg, 1985).
Presently it is used as a raw material in the synthesis of styrene, phenol, cyclohexane, aniline, and alkyl benzenes in the
manufacture of various plastics, resins, and detergents. Syntheses of many pesticides and pharmaceuticals also involve
benzene as a chemical intermediate (HSDB, 2003). The tire industry and shoe factories use benzene extensively in their
manufacturing processes. Benzene causes central nervous system damage acutely and bone marrow damage chronically
and is carcinogenic. It was formerly used as parasiticide (SIS 2003).
2. Physical and Chemical properties
Molecular weight (g/mol) 78.11
Melting point (°C) 5.5
Boiling point (°C) 80.1
Density (g/l at 20 °C, 1 atm) 0.88
Relative vapour density (air =1) 2.8
Solubility: Water (g/l) 1.8
Miscible in most organic solvents.
Conversion factors at 20 °C and 760 mm Hg:
1 ppb = 3.242 µg/m
3
1 µg/m
3
= 0.308 ppb
Sources: GDCh (1988), RIVM (1988) and ATSDR (1989), Verschueren 2001, HSDB (2003).
3. Indoor Air Exposure assessment
Emission sources
Probably the most important indoor source is cigarette smoking. Other remarkable indoor sources are emissions from
consumer products, including off-gassing from particle board. Similarly, living in the vicinity of hazardous waste sites
or industrial facilities may increase exposure to benzene (ATSDR, 1991).
In a German study, being a subject to environmental tobacco smoke together with driving a car and refuelling were
found significant determinants of exposure to benzene (Hoffman et al 2000). Similar results were reported in the US
TEAM study (Wallace, 1989). It appeared that personal sources (use of products emitting benzene, driving or riding in
automobiles), automobile exhaust and smoking (active and passive) were major sources of benzene to the general
population. Inhalation exposures accounted for more than 99% of the general population exposure to benzene in US
according to Hattemer-Frey et al (1990).
The INDEX project Final report
94
The average daily intake for an adult in Canada was estimated to be about 200 µg/day in total: 14 µg from ambient air,
140 µg from indoor air, 1.4 µg each from food and drinking water and 50 µg from automobile-related activities. The
corresponding calculated average daily intake in the USA is 320 µg, with a daily intake from ambient and indoor air
ranging between 180 - 1300 µg. Cigarette smoking may add to that as much as 1800 µg/day and passive smoking 50
µg/day. Based on assumptions of spending 2 hours per day in urban ambient air at 7 µg/m
3
, 21 hours per day in indoor
air at 4 µg/m
3
, and 1 hour per day inside a vehicle at 50 µg/m
3
(typical for large roads and heavy traffic), calculated
relative uptakes from urban ambient air, indoor air, air inside cars, and intake from food were 9; 53; 30 and 8%,
respectively (WHO, 1996).
Indoor air and exposure concentrations
Mean indoor concentrations are typically higher than the respective ambient levels all over Europe. The trend showing
increasing ambient concentrations of benzene from northern to southern European cities (Cocheo et al 2000) seems to
be similar for the indoor air concentrations (Jantunen et al 1999). In Northern Europe (Helsinki) the mean indoor
concentration, 2.1 µg/m
3
, was slightly higher than the respective outdoor concentration, 1.7 µg/m
3
, if tobacco smoke
was not present indoors (Edwards et al 2001). When tobacco smoke was present in indoor air, indoor concentration of
benzene was more than twofold compared to outdoor level. Personal exposures were typically higher than indoor
concentrations. The highest mean exposure in Helsinki was found due to active smoking being, 2.4 times higher than
the mean indoor concentration in homes without tobacco smoke. In addition to smoking, residential indoor benzene
concentration, time spent in a car, workplace concentration and time spent in home workshop determined personal
exposures to benzene (Edwards et al 2001, Edwards and Jantunen 2001). Similar pattern between mean indoor, outdoor
and personal exposure concentrations could be seen in central and southern European cities, although the concentrations
were higher. Mean indoor concentrations ranged between 2.3 and 12 µg/m
3
in central European cities such as Basel,
Erfurt, Hamburg, Prague and Antwerp (Jantunen et al 1999, Schneider et al 1999 and 2001, Cocheo et al 2000) and
between 10 and 13 µg/m
3
in southern cities like Milan and Athens (Jantunen et al 1999, Cocheo et al 2000). The mean
personal exposures to benzene in central Europe ranged from 6 µg/m
3
in Basel, Switzerland (Jantunen et al 1999) to 14
µg/m
3
in Germany (Hoffman et al 2000). Carrer et al (2000) studied personal exposures to benzene of 100 office
workers in Milan showing an average exposure of 21.1 µg/m
3
. The highest reviewed mean population exposure to
benzene in Southern Europe was measured in Murcia being 23 µg/m
3
(Cocheo et al 2000).
In a German population based study of children and teenagers (GerES IV), weekly mean exposure to benzene was
higher than the mean residential indoor concentration being 2.5 µg/m
3
and 1.6 µg/m
3
respectively. The highest exposure
and indoor concentrations were 7.7 µg/m
3
and 7.0 µg/m
3
respectively (Ullrich et al 2002).
Based on these results we can conclude, that in addition to outdoor sources of benzene, there are important indoor
sources like smoking, but clearly also emissions from other indoor sources.
Cumulative frequency distributions of the indoor and 48-hour personal exposure concentrations in European studies are
presented in Figure 3.1, Figure 3.2, Figure 3.3 and Figure 3.4.
Source related short time concentrations of benzene are presented in Figure 3.5. Lee and Wang (2004) studied
emissions of incense burning in chamber (18 m
3
) tests. They found that all 10 tested incense types caused
concentrations exceeding the Recommended Indoor Air Quality Objectives for Office Buildings and Public Places in
Hong Kong (HKIAQO) of 16.1 µg/m
3
. Maximum benzene concentrations peaked up to 117 µg/m
3
during incense
burning. Brown (2002) studied short time (30-50 min) concentrations of benzene in established and new buildings in
Australia, reporting mean concentrations of 7 µg/m
3
and <30 µg/m
3
respectively. The highest measured indoor
concentration was 81 µg/m
3
. Emission sources of this high concentration could not be identified, but high indoor
concentrations (16 – 19 µg/m
3
) in two other houses were related to attached garages. In general, indoor concentrations
of VOCs were much higher in new and recently renovated houses persisting above ‘baseline level’ for several weeks.
Concentration decay rates correlated with molecular volume, indicating the role of diffusion processes for limiting the
emissions. Elke at al (1998) measured short time concentrations of benzene inside buildings (60-min average), inside a
train and a car (30-min average). Slightly higher concentrations were measured during driving a car than when exposed
to tobacco smoke in a smoker compartment of a train or in a smoker day room.
Short time exposure scenarios for potential consumer activities were analysed in European Union Risk Assessment
Report (EU 2003). The highest benzene concentration, 150 – 240 µg/m
3
were expected to be present in a mainstream
tobacco smoke. Exposure of passive smokers to benzene from smoking was assumed to be 7 µg/m
3
, exposure from
painting and from car interior accessories 17 µg/m
3
and 12 µg/m
3
respectively.
The INDEX project Final report
95
0
20
40
60
80
100
0 10203040506070
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
Hel
Mil
Oxf
Pra
Figure 3.1. Cumulative frequency distributions of 30-hour indoor air concentrations of benzene in Athens (Ath, n=42),
Basel (Bas, n=47), Helsinki (Hel, n=188), Milan (Mil, n=41) Oxford (Oxf, n=40) and Prague (Pra, n=46) (EXPOLIS
2002).
0
20
40
60
80
100
0 10203040506070
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Ant
Ath
Cop
Mur
Pad
Rou
Figure 3.2. Cumulative frequency distributions of 5-day indoor air concentrations of benzene in Antwerp (Ant), Athens
(Ath), Copenhagen (Cop), Murcia (Mur), Padova (Pad) and Rouen (Rou) (n= 50 in each city) measured in the
MACBETH study (Cocheo et al 2000).
The INDEX project Final report
96
0
20
40
60
80
100
0 5 10 15 20
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Benzene 28-day
Figure 3.3. Cumulative frequency distribution of 28-day indoor air concentrations of benzene in England (GM= 3.0
µg/m3, max 93.5 µg/m3, n=796, Brown et al 2002).
0
20
40
60
80
100
0 10203040506070
Exposure (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
GeS
Hel
Oxf
Pra
Figure 3.4. Cumulative frequency distributions of 48-hour personal exposure concentrations of benzene in Athens
(Ath), Basel (Bas), Helsinki (Hel), Oxford (Oxf) and Prague (Pra) (EXPOLIS 2002), and 1-week mean concentrations
of the German Survey GerES II (Hoffman et al 2000).
The INDEX project Final report
97
0
20
40
60
80
100
120
140
Incence
burning
New buildings In a car Room with
ETS
Train with
ETS
Concentration (µg/m
3
)
Benzene
Figure 3.5. Short time benzene concentrations related to specific microenvironments or emission sources. Sampling
times of ‘New buildings’ (Brown 2002), ‘In a car’, ‘Room with ETS’, and ‘Train with ETS’ (Elke at al 1998) were 30-
50 min, 30-min, 60-min, 30-min respectively. During incense burning grab samples were obtained (Lee and Wang
2004).
4. Toxicokinetics
Absorption
Benzene is readily absorbed from oral and inhalation exposures. Less than 1% is absorbed through the skin. Pekari et al.
(1992) studied respiratory absorption of benzene in three males exposed to 5 and 32 mg/m
3
(1.7 and 10 ppm) benzene
for 4 hours each. Absorption of benzene was determined by measuring differences in concentration between inhaled and
exhaled air. The average absorption was 52% at the low concentration and 48% at the high concentration.
Most human respiratory absorption studies were conducted at concentrations probably conducing to saturation of the
benzene metabolism. The observed decline in absorption with increasing exposure time is apparently due to respiratory
excretion of unmetabolized benzene. Animal studies indicate that the metabolism of benzene is saturated at exposure
concentrations in excess of 32 mg/m
3
. Because absorption at low dose levels is most relevant for derivation of exposure
limits, the estimates of 48–52% retention derived by Pekari et al. (1992) are the most relevant values for evaluating
absorption in humans. Absorption of benzene through the oral route is complete (100%).
Distribution
Results from test animal studies indicate that absorbed benzene is widely distributed throughout the body,
independently of the route of the route of exposure. The parent compound is preferentially stored in the fat, although the
relative uptake in tissues also appears to be dependent on the perfusion rate of tissues by blood. Steady-state benzene
concentrations in rats exposed via inhalation to 1600 mg/m3 (500 ppm) for 6 hours were blood, 1.2 mg%; bone marrow,
3.8 mg%; and fat, 16.4 mg% (Rickert et al., 1979). Benzene was also found in the kidney, lung, liver, brain, and spleen.
Levels of the benzene metabolites phenol, catechol, and hydroquinone were higher in the bone marrow than in blood,
with phenol being eliminated more rapidly than catechol or hydroquinone after exposure. Benzene also has been shown
to cross the human placenta and has been found in the cord blood in amounts equal to or greater than those in maternal
blood (Dowty et al. 1976).
Metabolism and elimination
Despite extensive research, the metabolism of benzene is still not thoroughly understood. It is generally accepted that
benzene itself is not directly responsible for causing the toxic effects; however, the metabolic products responsible for
the noncancer and carcinogenic effects of benzene exposure have not been clearly defined. The evidence suggests that
The INDEX project Final report
98
several metabolites, as well as interactions between these metabolites, may be necessary to explain the toxic effects of
benzene.
Reviews of benzene metabolism are given in ATSDR, 1997; Snyder et al., 1993b; Snyder and Hedli, 1996; Ross, 1996;
Witz et al.,1996 (see also Figure 4.1). The liver is the major site of benzene metabolism. Benzene is converted to a
number of metabolites (phenol, catechol, hydroquinone, 1,2,4-benzenetriol, benzoquinone and trans,trans-
muconaldehyde) by the cytochrome P-450 dependent mixed-function oxidase system. The P-450 enzyme CYP 2E1
appears to exhibit the greatest affinity for benzene and is the most active in benzene metabolism. CYP 2B1 is also
capable of hydroxylating benzene, but contributes to the metabolism only at higher concentrations. Oxidative
metabolism by CYP 2E1 is required for manifestation of the haematotoxic and genotoxic effects of benzene as
confirmed recently by studies on the benzene in vivo metabolism in transgenic CYP 2E1 knockout mice (Valentine et
al. 1996).
In the attempt to explain bone marrow toxicity the hypothesis is brought forward that phenol, catechol, and
hydroquinone are transported to the bone marrow and converted by peroxidase-mediated reactions to electrophilic
compounds. For example, in the bone marrow, phenol is rapidly oxidized by the marrow peroxidases to reactive,
protein-binding species such as biphenols and biphenoquinones (Sawahata et al. 1985). Hydroquinone also is oxidized
in the marrow via a peroxidase-catalyzed reaction to an extremely short-lived semiquinone radical intermediate. This
forms p-benzoquinone, a reactive metabolite, known to cause DNA damage and cytotoxic effects in benzene-exposed
animals (Smith et al. 1989). However, trans-trans-muconaldehyde may also play a role (Snyder et al. 1989).
Pharmacokinetic models have been developed for rats and mice and used for risk assessments based on animal cancer
data (WHO, 1993; Cox and Ricci, 1992). Physiologically based toxicokinetic models have also been developed using
human data (WHO, 1993; Watanabe et al., 1994), but so far these have not been used for risk assessment.
The metabolism of benzene can be inhibited by toluene, leading to decreased toxicity. On the other hand, ethanol can
increase the metabolism of benzene, primarily by inducing xenobiotic metabolizing enzymes (Medinsky at al., 1994).
The average half-time of benzene in humans is 28 hours (WHO, 1987).
Following inhalation exposure to benzene, the major route of elimination of unmetabolized benzene in humans and
animals is in exhaled air. Most benzene is metabolized and the metabolites are excreted after phase-II-conjugation
predominantly in the urine. Phenolic metabolites are conjugated with sulfate or glucuronic acid (Henderson et al. 1989).
Small amounts of the glucuronides may enter the bile and are found in the feces.
5. Health effects
Effects of short-term exposure
The acute toxicity of benzene is low. In general, acute symptoms are dependent on both the concentration and duration
of exposure. Exposure to 24 g/m
3
for 30 min is life-threatening; 5 g/m
3
for 60 min produces significant symptoms; 150-
500 mg/m
3
for 5 hr results in headache and weakness; whereas exposure to 80 mg/m
3
or less for 8 hr results in no
demonstrable acute effect (Irons, 1992). Exposure at the odor threshold (2.8 mg/m³) for a brief duration has been
reported to enhance the electropotential of the brain (Haley, 1977).
Major concerns of systemic benzene toxicity include aplastic anemia and acute myelogenous leukemia (IARC, 1987;
Reprotext, 1993). Both of these conditions are typically seen in the chronic and subchronic exposures, but may be of
concern following acute exposures as well. Myeloid and erythroid components of the bone-marrow are specific targets
of benzene toxicity, which leads to aplastic anemia (IARC, 1982).
The long elimination half-life for benzene (13.1 hours for the longer phase of a 2-compartment model) is due to the
formation of catechol, quinone, and hydroquinone in the bone marrow. These reactive metabolites are not readily
excreted, and are cytotoxic to the germinal cells in the bone marrow (Greenlee et al., 1981). A 3-compartment model
was fitted to human data on benzene disposition and bonemarrow metabolism (Watanabe et al., 1994). The general
relationship between cumulative quantity of metabolites produced and inhalation concentration was not linear, but was
S-shaped, inflecting upward at low concentrations, and saturating at high concentrations.
The INDEX project Final report
99
Figure 4.1: Benzene Metabolism (taken from OEHHA, 2001)
Male C57BL/6J mice (7-Wgroup) were exposed to benzene 0, 33, 99, 319 or 961 mg/m
3
(0, 10.2, 31, 100 or 301 ppm)
in wholebody dynamic inhalation chambers for 6 hours/day for 6 consecutive days (Rozen et al. 1984). Control mice
were exposed to filtered, conditioned air only. Erythrocyte counts were depressed in C57BL/6 mice only at 319 and 961
mg/m
3
. At exposures of 33 and 99 mg/m
3
, respectively, depressions of the proliferative responses of bone-marrow-
derived B-cells and splenic T-cells occurred in C57BL/6J mice without causing a concomitant depression in number of
splenic T- or B- cells. Peripheral lymphocyte counts were depressed at all levels. No correlation was made of reduced
lymphocytes with general effects. These results demonstrate that short-term inhaled benzene even at low exposure
concentrations can cause reduction in certain immune associated processes.
Results from subacute exposures further illustrate the hematotoxic effects of benzene and the potential for
immunotoxicity. Inhalation of 334 mg/m³ (103 ppm) benzene for 6 hours/day for 7 days by mice caused decreased
spleen and marrow cellularities and decreased spleen weights (Green et al., 1981). Benzene inhalation at concentrations
of 0, 32, 96, 319, and 958 mg/m³ (0, 10, 30, 100, and 300 ppm) for 6 hours/day for 5 days resulted in a decreased host-
resistance to bacterial infection by Lysteria monocytogenes (Rosenthal and Snyder, 1985). The numbers of L.
monocytogenes bacteria isolated from the spleen were increased in a dose-dependent manner on day 4 of infection. The
total numbers of T- and B-lymphocytes in the spleen and the proliferative ability of the splenic lymphocytes were
decreased in a dose-dependent manner by benzene exposures of 96 mg/m³ or greater. In this study, no decrement in host
resistance or immune response was observed at 32 mg/m³ benzene. Later studies in mice have also shown that exposure
to 32 mg/m³ for a subacute duration does not significantly alter hematological parameters in blood, spleen, thymus, or
bone marrow (Farris et al., 1996; 1997). Farris et al. (1997) reported the hematological consequences of benzene
inhalation in B6C3F1 mice exposed to 3.2 , 16, 32, 319 and 639 mg/m³ (1, 5, 10, 100, and 200 ppm) benzene for 6
hr/day, 5 days/week for 1, 2, 4, or 8 weeks and a recovery group. There were no significant effects on hematopoietic
parameters from exposure to 10 ppm benzene or less. Thus 32 mg/m³ was a NOAEL for 1 week of exposure (and
longer). Exposure to 319 and 639 mg/m³ benzene reduced the number of total bone marrow cells, progenitor cells,
differentiating hematopoietic cells, and most blood parameters. Replication of primitive progenitor cells in the bone
marrow was increased during the exposure period as a compensation for the cytotoxicity. At 639 mg/m³, the primitive
progenitor cells maintained an increased percentage of cells in S-phase through 25 days of recovery compared with
controls.
The INDEX project Final report
100
In addition to myelotoxicity, acute exposure to benzene may disrupt erythropoiesis and result in genotoxicity.
Erythropoiesis, as measured by uptake of radiolabeled iron in the bone-marrow, has been shown to be inhibited by
subcutaneous injection of 10 mmol/kg benzene in mice (Bolcsak and Nerland, 1983). Inhalation of 9.6 mg/m³ benzene
for 6 hours by rats resulted in a significant increase over controls in the frequency of sister chromatid exchanges in
peripheral blood lymphocytes (Erexson et al., 1986).
Reproductive and developmental effects have been reported following benzene exposure. Coate et al. (1984) exposed
groups of 40 female rats to 3.2 , 32, 128, 319 and 639 mg/m³ (0, 1, 10, 40, and 100 ppm) benzene for 6 hours/day
during days 6-15 of gestation. In this study, teratologic evaluations and fetotoxic measurements were done on the
fetuses. A significant decrease was noted in the body weights of fetuses from dams exposed to 319 mg/m³. No effects
were observed at a concentration of 128 mg/m³.
Keller and Snyder (1986) reported that exposure of pregnant mice to concentrations as low as 16 mg/m³ (5 ppm)
benzene on days 6-15 of gestation (6 hr/day) resulted in bone-marrow hematopoietic changes in the offspring that
persisted into adulthood. However, the hematopoietic effects (e.g., bimodal changes in erythroid colony-forming cells)
in the above study were of uncertain clinical significance.
Keller and Snyder (1988) exposed Swiss Webster mice to benzene in utero from day six through day 15 of gestation.
Pregnant mice were administered benzene via inhalation at doses of 5, 10 or 20 ppm. On day 16 of gestation, two days
after birth and six weeks after birth, offspring were tested for levels of blood haemoglobin and levels of hematopoietic
cells in the blood, bone marrow and liver. No adverse effects were observed for fetuses at day 16 of gestation. However,
two days after birth, offspring exhibited reduced numbers of circulating erythroid precursor cells (all doses), increased
numbers of hematopoietic blast cells in the liver (20 ppm only), increased granulopoietic precursor cells (20 ppm only)
and decreased numbers of erythropoietic precursor cells (20 ppm only). Mice examined six weeks after birth also
demonstrated increased granulopoiesis in the 20 ppm dose group.
An exposure of 1600 mg/m³ (500 ppm) benzene through days 6-15 of gestation was teratogenic in rats while 160 mg/
resulted in reduced fetal weights on day 20 of gestation. No fetal effects were noted at an exposure of 32 mg/m³ (Kuna
and Kapp, 1981). An earlier study by Murray et al. (1979) showed that inhalation of 1600 mg/m³ benzene for 7
hours/day on days 6-15 and days 6-18 of gestation in mice and rabbits, respectively, induced minor skeletal variations.
Tatrai et al. (1980) demonstrated decreased fetal body weights and elevated liver weights in rats exposed throughout
gestation to 150 mg/m³ (47 ppm).
A recent study did not find any increased risk of spontaneous abortion among the wives of 823 male workers
occupationally exposed to benzene levels of less than or around 15 mg/m
3
(Strucker et al., 1994).
An attempt by Nielsen and Alarie (1982) to determine the inhalation RD
50
(the concentration of a compound inducing a
40% decrease in respiratory rate in male Swiss-Webster mice) for benzene was not successful. These investigators
showed that inhalation of 18,5 g/m³ (5800 ppm) benzene in mice caused an increase in respiratory rate beginning at 5
minutes following onset of exposure. They speculated that the stimulation of respiratory rate resulted from the action of
benzene on the central nervous system. In this study, benzene was not irritating to the upper airways of the animals.
Summary of short-term exposure effect levels (noncancer)
NOAEL
mg/m³
LOAEL
mg/m³
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
128
EXP
319
EXP
Developmental: decreased fetal body weights
(severe adverse effect!)
Rats, 6 hours per day
during days 5-16 of
gestation
Coate et al., 1984;
(supported by Kuna
and Kapp, 1981;
Keller and Snyder,
1988)
OEHHA 1999
REL: 1.3
16
EXP
Developmental: Altered hematopoiesis in
offspring
(high degree of uncertainty!)
Mice, 6 hours per day
during days 5-16 of
gestation
Keller and Snyder,
1988
OEHHA 2001
MADL
inhal.
: 1.3
µg/day
33
EXP
8
ADJ
47
HEC
Haematological: depressed peripheral
lymphocytes and mitogen-induced blastogenesis
of femoral B-lymphocytes
(less Serious LOAEL) .
Mice, 6 hours per day
(for 6 consecutive
days)
Rozen et al. 1984
ATSDR 1997
MRL: 0.16
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
The INDEX project Final report
101
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration; MADL
inhal.
maximum allowable daily level (OEHHA)
OEHHA (1999)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Derivation of Acute Reference Exposure Level (for a 6-hour exposure) (Level Protective Against Severe Adverse
Effects):
Study Coate et al., 1984; (supported by Kuna and Kapp, 1981; Keller
and Snyder, 1988)
Study population pregnant female rats
Exposure method inhalation of 0, 1, 10, 40, or 100 ppm
Critical effects decreased fetal body weights
LOAEL 100 ppm
NOAEL 40 ppm
Exposure duration 6 hours per day (for 5 days)
LOAEL uncertainty factor 1
Interspecies uncertainty factor 10
Intraspecies uncertainty factor 10
Cumulative uncertainty factor 100
Reference Exposure Level 0.4 ppm (1.3 mg/m³; 1,300 µg/m³)
While benzene exposure results in decreased immune response and hematopoietic effects in laboratory animals
following 5 day exposures, it was problematic to extrapolate from these repeated dose studies for these endpoints. Thus,
no level protective against mild adverse effects for one-hour is being recommended. The REL is based on
developmental toxicity, a severe adverse effect.
Pregnant female rats (40 per group) were exposed for 6 hours/day on days 6-15 of gestation to benzene concentrations
of 0, 1, 10, 40, and 100 ppm (0, 3.24, 32.4, 129.6, and 324 mg/m³) (Coate et al., 1984). The mean fetal weights from the
females treated with 100 ppm benzene were significantly decreased (p < 0.05) compared to controls. No teratogenic,
fetotoxic, or maternally toxic effects were observed in rats exposed to 40 ppm (129.6 mg/m³) benzene or less. The 40
ppm (129.6 mg/m³) concentration is considered a NOAEL for reduced fetal weight. The value of 40 ppm for a 6-hour
exposure was extrapolated to a 1-hour exposure using the equation Cn * T = k, where n = 2. The resulting 100 ppm
extrapolated value was used to determine the level protective against severe adverse effects using uncertainty factors of
10 for intraspecies and 10 for interspecies variation. The level protective against severe adverse effects for benzene is
therefore 1.0 ppm or 3.24 mg/m³.
Kuna and Kapp (1981) found direct teratogenic effects measured as decreased crown-rump length, exencephaly, and
angulated ribs in rats when pregnant females were exposed 6 hours/day during days 6-15 of gestation to a concentration
of 500 ppm. In this study, a concentration of 50 ppm during gestation resulted in lower fetal weights measured on day
20 of gestation. No fetal effects were noted at an exposure of 10 ppm (32 mg/m³). Keller and Snyder (1988) reported a
NOAEL of 10 ppm for developmental hematopoietic effects in mice. The highest reported NOAEL (i.e., 40 ppm)
consistent with reported LOAEL values was chosen for the derivation of the Reference Exposure Level (severe adverse
effect level, in this case) for benzene.
OEHHA (2001)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
From all the available animal studies the most sensitive study was a developmental toxicity study in mice using
endpoints reflecting altered hematopoiesis (blood cell formation) (Keller and Snyder, 1988). Exposures were by
inhalation at 0, 5, 10 and 20 ppm benzene concentrations in air, 6 h/day, during gestation days 6 to 15. The LOEL for
developmental hematopoietic effects was 5 ppm based on a significantly reduced relative number of early nucleated red
cells in a sample of nucleated cells from the peripheral blood of 2-day old neonates that had been exposed to benzene in
utero. Because the Keller and Snyder study provided a LOEL rather than a NOEL, a NOEL for purposes of assessment
was calculated by dividing the LOEL of 5 ppm by 10, resulting in a NOEL of 0.5 ppm.
The following calculations were performed to derive the Maximum Allowable Daily Level (MADL) for benzene:
Conversion of air concentration in ppm to mg/m
3
using a molecular weight for benzene of 78.11 daltons and a partial
molar volume of 24.45 at 25°C (=1.6 mg/m
3
). Conversion of air concentration for 6 hour (h) exposure to a 24 h day
The INDEX project Final report
102
(=0.40 mg/m
3
). Calculation of NOEL dose for 30 g mouse with an inhalation rate of 0.063 m
3
/day (Bond et al. 1986,
Depledge 1985) (=0.84 mg/kg/day). Calculation of NOEL dose for a 58 kg woman (=49 mg/day).
The MADL is derived by dividing the NOEL by one thousand. Thus, the adjusted NOEL was divided by 1000 to obtain
the MADL
inhalation
= 49 µg/day
ATSDR (1997)
Agency for Toxic Substances and Disease Registry
Derivation of the Minimal Risk Level (MRL):
ATSDR derived an acute-duration inhalation MRL of 0.05 ppm for benzene based on a LOAEL of 10 ppm for
immunotoxicity (reduced lymphocyte proliferation) following mitogen stimulation in mice (Rozen et al. 1984). The
animal LOAEL was converted to a human equivalent concentration (LOAEL
HEC
) of 14.7 ppm and divided by an
uncertainty factor of 300 (10 for the use of a LOAEL, 3 for extrapolation from animals to humans, and 10 for human
variability) to yield the MRL. The mice were exposed 6 hours/day for 6 days.
Effects noted in the study (Rozen et al. 1984) and corresponding doses: 10.2 ppm: No adverse effect on erythrocytes,
depressed peripheral lymphocytes and mitogen-induced blastogenesis of femoral B-lymphocytes (Less Serious
LOAEL). 31 ppm: No adverse effect on erythrocytes, depression of mitogen-induced blastogenesis of splenic T-cells.
100 ppm: Depressed erythrocyte counts. Dose end point used for MRL derivation: 10.2 ppm. Uncertainty factors used
in MRL derivation: 300, 10 for use of a LOAEL, 3 for extrapolation from animals to humans, 10 for human variability.
The ratio of the blood/gas partition coefficients was assumed to be 1. The concentration was adjusted for intermittent
exposure by multiplying the LOAEL (10.2 ppm) by 6/24 to correct for less than a full day of exposure. The resulting
number (adjusted LOAEL) is 2.50 ppm.
The human equivalent concentration (HEC) was calculated using Formula 4-l1, from Interim Methods for Development
of Inhalation Reference Concentrations, EPA 1990h:
LOAEL
[HEC]
(mg/m
3
or ppm) = LOAEL [ADJ] (mg/m
3
or ppm) x (V
A
/BW)
A
/ (V
A
/BW)
H
where: LOAEL
[HEC]
= the LOAEL human equivalent concentration
LOAEL
[ADJ]
= the LOAEL adjusted for duration (see above)
(V
A
/BW)
A
/ (V
A
/BW)
H
= the ratio of the alveolar ventilation rate (mL/min or L/hr) divided by Body weight (kg) of the
animal species to the same parameters for humans.
Values for this ratio were taken from EPA 1988e, as follows: (V
A
)
A
= ventilation rate for male B6C3F1 mice,
subchronic, 0.053 m
3
/day x 1,000 L/m
3
x 1 day/24 hr= 2.208 L/hr = 2.21 L/hr. (BW)
A
= body weight for male B6C3F1
mice, subchronic, 0.0316 kg (V
A
)
H
= ventilation rate for human adult male, 20 m
3
/day x 1,000 L/m
3
x 1 day/24 hr=
833.33 L/hr = 833 L/hr. (BW)
H
= body weight for human adult male, 70 kg. Resulting in a LOAEL
[HEC]
given by: 2.50
ppm x (2.21 L/hr / 0.0316 kg ) / (833 L/hr / 70 kg ) = 14.7 ppm (47 mg/m
3
).
Other additional studies or pertinent information that lend support to this MRL: Increased number of MN-PCEs,
decreased numbers of granulopoietic stem cells (Toft et al. 1982) lymphopenia (Cronkite et al. 1985), lymphocyte
depression, and increased susceptibility to bacterial infection (Rosenthal and Snyder 198.5) are among the adverse
hematological and immunological effects observed in several other acute-duration inhalation studies. The study by
Rozen et al. (1984) shows benzene immunotoxicity (reduced mitogen-induced lymphocyte proliferation) at a slightly
lower exposure level than these other studies. C57BI/6J mice were exposed to 0, 10.2, 31, 100, and 301 ppm benzene
for 6 days at 6 hrs/day. Control mice were exposed to filtered, conditioned air only. Lymphocyte counts were depressed
at all exposure levels while erythrocyte counts were elevated at 10.2 ppm, equal to controls at 31 ppm and depressed at
100 and 301 ppm. Femoral B-lymphocyte and splenic B-lymphocyte numbers were reduced at 100 ppm. Levels of
circulating lymphocytes and mitogeninduced blastogenesis of femoral B-lymphocytes were depressed after exposure to
10.2 ppm benzene for 6 days. Mitogen-induced blastogeneses of splenic T-lymphocytes were depressed after exposure
to 31 ppm of benzene for 6 days. In another study, mice exhibited a 50% decrease in the population of erythroid
progenitor cells (CFU-E) after exposure to 10 ppm benzene for 5 days, 6 hours per day (Dempster and Snyder 1991). In
a study by Wells and Nerland (1991), groups of 4-5 male Swiss-Webster mice were exposed to 0, 3, 25, 55, 105, 199,
303, 527, 1,150, or 2,290 ppm benzene for 6 hours/day for 5 days. The number of leukocytes in peripheral blood and
spleen weights were significantly decreased compared with untreated controls at all concentrations ¡Ý25 ppm.
Therefore, 3 ppm was the NOAEL and 25 ppm was the LOAEL for these effects. Other endpoints were not monitored
in this study. These data support the choice of Rozen et al. (1984) as a critical study.
The INDEX project Final report
103
Effects of long-term exposure (noncancer)
The primary toxicological effects of chronic benzene exposure are on the hematopoeitic system. Neurological and
reproductive/developmental toxic effects are also of concern at slightly higher concentrations. Impairment of immune
function and/or various anemias may result from the hematotoxicity. The hematologic lesions in the bone marrow can
lead to peripheral lymphocytopenia and/or pancytopenia following chronic exposure. Severe benzene exposures can
also lead to life-threatening aplastic anemia. These lesions may lead to the development of leukemia years after
apparent recovery from the hematologic damage (DeGowin, 1963).
Several types of blood dyscrasia, including pancytopenia, aplastic anaemia, thrombocytopenia, granulocytopenia and
lymphocytopenia have been noted following exposure to benzene. As in experimental animals, the primary target organ
for benzene that results in haematological changes is the bone marrow. It has been suggested that the cells at highest
risk are the rapidly proliferating stem cells (WHO, 1993). Most of these cases were reported several years ago, when
benzene was used as a solvent in different workplaces. An increased frequency of anaemia was detected among shoe
workers, rotogravure workers and rubber factory workers with prolonged exposure to high benzene concentrations
(hundreds of milligrams of benzene per m
3
) (WHO, 1993).
Kipen et al. (1988) performed a retrospective longitudinal study on a cohort of 459 rubber workers, examining the
correlation of average benzene exposure with total white blood cell counts taken from the workers. These researchers
found a significant (p < 0.016) negative correlation between average benzene concentrations in the workplace and white
blood cell counts in workers from the years 1940-1948. A reanalysis of these data by Cody et al. (1993) showed
significant decreases in RBC and WBC counts among a group of 161 workers during the 1946-1949 period compared
with their pre-exposure blood cell counts. The decline in blood counts was measured over the course of 12 months
following start of exposure. During the course of employment, workers who had low monthly blood cell counts were
transferred to other areas with lower benzene exposures, thus potentially creating a bias towards non-significance or
removing sensitive subjects from the study population. Since there was a reported 75% rate of job change within the
first year of employment, this bias could be highly significant. In addition, there was some indication of blood
transfusions used to treat some “anemic” workers, which would cause serious problems in interpreting the RBC data,
since RBCs have a long lifespan in the bloodstream. The exposure analysis in this study was performed by Crump and
Allen (1984). The range of monthly median exposures was 30-54 ppm throughout the 12-month segment examined.
Despite the above-mentioned potential biases, workers exposed above the median concentrations displayed significantly
decreased WBC and RBC counts compared with workers exposed to the lower concentrations using a repeated
measures analysis of variance.
Tsai et al. (1983) examined the mortality from all cancers and leukemia, in addition to hematologic parameters in male
workers exposed to benzene for 1-21 years in a refinery from 1952-1978. The cohort of 454 included maintenance
workers and utility men and laborers assigned to benzene units on a “regular basis”. Exposures to benzene were
determined using personal monitors; the median air concentration was 0.53 ppm in the work areas of greatest exposure
to benzene. The average length of employment in the cohort was 7.4 years. The analysis of overall mortality in this
population revealed no significant excesses. Mortality from all causes and from diseases of the circulatory system was
significantly below expected values based on comparable groups of U.S. males. The authors concluded the presence of
a healthy worker effect. An internal comparison group of 823 people, including 10% of the workers who were
employed in the same plant in operations not related to benzene, showed relative risks for 0.90 and 1.31 for all causes
and cancer at all sites, respectively (p < 0.28 and 0.23). A subset of 303 workers was followed for medical surveillance.
Up to four hematological tests per year were conducted on these workers. Total and differential white blood cell counts,
haemoglobin, hematocrit, red blood cells, platelets and clotting times were found to be within normal (between 5% and
95% percentile) limits in this group. Although the exposure duration averaged only 7.4 years, the study was considered
to be chronic since 32% of the workers had been exposed for more than 10 years.
Collins et al. (1997) used routine data from Monsanto’s medical/industrial hygiene system to study 387 workers with
daily 8-hour time-weighted exposures (TWA) averaging 0.55 ppm benzene (range = 0.01 – 87.69 ppm; based on 4213
personal monitoring samples, less than 5% of which exceeded 2 ppm). Controls were 553 unexposed workers. There
was no increase in the prevalence of lymphopenia, an early, sensitive indicator of benzene toxicity, among exposed
workers (odds ratio = 0.6; 95% confidence interval = 0.2 to 1.8), taking into account smoking, age, and sex. There also
was no increase in risk among workers exposed 5 or more years (odds ratio = 0.6; 95% confidence interval = 0.2 to 1.9).
There were no differences between exposed and unexposed workers for other measures of hematotoxicity, including
mean corpuscular volume and counts of total white blood cells, red blood cells, haemoglobin, and platelets.
Rothman et al. (1996) compared hematologic outcomes in a cross-sectional study of 44 male and female workers
heavily exposed to benzene (median = 31 ppm as an 8-hr TWA) and 44 age and gender-matched unexposed controls
from China. Hematologic parameters (total WBC, absolute lymphocyte count, platelets, red blood cells, and hematocrit)
The INDEX project Final report
104
were decreased among exposed workers compared to controls; an exception was the red blood cell mean corpuscular
volume (MCV), which was higher among exposed subjects. In a subgroup of 11 workers with a median 8 hr TWA of
7.6 ppm (range = 1-20 ppm) and not exposed to more than 31 ppm on any of 5 sampling days, only the absolute
lymphocyte count (ALC) was significantly different between exposed workers and controls (p = 0.03). Among exposed
subjects, a dose response relationship with various measures of current benzene exposure (i.e., personal air monitoring,
benzene metabolites in urine) was present only for the total WBC count, the absolute lymphocyte count, and the MCV.
Their results support the use of the absolute lymphocyte count as the most sensitive indicator of benzene-induced
hematotoxicity.
Ward et al. (1985) exposed male and female CD-1 mice and Sprague-Dawley rats to 0, 1, 10, 30 or 300 ppm (0, 3.2, 32,
96 or 960 mg/m3) benzene, 6 hours/day, 5 days/week for 91 days and measured various hematological endpoints. The
study identified both a LOAEL of 300 ppm and a NOAEL of 30 ppm. The male mouse appeared to be the most
sensitive sex/species in this study. The exposure-response relationships for the different hematological endpoints for the
male mouse were modeled using a BMD modeling approach and decreased hematocrit (i.e., volume percentage of
erythrocytes in whole blood) was chosen as the critical effect.
An examination of 32 patients, who were chronically exposed to benzene vapors ranging from 150 to 650 ppm for 4
months to 15 years, showed that pancytopenia occurred in 28 cases. Bone marrow punctures revealed variable
hematopoeitic lesions, ranging from acellularity to hypercellularity (Aksoy et al., 1972).
In an evaluation of the literature, a WHO Task Group (WHO, 1993) drew the conclusion that bone marrow depression
or anaemia would not be expected to occur in workers exposed for 10 years to 3.2 mg/m
3
(1 ppm) or less.
Intermediate-duration inhalation and oral exposures to benzene induced neurological effects in animals that included
reduced limb grip strength, behavioral disturbances, and changes in brain levels of monoamine transmitters and
acetylcholinesterase (Dempster et al. 1984; Frantik et al. 1994; Hsieh et al. 1988; Li et al. 1992).
Older studies on workers exposed to benzene, toluene and xylene have shown decreased levels of agglutinins and IgG
and IgA immunoglobulins, and increased levels of IgM.
A loss of leukocytes has been observed in several studies in highly exposed workers as well as reduced numbers of T
lymphocytes. In a study of refinery workers exposed to benzene concentrations of less than 32 mg/m
3
, no such
immunological effects were seen (WHO, 1993).
Summary of long-term exposure effect levels (noncancer)
NOAEL
mg/m³
LOAEL
mg/m³
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
1.7
Study
0.60
ADJ
Haematological effects (total and differential
white blood cell counts, haemoglobin,
hematocrit, red blood cells, platelets and clotting
times)
Occupational, 7.4y Tsai et al. 1983
OEHHA 1999
REL: 0.06
44
BMC
23
BMCL
8.2
ADJ-BMCL
Decreased lymphocyte counts Occupational, 6.3y
(0.7-16y)
Rothman et al. 1996 EPA-IRIS 2003
RfC: 0.03
(3.2) 32
EXP
Blood cell depression (lymphocytes) Mouse, 6-178d
NOAEC has been
derived, not observed
Dempster and Snyder,
1990, Rozen et al.
1984, Baarson et al.,
1984, Green et al.,
1981a,b, and other
studies
ECB 2003
2.5
EXP
1.05
HEC
Neurological: increased (rapid response time)
(Less Serious minimal LOAEL).
Mice, 30d
(2h/d, 6d/w)
Li et al. 1992
intermediate duration
study !
ATSDR 1997
MRL
INT
: 0.013
(intermediate!)
The INDEX project Final report
105
OEHHA (1999)
Office of Environmental Health Hazard Assessment
Derivation of Chronic Reference Exposure Level (REL)
Study Tsai et al. (1983)
Study population 303 Male refinery workers
Exposure method Occupational exposures for 1-21 years
Critical effects Hematological effects
LOAEL Not observed
NOAEL 0.53 ppm
Exposure continuity 8 hr/day (10 m
3 per 20 m3 day), 5 days/week
Exposure duration 7.4 years average (for the full cohort of 454); 32% of the workers were
exposed for more than 10 years
Average occupational exposure 0.19 ppm
Human equivalent concentration 0.19 ppm
LOAEL uncertainty factor 1
Subchronic uncertainty factor 1
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 10
Cumulative uncertainty factor 10
Inhalation reference exposure level 0.02 ppm (20 ppb; 0.06 mg/m
3; 60 µg/m3)
Both the animal and human databases for benzene are excellent. The human data presented by Tsai et al. (1983) and
associates were selected over animal studies because the collective human data were considered adequate in terms of
sample size, exposure duration, and health effects evaluation. Although the study by Tsai et al. (1983) is a free-standing
NOAEL, the endpoint examined is a known sensitive measure of benzene toxicity in humans. In addition, the LOAEL
for the same endpoint in workers reported by Cody et al. (1993) help form a dose-response relationship and also yield
an REL which is consistent with that derived from Tsai et al. (1983). The study by Cody et al. (1993), since it failed to
identify a NOAEL and was only for a period of 1 year, contained a greater degree of uncertainty in extrapolation to a
chronic community Reference Exposure Level. The recent results of Collins et al. (1997) that included a NOAEL of
0.55 ppm and of Rothman et al. (1996) that included a LOAEL of 7.6 ppm are consistent with those of Tsai et al.
Therefore the study by Tsai et al. (1983) was used as the basis for the chronic REL for benzene.
In the Cody et al. (1993) study, significant hematological effects, including reduced RBC and WBC counts, were
observed in 161 male rubber workers exposed to median peak concentrations (i.e., only the peak concentrations for any
given exposure time were reported) of 30-54 ppm or more for a 12-month period during 1948. The 30 ppm value was
considered a 1-year LOAEL for hematological effects. The 30 ppm value is the low end of the range of median values
(30-54 ppm) reported by Crump and used in the Kipen et al. (1988) and Cody et al. (1993) studies. An equivalent
continuous exposure of 10.7 ppm can be calculated by assuming that workers inhaled 10 m
3 of their total 20 m3 of air
per day during their work-shift, and by adjusting for a normal 5 day work week. Application of uncertainty factors for
subchronic exposures, estimation of a NOAEL, and for protection of sensitive subpopulations (10 for each) results in an
REL of 0.01 ppm (10 ppb; 30 µg/m
3). This is comparable to the REL based on Tsai et al. (1983).
Ward et al. (1996) determined a relationship between occupational exposures to benzene and decreased red and white
cell counts. A modeled dose-response relationship indicated a possibility for hematologic effects at concentrations
below 5 ppm. However, no specific measures of the actual effects at concentrations below 2 ppm were taken, and the
Tsai et al. (1983) data were not considered in their analysis. The purpose of this study was to characterize the trend for
effects at low concentrations of benzene. A NOAEL or LOAEL was not identified in the study. The selection of a
NOAEL of 0.53 ppm is therefore not inconsistent with the results of the Ward et al. (1996) study.
For comparison with the REL of 20 ppb based on human data, OEHHA estimated a REL based on the chronic
inhalation study in mice by Baarson et al. (1984), which showed that bone-marrow progenitor cells were markedly
suppressed after intermittent exposures (6 hr/day, 5 days/week) to 10 ppm benzene for 6 months. An extrapolation of
this value to an equivalent continuous exposure resulted in a concentration of 1.8 ppm. Application of an RGDR of 1
for a systemic effect and uncertainty factors of 3 and 10 for inter- and intraspecies variability, and 10 for estimation of a
NOAEL from the LOAEL would result in an REL of 6 ppb (20 µg/m
3). The Farris et al. (1997) 8 week study indicated
a LOAEL of 100 ppm and a NOAEL of 10 ppm for hematological effects. Application of an RGDR of 1 and UFs of 10
for subchronic, 3 for interspecies and 10 for intraspecies extrapolation (total UF = 300) also resulted in an estimated
REL of 6 ppb, in reasonable agreement with the proposed REL of 20 ppb. One could also crudely approximate an
inhalation REL from the oral NTP bioassay where a dose of 25 mg/kgday was associated with hematological effects.
The INDEX project Final report
106
The concentration approximately equivalent to a 25 mg/kg dose for a 70 kg human breathing 20 cubic meters per day is
27 ppm. Assuming this is a LOAEL and applying an RGDR of 1 for systemic effects, a 3 fold UF for extrapolation to
humans, a 10-fold UF for LOAEL to NOAEL extrapolation and a 10-fold UF for intraspecies extrapolation yields a
REL of 90 ppb. There are a number of uncertainties to this approach, yet it comes within a factor of 5 of the proposed
REL based on human studies.
IRIS (2003)
Integrated Risk Information System (IRIS) – U.S.EPA
Determination of the Reference Concentration for Chronic Inhalation Exposure (RfC)
Critical effect Exposures* UF MF RfC
Decreased lymphocyte count (Human
occupational inhalation study of
Rothman et al., 1996)
BMCL = 8.2 mg/m
3
300 1 0.03 mg/m
3
*Conversion factors: MW = 78.11. BMCL = 7.2 ppm, 8-hour TWA. Assuming 25ºC and 760 mm Hg, BMCL (mg/m
3
) = 7.2 ppm x MW/24.45 = 23.0
mg/m
3
. BMCL
ADJ
= 23.0 mg/m
3
x 10 m
3
/20 m
3
x 5 days/7days = 8.2 mg/m
3
. (The BMC was based on a benchmark response of one standard deviation
change from the control mean.)
The RfC is based on BMD modeling of the absolute lymphocyte count (ALC) data from the occupational epidemiologic
study of Rothman et al. (1996), in which workers were exposed to benzene by inhalation. A comparison analysis based
on BMD modeling of hematological data from the Ward et al. (1985) subchronic experimental animal inhalation study
was also conducted. In addition, comparison analyses using the LOAEL from the Rothman et al. (1996) study and the
NOAEL from the Ward et al. (1985) study were performed.
BMD modeling of the ALC exposure-response data from Rothman et al. (1996) was done using U.S. EPA's Benchmark
Dose Modeling Software (version 1.20). The data are rather supralinear, that is, the change in ALC per unit change in
exposure decreases with increasing exposure; therefore, in order to fit the data with one of the available continuous
models, the exposure levels were first transformed according to the equation d’ = ln(d+1). Then the exposure-response
data were fitted using the continuous linear model, which provided a good fit (p=0.54). A two-degree polynomial and a
power model also fit the data, but the linear model was selected because it is the most parsimonious. The parameters
were estimated using the method of maximum likelihood. A constant variance model was used.
In the absence of a clear definition for an adverse effect for this continuous endpoint, a default benchmark response of
one standard deviation change from the control mean was selected, as suggested in EPA's draft Benchmark Dose
Technical Guidance Document (U.S. EPA, 2000). This default definition of a benchmark response for continuous
endpoints corresponds to an excess risk of approximately 10% for the proportion of individuals below the 2nd
percentile (or above the 98th percentile) of the control distribution for normally distributed effects (see U.S. EPA,
2000). A 95% lower confidence limit (BMCL) on the resulting BMC was calculated using the likelihood profile
method. Transforming the results back to the original exposure scale yields a BMC of 13.7 ppm (8-hr TWA) and a
BMCL of 7.2 ppm (8-hr TWA).
As suggested in the draft technical guidance document (U.S. EPA, 2000), the BMCL is chosen as the point of departure
for the RfC derivation. An adjusted BMCL is calculated by converting ppm to mg/m3 and adjusting the 8-hour TWA
occupational exposure to an equivalent continuous environmental exposure. The BMCL is first converted to mg/m3
using the molecular weight of 78.11 for benzene and assuming 25ºC and 760 mm Hg: 7.2 ppm × 78.11/24.45 = 23.0
mg/m3. The converted value is then adjusted from the 8-hour occupational TWA to a continuous exposure
concentration using the default respiration rates (U.S. EPA, 1994): BMCLADJ = 23.0 mg/m3 × (10 m3/20 m3) × 5
days/7 days = 8.2 mg/m3.
The RfC is then derived by dividing the adjusted BMCL by the overall UF of 300: RfC = BMCLADJ/UF = 8.2 mg/m3
÷ 300 = 3 × 10-2 mg/m3. The overall UF of 300 comprises a UF of 3 for effect-level extrapolation, 10 for intraspecies
differences (human variability), 3 for subchronic-to-chronic extrapolation, and 3 for database deficiencies (see Section
I.B.3).
For comparison, an RfC was also calculated based on the LOAEL of 7.6 ppm (8-hr TWA) from the Rothman et al.
(1996) study. Converting the units and adjusting for continuous exposure as above results in a LOAEL
ADJ
of 8.7
mg/m3. The LOAEL
ADJ
is then divided by an overall UF of 1000 to obtain the RfC: 8.7 mg/m3 ÷ 1000 = 9 × 10-3
mg/m3. The combined UF of 1000 represents UFs of 10 to account for the use of a LOAEL because of the lack of an
appropriate NOAEL, 10 for intraspecies differences in response (human variability), 3 for subchronic-to-chronic
The INDEX project Final report
107
extrapolation, and 3 for database deficiencies. The value of 9 × 10-3 mg/m3 is in good agreement with the RfC of 3 ×
10-2 mg/m3 calculated from the BMC.
ATSDR (1997)
Agency for Toxic Substances and Disease Registry
Derivation of the Minimal Risk Level (MRL):
The intermediate-duration inhalation MRL of 0.004 ppm was derived from a LOAEL value of 0.78 ppm for
neurological effects of intermediate-duration inhalation exposure of mice to benzene (Li et al. 1992). The ratio of the
blood/gas partition coefficients was assumed to be 1. The dose was adjusted for intermittent exposure by multiplying
the LOAEL (0.78 ppm) by 2 hours/24 hours and by 6 days/7 days to adjust to continuous exposure. The resulting
adjusted LOAEL 0.056 ppm was then converted to a human equivalent concentration (HEC) by multiplying by the ratio
of the mouse ventilation rate (2.21 L/hour) to its body weight (0.0316 kg) divided by the ratio of the human ventilation
rate (833 L/hour) to human body weight (70 kg) (formula 4-l1, EPA 1990h; ventilation rate and body weight values
taken from EPA 1988e). The resulting LOAEL (HEC) of 0.33 ppm was then divided by an uncertainty factor of 90 (3
for the use of a minimal LOAEL, 3 for extrapolation from animals to humans after adjusting for the human equivalent
concentration, and 10 for human variability), yielding the MRL of 0.004 ppm (see Appendix A). ATSDR (1997) did not
derive a chronic-duration inhalation MRL for benzene due to lack of suitable data.
ECB (2003)
European Chemical Bureau - Institute for Health and Consumer Protection - European Union Risk Assessment Report
In its recent risk assessment draft report (2003) ECB concludes that chronic benzene exposure leads to depression of
white blood and red blood cells. This effect is reversible after long time exposures (years) with low concentrations
(reported concentration range: > 32-64 mg/m³ = 10-20 ppm). Exposure to 192 mg/m³ (60 ppm) of benzene for about a
week may be associated with an increased proportion of large granular lymphocytes, and not severe narrow effects nor
specific cytopenias. At higher concentrations, benzene may lead to aplastic anemia which can be fatal. Jandl's (1977)
review suggests a fatal outcome in 13% of the cases (as opposed to 85% for idiopathic aplastic anaemia).
The prevalence of leucopenia correlates with the concentration of benzene as shown by data of Yin et al. (1987) as well
as by Kipen et al. (1988, 1989). Taken these data, the LOAEC (lowest observed adverse effect concentration) for
leucopenia is in the range between 40 mg/m³ (12.5 ppm) and 64 mg/m³. A higher prevalence for leucopenia is given at
concentrations above 320 mg/m³ (100 ppm).
The case control studies presented recently by Rothman et al. (1996a, 1996b) and Dosemeci et al. (1997) have shown,
that the most sensitive reaction in humans to chronic benzene exposure is lymphopenia. The data show that a collective
of workers exposed to benzene concentrations in a range between 1 and 31 ppm had significantly reduced lymphocyte
counts as compared to a cohort of non-exposed workers.
Thus, for blood cell depression an overall LOAEC is suggested to be 32 mg/m³ (10 ppm). For depression of
lymphocytes, a NOAEC of 3.2 mg/m³ (1 ppm) has been derived. In conclusion, the NOAEC for effects of benzene is
assumed to be 3.2 mg (1 ppm).
Genotoxic effects
WHO (2000) Air Quality Guidelines for Europe 2000
There are numerous studies on chromosomal effects in exposed workers (2, 22). Both structural and numerical
chromosome aberrations have been observed. In most cases the benzene exposure has also been high enough to produce
haematological effects. The chromosomal effects in these studies are evident at concentrations of around 320 mg/m
3
(100 ppm) or higher, but in some studies effects were reported in workers chronically exposed to levels of around 32
mg/m
3
(10 ppm) (2, 23, 24). Tompa et al. (25) reported that the frequency of chromosome aberrations decreased when
exposure levels decreased from 3–69 mg/m
3
to 1–18 mg/m
3
. In the study by Karacic et al. (24), a decrease in sister
chromatid exchanges but not in chromosomal aberrations was noted in a group of female workers when examined with
a 5-year interval during which the mean benzene concentration had decreased from 26 to 16 mg/m
3
. Smoking did not
influence the results (23, 24, 26).
Somatic mutations as an endpoint of benzene-induced genotoxic effects in heavily exposed workers was studied
recently by Rothman et al. (27). They used the glycophorin A (GPA) mutation assay. The results suggested that benzene
induces gene-duplicating mutations, presumably through recombination mechanisms, but not gene-inactivating
mutations due to point mutations or deletions.
The INDEX project Final report
108
RIVM (2001)
National Institute of Public Health and the Environment - The Netherlands
The genotoxicity of benzene has been studied extensively. Benzene appears to have a peculiar genotoxic profile.
Benzene is hardly, if at all, able to induce gene mutations in in vitro test systems, despite its potent clastogenic
properties in vitro and in vivo. Several studies have demonstrated induction of both numerical and structural
chromosomal aberrations, sister chromatid exchanges, and micronuclei in experimental animals and humans after in
vivo benzene exposure (various routes of exposure). After short-term exposure to benzene levels of 3 to 30 mg/m
3
experimental animals showed chromosomal effects (Vermeirre et al. 1993) and some studies in humans have
demosnstrated chromosomal effects at mean workplace exposures as low as 4 to 7 mg/m
3
(EU 1999).
There appears to be a lack of evidence for direct DNA interaction under normal in vivo conditions: despite the
demonstrated ability of several benzene metabolites to form DNA adducts under in vitro conditions, DNA adduct
formation in bone marrow cells in experimental animals in vivo could only be demonstrated under quite specific and
very high exposure regimes (EU 1999; Health Council of the Netherlands 1997).
IRIS (2000)
Integrated Risk Information System (IRIS) – U.S.EPA
Current evidence indicates that benzene-induced myelotoxicity and genotoxicity result from a synergistic combination
of phenol with hydroquinone, muconaldehyde, or catechol. Molecular targets for the action of these metabolites,
whether acting alone or in concert, include tubulin, histone proteins, topoisomerase II, and other DNA-associated
proteins. Damage to these proteins would potentially cause DNA strand breakage, mitotic recombination, chromosomal
translocations, and malsegregation of chromosomes to produce aneuploidy. If these effects took place in stem or early
progenitor cells, a leukemic clone with selective advantage to grow could arise as a result of protooncogene activation,
gene fusion, and suppressor-gene inactivation. Epigenetic effects of benzene metabolites on the bone marrow stroma,
and perhaps the stem cells themselves, could then foster development and survival of a leukemic clone. Since these
plausible events have not been conclusively demonstrated, this remains a hypothesis (Smith, 1996).
UK Environment Agency (2003)
Benzene induces both numerical and structural chromosomal changes. Chromosome aberrations and micronuclei have
been commonly reported in laboratory animals exposed to benzene either in the atmosphere or by oral administration.
Often the positive results are seen throughout the tested dose range (ATSDR, 1997; WHO, 1993). Chromosomal
damage has been detected in the peripheral blood of workers by many teams of investigators (ATSDR, 1997), and
chromosomal effects have been claimed even at exposures of 4–7 mg m–3 (WHO, 2000).
Chromosomal aberrations may act as predictors of cancer risk, and their role in subsequent leukaemia is thought to
involve chromosome numbers 5 and 7, and also possibly numbers 8 and 21 (EBS, 1996; Irons and Stillman, 1996).
Aneuploidy may also be a precursor of leukaemogensis (EBS, 1996; Irons and Stillman, 1996). Other indicators of
genetic damage are sister chromatid exchange (SCE), DNA crosslinking, DNA adduct formation and DNA repair
(reviewed in Paustenbach et al, 1993; Snyder et al, 1993; EBS, 1996). Although benzene is not mutagenic in the Ames
test, there are a number of reports of treatment-related mutations in the tissues of mice exposed by inhalation (ATSDR,
1997).
Carcinogenic effects
Both IARC and the US EPA have assigned benzene their highest cancer classification, Group 1 (“is carcinogenic to
humans”) (IARC, 1987) and Category A (“known human carcinogen”) (USEPA, 2002), respectively. A large number of
epidemiological studies provide conclusive evidence of a causal association between benzene exposure and leukaemia,
acute non-lymphocytic leukaemia (ANLL) certainly, and also possibly chronic non-lymphocytic and chronic
lymphocytic leukaemias (USEPA, 2002). There have also been occasional reports of an increased risk in humans of
other neoplastic changes in the blood or lymphatics, including Hodgkin’s and non-Hodgkin’s lymphoma and
myelodysplastic syndrome (USEPA, 2002).
The vast majority of the evidence for an association with ANLL derives from occupational epidemiology, where
benzene is often a constituent of a complex mixture. These include studies in the shoe-making, printing, petrochemical,
chemical, coke production and rubber manufacturing industries (Aksoy et al, 1974; Christie et al, 1991; Decouflé et al,
1983; Hurley et al, 1991; Paxton et al, 1994a; Rushton, 1993; Tsai et al, 1983; Vigliani and Forni, 1976; Yin et al,
1989). Many of the workforces were exposed to benzene at levels markedly higher than those experienced in these
industries today.
The INDEX project Final report
109
There have been a number of cohort and nested case control studies that have estimated exposure for each worker.
Attempts have been made to develop dose–response relationships using cumulative exposure, which is, assuming a
symmetrical contribution of exposure concentration and exposure duration. The cancer mortality seen in the “Pliofilm”
cohort, which included workers from three factories manufacturing rubber film in the USA, has been most often used in
cancer risk assessment. A wide range of exposures were encountered, with relatively few other chemicals involved, and
a clear dose–response was seen between leukaemia risk and benzene exposure. There have been several publications
from this cohort giving mortality results (Infante et al, 1977; Paxton et al, 1994a; Rinsky et al, 1981, 1987; Wallace,
1996) and risk assessments using different methods of exposure estimation and different mathematical models (Austin
et al, 1988; Brett et al, 1989; Byrd and Barfield, 1989; Crump, 1994, 1996; Crump and Allen, 1984; Paustenbach et al,
1992; Paxton, 1996; Paxton et al, 1994b; Rinsky et al, 1989; Schnatter et al, 1996a,b). For example, the analysis of
Paxton et al (1994a), based on 14 cases of leukaemia in the exposed male workers and three previous estimates of
worker exposure (Crump and Allen, 1984; Paustenbach et al, 1992; Rinsky et al, 1981), reported standard mortality
ratios in the highest exposure group (>1600 mg m
–3
yr
–1
) of between 10.3 and 20.
Four other cohort studies have quantified benzene exposures for all workers. Bond et al (1986), in a follow-up of a
study by Ott et al (1978) of 956 Dow Chemical workers, found three deaths from leukaemia compared with 1.9
expected. Wong (1987a,b) followed up 7676 chemical workers, 3536 of whom were continuously exposed to benzene.
Six leukaemia deaths were reported in the exposed group compared with 4.5 expected. An update of a subgroup of this
population has been reported by Ireland et al (1997). There were three cases of leukaemia observed at exposures at or
above 6 ppm (19.2 mg m–3) compared with 0.65 expected.
Increased risks of non-haematopoietic cancers have been found in a few studies of occupational cohorts exposed to
benzene or mixtures containing benzene. These include bladder, stomach, prostate and lung neoplasms (ATSDR, 1997),
although, in general, the risks are only moderately raised; also, in many studies, exposure to multiple chemicals
complicates the interpretation of the results.
Benzene has been implicated as a potential risk factor for the development of childhood leukemia (OEHHA, 1997; Ries
et al., 1997; Smith and Zhang, 1998; U.S. EPA, 1998).
There are little data on the effects of direct exposure of children to benzene. Because of their small size, increased
activity, and increased ventilation rates compared to adults, children may have greater exposure to benzene in the air, on
a unit body weight basis (U.S. EPA, 1998). Although no human cancer studies of benzene-exposed children are
available, there is good reason to believe that childhood exposure to benzene would also contribute to adult-onset
leukemias. Also, there is some evidence to suggest that exposure to benzene is associated with childhood leukemia.
Paternal exposure to benzene prior to conception in humans has been associated in some studies with increased
childhood leukemia, especially of the acute nonlymphocytic type, findings that are supported by observations in animals
of benzene-induced DNA damage to sperm. Maternal exposure to benzene in humans also has been associated with
increased incidences of childhood leukemia. These findings are supported by observations in animals of benzene-
induced transplacental genotoxicity, altered hematopoiesis, and of carcinogenicity, following exposure in utero and
continuing until weaning. It should be noted that other epidemiological studies that represented a large number of cases
of various subtypes of leukemia (Kaatsch et al., 1998) or acute lymphocytic leukemia only (Shu et al., 1999) did not
find an association with paternal benzene exposure. Thus a casual relationship would be difficult to establish.
Summary of cancer (leukemia) risk values
Risk value Unit Risk specific concentration
µg/m
3
(at an excess lifetime
cancer risk of 10
-4
)
Risk value name Study Source; Ref.
Value mg/m³
6 · E-6
(µg/m
3
)
-1
17 Unit risk (UR) and guideline
value (GV)
Pliofilm cohort
Crump, 1994
WHO 1995
2.2 - 7.8 · E-6 (µg/m
3
)
-1
13 - 45 UR Pliofilm cohort
multiple: Rinsky et al., 1981, 1987;
Paustenbach et al., 1993; Crump and
Allen, 1984; Crump, 1992, 1994;
U.S. EPA, 1998.
US-EPA 1998
5 · E-8
a
-
6 · E-6
b
(µg/m
3
)
-1
20-36
UR
a
lowest plausible estimate
b
highest plausible estimate
multiple:
a
Wong and Raabe (1995)
b
Crump, 1994 (Pliofilm cohort)
EU 1999
1.5 · E+1 mg/m
3
30 TC
05
(Tumorigenic
concentration associated
with a 5% increase in
incidence) - acute
myelogenous leukemia
Pliofilm cohort
Rinsky et al., 1987
Health Canada
1991
The INDEX project Final report
110
2.4 · E-2 mg/m
3
24 CR
inhal.
(Lifetime inhalatory
excess cancer risk at the 1 in
10
4
level) - haematopoietic
system
multiple:
Wong and Raabe (1995)
RIVM 1999
0.054 (mg/kg
d)
-1
4.1 using mean children’s
breathing rate of 0.45m
3
/kg-
day;
7.9 using mean adult
breathing rate of
0.235m
3
/kg-day
Inhalation cancer potency Pliofilm Cohort: Rinsky et al., 1987;
Paxton et al., 1994
Chinese Worker Cohort: Hayes et al.,
1997
OEHHA 2003
2 · E-5 (ppm)
-1
16 Lifetime leukemia (AML)
risk equivalent
Pliofilm cohort
multiple
Crump, 1994
USEPA, 1998
NICNAS 2001
WHO (1995)
Air Quality Guidelines for Europe 2000
The WHO derived in 1995 a Unit risk (UR) for leukemia of 6 · 10
–6
(µg/m
3
)
-1
on the basis of risk calculations of Crump
(1994). Based on this UR, air quality guidelines corresponding to excess lifetime cancer risks of 10
-4
, 10
-5
and 10
-6
are
17, 1.7 and 0.17 µg/m
3
, respectively (WHO, 2000)
US EPA (1998)
The US EPA is currently in the process of reviewing the evidence relating to the risk of benzene. Their provisional
conclusion is that there is insufficient evidence to reject a linear dose-response curve for benzene carcinogenicity at
low-dose exposures and that the approach of using a linear dose-response curve is still to be recommended. The US
EPA gives a range of risk estimate for leukemia at 1 µg/m
3
from 2.2 10
–6
to 7.8 10
–6
.
European Commission (1999)
The EU-Working Group concluded that it is reasonable to assume that benzene-induced effects at low exposure levels
differ quantitatively as well as qualitatively from those induced at high occupational exposures. It is not clear whether
the underlying mechanism of benzene carcinogenicity is direct DNA interaction. In view of this uncertainty the EU
Working Group decided, on precautionary grounds, to hold on to a non-threshold extrapolation method. Though it was
not possible to give a precise estimate of the risk associated with benzene it was possible to define a range within which
that risk was likely to lie. The UR calculated by the WHO (6 · 10
–6
) was considered to result in the highest plausible
estimate of risk. The lowest UR which the EU Working Group felt was likely to be plausible was in the order of 5 · 10
8
, an estimate which is consistent with the Dutch Health Council’s analysis of the Wong and Raabe data. This range of
URs means that an excess lifetime risk for leukemia of 10
–6
is associated with annual average concentrations varying
between 0.17 and 20 µg/m
3
(EU 1999).
On the basis of this risk estimation the European Commission has adopted an ambient air quality limit value for
benzene of 5 µg/m
3
to be met on 1 January 2010 (Directive 2000/69/EC). This limit value has been proposed by the EU
working group taking into account practical considerations; 5 µg/m
3
is as low as reasonably achievable in 2010.
Health Canada (1991)
Health Canada based their risk values on the incidence of leukemia in occupationally-exposed humans, resulting in a
TC
05
(Tumorigenic concentration associated with a 5% increase in incidence) of 1.5E+1 mg/m
3
. The Health Canada
TC
05
can be divided by 5000 to represent a 1 in 100,000 risk level of 3 · 10
–3
mg/m
3
. It should be noted that the Health
Canada TC
05
is computed directly from the dose-response curve within or close to the experimental range.
RIVM (1997)
National Institute of Public Health and the Environment - The Netherlands
The figure of 24 µg/m
3
(risk specific concentration at an excess lifetime cancer risk of 10
-4
) was derived from the
estimate as presented by the Health Council of 12 µg/m
3
being associated with an excess lifetime cancer risk of 10
-6
.
The figure of 12 µg/m
3
was based on the unit risk estimate of the US-EPA of 8.3 · 10
–6
as presented in the Dutch
Integrated Criteria Document on benzene (RIVM, 1987) increased with a factor of 100. In combination with the unit
risk of the US EPA the 95% upper confidence limit of the cancer risk in the Pliofilm cohort was calculated to be 0.052
The INDEX project Final report
111
(=6234* · 8.3 · 10
–6
). Linear interpolation between the two points on the dose response curve, i.e. [12 µg/m
3
, 10
–6
] and
[6234 µg/m
3
, 0.052] gives an 10
–4
excess cancer risk at 24 µg/m
3
.
* The average exposure in the Pliofilm cohort was 128 mg/m
3
. Converting this value to a continuous lifetime exposure
for the general population results in a value of 6234 µg/m
3
(128 mg/m
3
· 10/75 years/lifetime · 5/7 days/week · 48/52
weeks/year · 10/18 inhalation volume/day).
European Commission (1999)
The EU-Working Group concluded that it is reasonable to assume that benzene-induced effects at low exposure levels
differ quantitatively as well as qualitatively from those induced at high occupational exposures. It is not clear whether
the underlying mechanism of benzene carcinogenicity is direct DNA interaction. In view of this uncertainty the EU
Working Group decided, on precautionary grounds, to hold on to a non-threshold extrapolation method. Though it was
not possible to give a precise estimate of the risk associated with benzene it was possible to define a range within which
that risk was likely to lie. The UR calculated by the WHO (6 · 10
–6
) was considered to result in the highest plausible
estimate of risk. The lowest UR which the EU Working Group felt was likely to be plausible was in the order of 5 10
–8
,
an estimate which is consistent with the Dutch Health Council’s analysis of the Wong and Raabe data. This range of
URs means that an excess lifetime risk for leukemia of 10–6 is associated with annual average concentrations varying
between 0.17 and 20 µg/m
3
(EU 1999).
On the basis of this risk estimation the European Commission has adopted an ambient air quality limit value for
benzene of 5 µg/m
3
to be met on 1 January 2010 (Directive 2000/69/EC). This limit value has been proposed by the EU
working group taking into account practical considerations; 5 µg/m
3
is as low as reasonably achievable in 2010.
OEHHA (2003)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
The inhalation cancer potency of benzene was estimated from observations of leukemia among U.S. rubber
hydrochloride workers exposed to benzene (Pliofilm Cohort) (Rinsky et al., 1987; Paxton et al., 1994) and benzene-
exposed workers from various industries in China (Chinese Worker Cohort) (Hayes et al., 1997). In estimating potency,
observations were assumed to follow a Poisson distribution. Relative risk was assumed to increase linearly with
cumulative exposure, and leukemia onset was assumed unaffected by benzene exposure after 30 years. Cancer potency
estimates, obtained from the occupational data, were adjusted using lifetable analyses to those expected for the
California population from constant exposure to benzene.
Studies in human volunteers, workers, and animals suggest that humans retain approximately 50 percent of inhaled
benzene and absorb 100 percent of ingested benzene at environmentally relevant levels of exposure (OEHHA, 2001a).
Based on these differences in absorption of benzene by route of exposure, the oral cancer potency was derived from the
inhalation cancer potency.
The evidence regarding the shape of the exposure-response curve for benzene-induced leukemia supports the use of
linear risk models to extrapolate to low doses (U.S. EPA, 1998; OEHHA, 2001a,b). Saturation of metabolism and
formation of reactive compounds may result in nonlinearity at high exposures.
OEHHA (2001a) reviewed the scientific evidence for associations of benzene and various human cancers. The strongest
association is for acute non-lymphocytic leukemia and myelodysplastic syndromes (ANLL/MDS). MDS, if not fatal,
often progresses to ANLL. Evidence suggests that benzene causes other forms of leukemia as well. Total leukemia
(e.g., all subtypes of leukemia as a related class of diseases) was judged to be appropriate as the basis of risk
assessment.
Cancer potency estimates were derived for all exposures and after removal of workers with the highest cumulative
exposures. Highly exposed workers were removed from the analysis because of reduced confidence in their exposure
assignments or because of observed non-linearity of response at higher exposures.
Upper-bound estimates of cancer potency from Pliofilm Cohort or Chinese Worker Cohort were very similar after
removal of the workers with the highest exposures from each cohort. Cancer potency estimates in units of ppm
-1
were
converted to units of (mg/kg-d)
-1
using default intake parameters of 70 kg body weight and 20 m
3
/d for breathing rate:
Potency/(mg/kg-d) = (upper-bound potency/ppm) x (ppm/3.19 mg/m
3
) x (70 kg) x (1/20 m
3
/d).
The INDEX project Final report
112
Thus, inhalation cancer potency estimates from the Pliofilm Cohort based on the Rinsky et al. or Crump and Allen
exposure estimates are 0.048 and 0.049 (mg/kg-day)
-1
, respectively, resulting in a geometric mean of 0.048 (mg/kg-
day)
-1
after rounding. The cancer potency estimate based on the Chinese Worker Cohort is 0.061 (mg/kg-day)
-1
. The
geometric mean estimate of these two cohort studies is 0.054 (mg/kg-day)
-1
. Cancer potency estimates from inhalation
exposures were converted to those expected from oral exposures by applying a factor of two since 50 percent of inhaled
benzene is absorbed and 100 percent of ingested benzene is absorbed (OEHHA, 2001a). The resulting oral cancer
potency estimates are 0.11 (mg/kg-day)
-1
The cancer potency estimates obtained from the Pliofilm Cohort and Chinese
Worker Cohort are consistent with estimates obtained from animal cancer bioassays of benzene, other epidemiological
studies of benzene-exposed workers, and epidemiological studies of leukemia and cigarette smoke (of which benzene is
a constituent) (OEHHA, 2001a).
The NO SIGNIFICANT RISK LEVEL (NSRL) for Proposition 65 is the intake associated with a lifetime cancer risk of
10
-5
. The cancer potency estimates derived above were used to calculate NSRLs for benzene, which are 13 µg/day for
inhalation exposures and 6.4 µg/day for oral exposures.
NICNAS (2001)
National Industrial Chemicals Notification and Assessment Scheme – Australia
NICNAS considered the Pliofilm cohort as the most reliable data set on which to establish the risk estimates. Data from
the most recent follow-up of the Pliofilm cohort indicate that the risk for leukaemia is significantly elevated at
cumulative exposures >50 ppm-years. However, this finding derives from a single cohort study with insufficient
statistical power to rule out the possibility of some increase in leukaemia risk at lower exposures. The additional risk for
leukaemia attributable to environmental exposure to benzene can be predicted by low-dose extrapolation of the
quantitative estimates for occupational exposure to benzene. Crump (1994) and USEPA (1998a) both predict the
number of additional leukaemia cases at two lifetime exposure levels, namely 1 ppm and 1 ppb, however, only the latter
is relevant for exposure to benzene in the general environment.
The risk characterisation is based on the most conservative prediction of the Pliofilm cohort (Crump, 1994 and USEPA,
1998a), that is, a lifetime leukaemia (AML) risk equivalent to 2 additional cases of leukaemia per 100,000 population at
1 ppb. By extrapolation, the lifetime leukaemia (AML) risk equivalent for increasing exposure levels can be calculated
as follows:
1 ppb 2 additional leukaemia/ 10
5
population
10 ppb 20 additional leukaemia / 10
5
population
20 ppb 40 additional leukaemia / 10
5
population
The NICNAS estimated that the average 24 hour lifetime exposure to benzene, from all sources (including an estimated
ambient air component of 11%), of an individual not exposed to environmental tobacco smoke living in an urban area
of an Australian city is 5.2 ppb. The predicted excess lifetime risk of leukaemia is therefore 1/10,000, or 1.2% of the
lifetime risk of contracting leukaemia of any cause (1 in 118, or 85/10,000 population, based on 1996 incidence figures
for Australia (AIHW, 1999).
AIHW. 1999. Cancer in Australia 1996 – incidence and mortality data for 1996 and selected data for1997 and 1998. Bruce, ACT,
Australian Institute of Health and Welfare.
NICNAS. 2001. National Industrial Chemicals Notification and Assessment Scheme, Benzene, Priority Existing Chemical
Assessment Report No. 21 (2001)
Susceptible population
The potential magnitude or range of susceptibilities cannot be accurately quantified at this time; however, current
evidence suggests individual differences in susceptibility may be two or three orders of magnitude.
Benzene’s toxicity is intimately tied to its complex metabolism and distribution. Key enzymes involved in the
metabolism of benzene, including CYP2E1, the quinone reductase NQO1, and myeloperoxidase are polymorphic. For
example, a 7.6-fold difference in benzene-induced hematotoxicity in workers was observed among gene variants of
CYP2E1 and NQO1 (Rothman et al., 1997). Thus the expression of genetic polymorphisms may modulate the
sensitivity of an individual or ethnic group to the effects of benzene exposure.
Aksoy et al. (1976) suggested familial susceptibility as one of the main factors in the frequency of chronic benzene
toxicity. Two patients with pancytopenia associated with chronic exposure to benzene were cousins. One of these two
The INDEX project Final report
113
patients died of aplastic anemia; the son of this patient presented with leukopenia a short time after exposure to this
chemical. Genetic polymorphism with regard to the P-450 enzymes that metabolize benzene to its reactive metabolites
may render some individuals at higher risk to the toxic effects of benzene (Kato et al. 1995). Although there are
possible indications of genetic susceptibility to benzene, it is believed that all humans are susceptible to the
pancytopenic effects of this agent.
Children may represent a subpopulation at increased risk due to factors that could increase their susceptibility to effects
of benzene exposure (e.g., activity patterns), on key pharmacokinetic processes (e.g., ventilation rates, metabolism rates,
and capacities), or on key pharmacodynamic processes (e.g., toxicant-target interactions in the immature hematopoietic
system).
There is a possible relationship between thalassemia (abnormal haemoglobins) and exposure to benzene. Aksoy (1989)
presented a series of 44 pancytopenic patients, 4 of which had β-thalassemia heterozygotes. Two of these four patients
had high levels of fetal haemoglobin, while a third patient had pseudo-Pelger-Huet anomaly. Thus, some forms of β-
thalassemia may increase the deleterious effects of benzene on the hematopoietic system.
The occurrence of aplastic anemia in chronic benzene toxicity may be accelerated in individuals with viral hepatitis
(Aksoy 1989). Furthermore, children and fetuses may be at increased risk-because their hematopoietic cell populations
are expanding and dividing cells are at a greater risk than quiescent cells.
In vitro studies of benzene-caused chromosomal aberrations in human lymphocytes taken from people with different
health practices suggests that “unhealthy lifestyles, ” including smoking, excessive alcohol consumption, inadequate
physical exercise and sleep, excessive stress, and inadequate nutritional balance, could make cells more sensitive to the
production of chromosomal aberrations after exposure (Morimoto et al. 1983).
Interactions with other chemicals
It has been reported that high exposures to gasoline (300 to 2000 ppm) which contains benzene may be less hazardous
than an equivalent exposure to benzene alone, based on studies in mice and from modeling predictions (Bond et al.,
1997). Volatile aromatics in gasoline including toluene, ethylbenzene and xylenes compete with benzene for CYP2E1,
thus these co-exposures significantly reduce the overall metabolism of benzene relative to equivalent exposures of pure
benzene. However, these experiments employed relatively high concentrations of test material (1) 300 ppm gasoline
plus six ppm benzene, and (2) 2000 ppm gasoline plus 40 ppm benzene), levels at which metabolic saturation is
expected. At low levels of exposure, competition for the P450 binding sites would be minimal; thus, reduction in
benzene toxicity due to co-exposures would also be minimal.
Odour perception
Source: Devos et al. (1990)
Odour threshold: 1.2 mg/m
3
.
Source: Haley, 1977
Odour threshold: 2.8 mg/m³
Source: AIHA (1989) - Odor thresholds for chemicals with established occupational health standards.
Detection: 195 mg/m
3
(61 ppm); Recognition 309 mg/m
3
(97 ppm). Reported values range from 2.5 – 510 mg/m
3
(0.78-
160 ppm).
The INDEX project Final report
114
Summary of Benzene Dose Response Assessment
________________________________________________________________________________________________
Exposure other than inhalation: ingestion of contaminated tap water, potential exposure from dermal contact during
showering and bathing (additional inhalatory exposure due to volatilized benzene). Dietary and endogenous sources of
phenol, hydroquinone and other primary metabolites of benzene confer potentially large differences in susceptibility to
benzene toxicity.
Toxicokinetics: at low levels (<10 ppm) about half of the inhaled dose is retained, the rest being exhaled. Metabolism
of benzene occurs largely in the liver, although some metabolism may take place in the bone marrow. Benzene can
cross the placenta. Not benzene itself, but several metabolites, as well as interactions between these metabolites, are
thought to explain the toxic effects of benzene. The proportion of the putatively toxic metabolites (benzoquinone and
muconaldehyde) formed is greater at low doses than at high doses (saturable process at relatively low doses).
Health effect levels of short- and long-term exposure (noncancer):
NOAEL
mg/m³
LOAEL
mg/m³
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
Short-term exposure
128
EXP
319
EXP
Developmental: decreased fetal body weights
(severe adverse effect!)
Pregnant female rats,
6 hours per day (for 5
days)
Coate et al., 1984;
(supported by Kuna
and Kapp, 1981;
Keller and Snyder,
1988)
OEHHA 1999
REL: 1.3
16
EXP
Developmental: Altered hematopoiesis in
offspring
(high degree of uncertainty!)
Mice, 6 hours per
day during days 5-16
of gestation
Keller and Snyder,
1988
OEHHA 2001
MADL
inhal
.: 1.3
µg/day
33
EXP
8
ADJ
47
HEC
Haematologica-immunological: depressed
peripheral lymphocytes and mitogen-induced
blastogenesis of femoral B-lymphocytes (Less
Serious LOAEL) .
Mice, 6 hours per
day (for 6
consecutive days)
Rozen et al. 1984
ATSDR 1997
MRL: 0.16
Long-term exposure
1.7
Study
0.60
ADJ
Haematological effects (total and differential
white blood cell counts, haemoglobin,
hematocrit, red blood cells, platelets and
clotting times)
Occupational, 7.4y Tsai et al. 1983
OEHHA 1999
REL: 0.06
44
BMC
23
BMCL
8.2
ADJ-
BMCL
Decreased lymphocyte counts Occupational, 6.3y
(0.7-16y)
Rothman et al. 1996 EPA-IRIS 2003
RfC: 0.03
(3.2) 32
EXP
Blood cell depression (lymphocytes) Mouse, 6-178d
NOAEC has been
derived, not observed
Dempster and
Snyder, 1990, Rozen
et al. 1984, Baarson
et al., 1984, Green et
al., 1981a,b, and
other studies
ECB 2003
2.5
EXP
1.05
HEC
Neurological: increased rapid response time
(Less Serious minimal LOAEL).
Mice, 30d
(2h/d, 6d/w)
Li et al. 1992
intermediate duration
study !
ATSDR 1997
MRL
INT
: 0.013
(intermediate!)
Carcinogenicity: IARC: 1 ; U.S.EPA: A ; Genotoxic carcinogen
Unit risks derived from Pliofilm cohort study - leukemia in occupational settings.EPA (1998): 2.2 - 7.8 · 10
-6
(µg/m³)
-1
;
WHO(1995): 6.0 · 10
-6
(µg/m³)
-1
; OEHHA(2003; Pliofilm cohort and Chinese worker cohort) with respect to breathing
rate: children 24 · 10
-6
(µg/m³)
-1
and adults 12 · 10
-6
(µg/m³)
-1
Genotoxicity: Induction of both numerical and structural chromosomal aberrations, sister chromatid exchanges and
micronuclei in experimental animals and humans after in vivo benzene exposure (as low as 4-7 mg/m³). Lack of
evidence for direct DNA interaction under normal in vivo conditions. Genotoxicity may result from a synergistic
combination of phenol with hydroquinone, muconaldehyde, or catechol.
Odour threshold: 1.2 mg/m³ (Devos); 2.8 mg/m³ (Haley, 1977)
Susceptible sub-populations: The range of susceptibility to benzene toxicity within the general population could be
very large (two or three orders of magnitude). Significant sources of variability are:
The INDEX project Final report
115
- genetic polymorphisms in key enzymes involved in the metabolism of benzene,
- dietary and endogenous sources of primary metabolites of benzene,
- factors such as pre-existing inflammation, radiation exposure, and ethanol consumption can potentiate the toxic
effects of exposure to benzene.
- Benzene has been implicated as a potential risk factor for the development of childhood leukemia.
Remarks:
The INDEX project Final report
116
6. Risk Characterization
Cancer risk evaluation
The most severe human responses to benzene exposure are those related to the blood-forming organs. There is a wealth
of evidence demonstrating benzene’s ability, at high concentrations, to cause bone marrow damage giving rise to
clinical outcomes such as various anemias, myelodysplastic syndromes, and leukemia. Additional evidence suggests
that benzene may also be associated with increased incidence of childhood leukemia. Benzene is regarded as a
genotoxic chemical, causing chromosomal damage and mutations in test systems in vitro, in animals in vivo, and in
occupationally exposed humans.
The present risk characterization is adressing benzene-induced leukemia and is supported by an evaluation on
haematological effects, considered a promotional condition to tumour development. Since no safe exposure level can be
established for genotoxic carcinogens, the quantitative risk estimate is here based on a non-threshold linear
extrapolation at low levels using the WHO Unit Risk guideline set as 6 · 10
–6
(µg/m
3
)
-1
and described as the geometric
mean of the range of estimates of the excess lifetime risk of leukemia at an air concentration of 1 µg/m
3
. This factor,
applied to a single exposure value or to a population distribution of exposure values, provides an estimate of the
individual risk or the population-based risk, the latter indicating the potential excess cancers among a million people
continuously exposed to the given benzene concentration over a 70-year lifetime.
Population cancer risk estimates of benzene-induced leukemia
In Table 6.1 the available exposure data (see cumulative frequency distributions in Chapter 1.3) are listed along with the
lifetime cancer risk estimates at three different exposure levels. Indoors, risk estimates are based on the median and the
90
th
percentile of the cumulative exposure distribution, with the 90
th
percentile describing a “worse case“ scenario
among each indoor survey. Both indoor levels were corrected following the assumption that people spend 95% of their
time in their homes and resting time outdoors. Additionally, risk estimates are refered to median outdoor concentrations,
a scenario describing indoor exposure experienced in the absence of indoor sources of benzene.
Table 6.1 Excess lifetime cancer risks per one million people derived from available exposure data
Population-based studies
Description (Study, Year)
N
Median estimate
Indoors
a
90
th
% estimate
Indoors
a
Median estimate
Outdoors
b
Athens, 30h-TWA (Expolis, 96-98) 42 48 112 45
Basel 30h-TWA (Expolis, 96-98) 47 13 28 8
Helsinki 30h-TWA (Expolis, 96-98) 188 10 25 9
Milan 30h-TWA (Expolis, 96-98) 41 60 216 55
Oxford 30h-TWA (Expolis, 98-00) 40 16
c
44
c
n.a.
Prague 30h-TWA (Expolis, 96-98) 46 41 80 28
England 28d-TWA (BRE, 97-99) 796 18
c
60
c
n.a.
Germany 4w-TWA (GerES IV pilot
study, spring+summer 2001)
44 8 16 7
French National Survey 7d-TWA (IAQ
observatory; 2003-04)
109
14
c
30
c
n.a.
a
supposing that people spend 95% of their time at home
b
supposing outdoor air as the unique source of indoor benzene exposure
c
not adjusted for outdoor exposure (0.05%)
n.a. data not available
The corresponding benzene concentrations are given in Table 6.2, with an additional column pointing out the
percentage of individuals exposed at levels higher than the current (2004) and the finalised (2010) EU-limit value of 10
and 5 µg/m
3
, respectively (1-y average; Directive 2000/69/EC). This limit value has been proposed by the EU working
group taking into account the developed risk estimation and practical considerations (5 µg/m
3
is as low as reasonably
considered achievable in 2010). Also, in Figure 6.1 the outcomes of the available indoor surveys are presented together
with the results of the MACBETH project.
The INDEX project Final report
117
0,0
0,5
1,0
1,5
2,0
2,5
(
5
) G
e
r
m
a
n
y 20
01
w
inte
r
(
2)
H
e
l
si
n
k
i
[SF] 1996-98
(2) Bas el [CH] 1996-98
(
6
) F
r
an
c
e
20
03
-
0
4
(
2
)
O
xf
o
r
d
[U
K] 19
96
-
9
8
(
3
)
C
o
p
en
h
ag
n
[D
K]
1
9
97
-
98
(4) England 1
9
97-99
(3) Pa
d
ova [I] 19
9
7-
9
8
(2) Prague [CZ] 1996
-
98
(
3
)
An
t
wer
p
[
B]
1
9
97
-
98
(
3)
R
o
ue
n
[F
]
1
99
7-
9
8
(3
)
Ath e
n
s (
E)
19
9
7-
9
8
(
2) Athens [E]
1996-98
(3) M
u
rcia [E]
9
7-98
(1
)
G
e
rma
ny
1
99
0
-
92
(
2
) Mi
la
n [I
]
1
9
96
-
9
8
Population excess lifetime cancer risk per 10.000 people
2004 EU Limit Value
2010 EU Limit Value
Table 6.2: Benzene concentrations (µg/m
3
) measured and percentages of population exposed indoors at levels higher
than the EU-limit value (1y avg.): 10 µg/m
3
(current) and 5 µg/m
3
to be met the 01/01/10
% exceeding
Population-based studies Description
(Study, Year)
N
Median
Indoors
90
th
%
Indoors
Median
Outdoors
10 µg/m
3
current
5 µg/m
3
01/01/10
Athens, 30h-TWA (Expolis, 96-98) 42 8,0 19,2 7,5 35% 75%
Basel 30h-TWA (Expolis, 96-98) 47 2,3 4,8 1,3 < 10%
Helsinki 30h-TWA (Expolis, 96-98) 188 1,7 4,3 1,5 < 10%
Milan 30h-TWA (Expolis, 96-98) 41 10,0 37,5 9,1 50% 75%
Oxford 30h-TWA (Expolis, 98-00) 40 2,6 7,3 - 5% 20%
Prague 30h-TWA (Expolis, 96-98) 46 6,9 13,8 4,6 25% 75%
England 28d-TWA (BRE, 97-99) 796 3,0 9,0 - 15% 30%
Germany 4w-TWA (GerES IV pilot study,
spring+summer 2001)
44 1,3 2,7 1,1 < <
French National Survey 7d-TWA (IAQ
observatory; 2003-04)
109
2,3 5,0 - < 10%
n.a. data not available;
< out of the evaluation range (i.e. <5% of the environments investigated)
Figure 6.1:Cancer risk estimates of benzene induced leukemia (last minute implementation)
based on arithmetic mean and 90%ile (---) indoor levels measured among European surveys
(1) GerES II ; (2) EXPOLIS ; (3) MACBETH ; (4) BRE National survey ; (5) GerES IVP ; (6) French National
survey
The INDEX project Final report
118
Health hazard evaluation (noncancer)
Both epidemiology and animal experiments show that carcinogenic effects occur at benzene levels also expected to be
haematotoxic. Together with the observation that susceptibility to haematotoxicity by benzene evidently predisposes to
myelodysplastic syndromes as well as haematopoietic malignancies, this target organ toxicity is considered a
promotional condition to tumour development.
A complementary health hazard evaluation is here based on haematological effects (see Table 6.3) with an observed
chronic NOAEL of 600 mg/m
3
(OEHHA, corrected for continuous exposure) and an assessment factor of ten for inter-
individual variability. In the occupational study refered to, total and differential white blood cell counts, hemoglobin,
hematocrit, red blood cells, platelets and clotting times of the population studied were found to be within normal limits
(between 5% and 95% percentile) during an exposure duration averaged 7.4 years.
Table 6.3: Haematological effects:
Population exposed beyond given threshold levels (µg/m
3
)
NOAEL
NOAEL
10
Population-based studies
Description (Study, Year)
N 600 60
Athens, 30h-TWA (Expolis, 96-98) 42 < <
Basel 30h-TWA (Expolis, 96-98) 47 < <
Helsinki 30h-TWA (Expolis, 96-98) 188 < <
Milan 30h-TWA (Expolis, 96-98) 41 < 5%
Oxford 30h-TWA (Expolis, 98-00) 40 < <
Prague 30h-TWA (Expolis, 96-98) 46 < <
England 28d-TWA (BRE, 97-99) 796 < <
Germany 4w-TWA (GerES IV pilot
study, spring+summer 01)
44 < <
French National Survey 7d-TWA (IAQ
observatory; 2003-04)
109
< <
n.a. data not available; < out of the
evaluation range (i.e. <5% of the
environments investigated)
The studies
The EXPOLIS study has been designed to produce direct population based measurements of exposures for major air
pollutants, the target population being the adult urban population of Europe (measurements performed from fall 1996 to
winter 1997/1998). (Selection bias (Oglesby et al., 2000) In Basel, participants of direct monitoring as compared to non-
participants were more likely to live at streets with low traffic volume; although in Helsinki, traffic volume was neither
significantly related to participation in direct nor indirect monitoring, the point estimates indicate a tendency to
decreased participation with increasing traffic intensity at home.)
The Building Research Establishment (BRE, UK) has conducted a national representative survey of air pollutants in
876 homes in England, designed to increase knowledge of baseline pollutant levels and factors associated with high
concentrations (17 months, October 1997 to February 1999).
The GerES IVP is a one-year pilot study (February 2001 - March 2002) of the German Environmental Survey (GerES),
and includes about 560 children/teenagers (0 to 17 years) with a sub collective of 112 for personal, indoor and outdoor
air analyses. The aim of the study was to collect information on parameters influencing the response rate and to test the
suitability of the different instruments intended to be used for the main study. The here presented benzene exposure data
are based on indoor air measurements averaged over 4 weeks.
The French National Survey: Study details in
A nationwide survey on indoor air quality has been st up in France in 2003-2004, with the aim of assessing household
exposure to indoor pollutants. The target population is the national housing stock of approximately 24 million
permanently occupied housing units. For further details refer on Golliot et al., 2003 and Kirchner et al., 2004.
The INDEX project Final report
119
Comments
A widespread characterization of residential exposures to benzene is given by the outcome of the Expolis-study,
considered here as indicative of the urban population in Europe. Within this study, a specific bias has been reported
(Oglesby et al. 2000) concerning the selection of residences (participants) in Basel and Helsinki, with these data more
likely to be collected in lower trafficked areas of the towns.
In two further surveys (BRE and GerES IVP), one-month average benzene levels were measured in randomly
distributed residences in England and Germany. With the exception of the GerES IV pilot study, all studies took into
account possible seasonal effects on indoor benzene concentration.
Tendency in ambient benzene levels
Since outdoor air is representing a source of indoor benzene levels, the adoption, in most member states, of
environmental policies regarding benzene emissions, raises the question on whether the available/selected exposure data
could be considered descriptive of the nowadays situation, the comment requiring a brief introduction to the policies
adopted and on-going tendencies of benzene ambient levels.
Combustion processes are the largest source of benzene in the atmosphere, with road traffic generally the biggest single
source. Especially in urban locations, the emission by vehicles is expected to be the principal cause of increased
benzene concentrations. Emissions are resulting from
direct emission from the exhausts and from the
evaporation of fuels either by car or from fuel
distribution and refuelling. These emissions have been
declining in recent years, owing to legislation on
vehicle emissions, industrial emissions and fuel
distribution. In particular, important results were
obtained with the introduction of the automobile
catalyst converter in the last two decades and the
reduction of the benzene content of gasoline more
recently. The Air Quality Report of the first Auto-Oil
Programme (AOP1) estimated a 56% reduction in
urban emissions of benzene between 1990 and 2010.
As an example, the control of benzene in gasoline has
recently been described as the main reason for the
decrease of indoor benzene levels in Germany, together
with the reduction of the use of benzene in many
consumer products (Seifert, 2002; Schleibinger et al., 2001). Levels decreased by a factor of about 4 over one decade in
the Berlin metropolitan area (see Figure). Although an example of a successful application of environmental control
measures in Germany, a tendencial decrease is expected to occur in all member states adopting these policies. As a
result, both the Expolis and the BRE data, collected more than 5 years ago, are expected to describe a pessimistic
picture of the actual situation, with respect to the contribution of ambient benzene levels.
When examining the exposure data (Table 6.2) with respect to the contribution of outdoor air contamination, two
exposure scenarios could be depicted as follows:
- In highly trafficked urban areas (outdoor benzene levels >5 µg/m
3
), outdoor air is well contributing to indoor
benzene levels, as shown in the case of Milan and Athens, where median indoor-levels exceed outdoor-levels for
not more than 9%. In these locations the relative contribution of indoor specific sources (e.g. tobacco smoke) is
likely to be impaired.
- At low outdoor levels (benzene <5 µg/m
3
), indoor sources moderately (Basel) or marginally (GerES IVP,
Helsinki) contribute to exposure, with median indoor levels not more than three-fold the background benzene
concentration (0.6 to 1.9 µg/m
3
in European continental pristine air,
as reported by ECB 2004).
Benzene levels measured in Prague and Oxford (EXPOLIS) and in England (BRE) are expected to fall between the two
scenarios described.
As pointed out in Table 6.1, the additional lifetime cancer risk in Europe is expected to range from 8 to 216 · 10
–6
(µg/m
3
)
-1
,
resulting in a 27-fold increase experienced by worse-case residentials in Milan (37 µg/m
3
) with respect to
average residentials in Germany (1.3 µg/m
3
), the latter considered as the European background cancer risk estimate,
with 7.5 cases of benzene-induced leukemia per one million people. Moreover, almost the entire bulk of data is below
the exposure level (60 µg/m
3
) considered safe with respect to haematological effects, the most severe toxicological
enpoint other than cancer.
The INDEX project Final report
120
EU limit value for benzene in ambient air – Directive 2000/69/EC
Limit value for the protection of human health: 5 µg/m
3
(averaging period of a calendar year)
Margin of tolerance: 5 µg/m
3
(100 %) on 13 December 2000, reducing on 1 January 2006 and every 12 months thereafter by 1 µg/m
3
to reach 0 % by
1 January 2010
Date by which limit value is to be met: 1 January 2010, except within zones and agglomerations within which a time-limited extension has been
agreed in accordance with Article 3(2) of the directive. The limit value for benzene to be granted during conditional extension for a period of up to
five years shall, however, not exceed 10 µg/m
3
.
Result
Benzene is ubiquitous in the athmosphere, mainly due to anthropogenic sources (90%), with concentrations in the
European continental pristine air ranging from 0.6 to 1.9 µg/m
3
. It is a genotoxic carcinogen and hence no safe level of
exposure could be recommended. Results from nine monitoring surveys indicate that the European population is
experiencing in their homes an increased risk in contracting benzene induced leukemia, with respect to the estimated
background lifetime risk of 7-8 cases per one million people (considering the WHO unit risk factor). Based on the
available exposure data (Median levels±sd: 4.2±3.2 µg/m
3
; 90th levels±sd: 11.5±11.1 µg/m
3
; N = 9) two main scenarios
could be described as follows:
- People living in highly trafficked urban areas are found, on average, to experience an estimated 6 to 30-fold increase
in contracting benzene induced leukemia during their life, the benzene levels encountered in these areas not expected
to produce chronic effects other than cancer, in particular haematological effects, nor acute sensory effects such as
odour perception (odour threshold: 1.2 mg/m
3
) and sensory irritation. Also, the contributions of specific indoor
sources in these areas are likely to be impaired due to increased background outdoor levels.
- People living in rural areas or poorly trafficked towns were found, on average, to experience an estimated 1 to 5-fold
increase in contracting benzene induced leukemia during their life, this factor depending principally on the presence
of indoor sources.
The INDEX project Final report
121
Naphthalene
Synonyms: antimite, naphthalin, naphthaline, naphthene, tar camphor
CAS Registry Numbers: 91-20-3
Molecular Formula: C
10
H
8
1. Compound identification
Naphthalene is white crystalline powder with aromatic odour (of mothballs). It is a two-ring hydrocarbon isolated from
coal tar. It is used as intermediate in chemical synthesis, as insect repellents, fungicides, lubricants, preservatives, and,
formerly, as topical antiseptics (EPA/Cal 2003, HSDB 2003). Gasoline and diesel fuels contain naphthalene.
Naphthalene is used indoors as a moth repellent, though this use is decreased. It has also been used in the manufacture
of phthalic anhydride, phthalic and anthranilic acids, naphthols, naphthylamines, 1-naphthyl-n-methylcarbamate
insecticide, beta-naphthol, naphthalene sulfonates, synthetic resins, celluloid, lampblack, smokeless powder,
anthraquinone, indigo, perylene, and hydronaphthalenes (EPA/Cal 2003, HSDB 2003).
Naphthalene emissions to atmosphere are mainly originated from fugitive emissions and motor vehicle exhaust. Spills
into land and water during the storage, transport and disposal of fuel oil and coal tar are lost and released to atmosphere
due to volatilisation, photolysis, adsorption, and biodegradation. Naphthalene has a relatively short half-life, 3-8 hr, in
the atmosphere. It is assessed that the primary route of exposure is inhalation, especially in vicinity of heavy traffic, gas
stations or refineries. Usual indoor sources of naphthalene are unvented kerosene heaters and tobacco smoke (EPA/Cal
2003, HSDB 2003).
2. Physical and Chemical properties
Molecular weight (g/mol) 128.18
Melting point (°C) 80.5
Boiling point (°C) 218
Density, (g/cm3, at 20 °C, 1 atm) 4.42
Solubility Soluble in alcohol, acetate
Conversion factors at 20 °C and 760 mm Hg:
1 ppb = 5.321 µg/m
3
1 µg/m
3
= 0.188 ppb
Source: CRC (1995), Verschueren 2001, EPA/Cal (2003)
3. Indoor Air Exposure assessment
Indoor air and exposure concentrations
Average outdoor naphthalene concentrations were low in Europe, ranging from 1 to 4 µg/m
3
(Table 4.0.1, Jantunen et al
1999). Also residential indoor concentrations were elsewhere low, typically average concentrations below 2 µg/m
3
, but
in Athens clearly higher indoor levels were measured, being on average 90 µg/m
3
. Personal exposures to naphthalene
ranged from 1 µg/m
3
to 3 µg/m
3
elsewhere (Jantunen et al 1999, Hoffman et al 2000), but in Athens average exposure
was 46 µg/m
3
. In general we can conclude that exposures to naphthalene were usually low in Europe, but in Athens
remarkable indoor sources of naphthalene were present, because personal exposure and workplace concentrations were
lower than indoor concentrations (Figure 3.1, Table 4.0.1). Figure 3.2 shows the same distributions as Figure 3.1, but
without Athens to be able to see distributions better focused on their scale. Similarly, exposure distributions with and
without Athens are presented in Figure 3.3 and Figure 3.4.
The INDEX project Final report
122
Maroni et al (1995) reported typical median and 90
th
percentile naphthalene concentrations in indoor air being 2 µg/m
3
and 5 µg/m
3
, respectively. Kostiainen et al (1995) detected slightly lower indoor concentrations in Helsinki, Finland,
having 0.44 µg/m
3
and 1.63 µg/m
3
as a mean and maximum concentrations.
Bituminous material, commonly used in UK for damp proofing floors emits naphthalene (Brown et al 1990).
Naphthalene concentrations up to 970 µg/m
3
were found in homes having an objectionable smell, where the damp proof
membrane had been applied, compared with <300 µg/m
3
for control homes (EU 2003).
In an Italian study average indoor naphthalene concentration was 11 µg/m
3
, maximum level being 70 µg/m
3
(DeBortoli
et al 1986). Similar concentrations were measured in Canada, showing average and maximum concentrations of 14
µg/m
3
and 77 µg/m
3
, respectively (Chan et al 1990).
0
20
40
60
80
100
0 50 100 150 200 250 300 350
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
Hel
Mil
Oxf
Pra
Figure 3.1. Cumulative frequency distributions of indoor air concentrations of naphthalene in Athens (Ath, n=42), Basel
(Bas, n=47), Helsinki (Hel, n=188), Milan (Mil, n=38) Oxford (Oxf, n=40) and Prague (Pra, n=46) (EXPOLIS 2002).
0
20
40
60
80
100
03691215
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Bas
Hel
Mil
Oxf
Pra
Figure 3.2. Cumulative frequency distributions of indoor air concentrations of naphthalene in Basel (Bas, n=47),
Helsinki (Hel, n=188), Milan (Mil, n=38) Oxford (Oxf, n=40) and Prague (Pra, n=46) (EXPOLIS 2002).
The INDEX project Final report
123
0
20
40
60
80
100
0 50 100 150
Exposure (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
GeS
Hel
Oxf
Pra
Figure 3.3. Cumulative frequency distributions of 48-hour personal exposure concentrations of naphthalene in Athens
(Ath), Basel (Bas), Helsinki (Hel), Oxford (Oxf) and Prague (Pra) (EXPOLIS 2002), and 1-week mean concentrations
of the German Survey GerES II (GeS) (Hoffman et al 2000).
0
20
40
60
80
100
024681012
Exposure (µg/m
3
)
Cumulative frequency (%)
Bas
Hel
GeS
Oxf
Pra
Figure 3.4. Cumulative frequency distributions of 48-hour personal exposure concentrations of naphthalene in Basel
(Bas), Helsinki (Hel), Oxford (Oxf) and Prague (Pra) (EXPOLIS 2002), and 1-week mean concentrations of the German
Survey GerES II (GeS) (Hoffman et al 2000).
The INDEX project Final report
124
4. Toxicokinetics
Absorption
No empirical data that describe the rate or extent of naphthalene absorption following inhalation exposure were
identified. NTP (2000) developed a physiologically-based pharmacokinetic model to describe the uptake of naphthalene
in rats and mice following inhalation exposure. The model was calibrated using blood time course data for naphthalene
(parent compound). Results from this model suggest that inhaled naphthalene is absorbed rapidly into the blood
(Blood:air partition coefficient of 571). On the basis of estimates of naphthalene metabolism generated by the model,
approximately 22% to 31% of inhaled naphthalene is absorbed by rats and 65% to 73% of inhaled naphthalene is
absorbed by mice.
Assuming an average ambient concentration level of 5.19 µg/m
3
and an average inhalation rate of 15.2 m
3
/day (U.S.
EPA, 1996), an average daily dose of 1,127 ng/kgday can be calculated for a 70-kg adult. An estimated average daily
dose of 4,5 ng/kg-day can be calculated for a 10-kg child assuming an inhalation rate of 8.7 m
3
/day (U.S. EPA, 1996).
Distribution
There are limited data concerning the distribution of naphthalene in human tissues. Naphthalene was present in 40% of
the adipose tissue samples that were analyzed as part of the National Human Adipose Tissue Survey (EPA 1986). The
maximum concentration observed was 63 ng/g. Naphthalene was also detected in human milk samples (concentration
not reported) (Pellizzari et al. 1982). The sources of naphthalene in these milk and body fat samples are not known.
Three reports (Zinkham and Childs, 1958; Anziulewicz et al., 1959; Athanasiou et al., 1997) describe apparent
transplacental exposure of a fetus during pregnancy, which resulted in neonatal hemolysis. In the two older cases,
unspecified amounts of naphthalene had been ingested by the mother during pregnancy. The more recent report by
Athanasiou et al. (1997) documented the occurrence of hemolytic anemia in a neonate whose mother had inhaled
naphthalene during the 28
th
week of pregnancy.
Metabolism and elimination
The in vivo and in vitro metabolism of naphthalene in mammalian systems has been extensively studied (U.S. EPA
1998). As many as 21 metabolites, including oxidized derivatives and conjugates, have been identified in the urine of
animals exposed to naphthalene (Horning et al., 1980; Wells et al., 1989; Kanekal et al., 1990). Factors that potentially
influence the relative proportions of individual metabolites include species, tissue type, and tissue concentration of
naphthalene (U.S. EPA, 1998). The initial step in naphthalene metabolism is catalyzed by cytochrome P-450
monooxygenases, and results in the formation of the arene epoxide intermediate 1,2-naphthalene oxide (Figure 4.1).
1,2-Naphthalene oxide can undergo spontaneous rearrangement to form naphthols (predominately 1-naphthol). The
resulting intermediates may be further metabolized by oxidation reactions, resulting in the formation of di-, tri-, and
tetrahydroxylated intermediates (Horning et al., 1980). Some metabolites may undergo conjugation with glutathione,
glucuronic acid, or sulfate (ATSDR, 1995; U.S. EPA, 1998). Glutathione conjugates undergo additional reactions to
form cysteine derivatives (thioethers). These cysteine derivatives may be further metabolized to mercapturic acids and
may be excreted in the bile (U.S. EPA, 1998). Alternative pathway of naphthol metabolism involves enzymatic
hydration by epoxide hydrolase (U.S. EPA, 1998).
Bieniek (1994) analyzed the excretion patterns of 1-naphthol in three groups of workers occupationally exposed to
naphthalene. The mean excretion rate for these workers was 0.57 mg/hour, with a calculated excretion half-life of
approximately 4 hours. The highest urinary levels of 1-naphthol were reported for workers in a naphthalene oil
distribution plant. Peak 1-naphthol levels were detected in urine collected one hour after finishing the shift.
Excretion in the feces represents a minor pathway for naphthalene, and the possibility of excretion via exhalation of the
unmetabolized compound has not been examined in available studies.
The INDEX project Final report
125
Figure 4.1: Proposed Pathways For Naphthalene Metabolism
Source: U.S. EPA (1998)
5. Health effects
Information is scarce regarding dose-response relationships for health effects in humans with acute, subchronic, or
chronic exposure by any route. Reported health effects associated with indoor exposures to naphthalene are limited to
its principal indoor source: mothballs.
Effects of short-term exposure
Reports that establish associations between naphthalene exposure and health effects in humans are restricted to
numerous reports of hemolytic anemia or cataracts following acute exposure or occupational exposure to naphthalene,
either by ingestion or by inhalation of naphthalene vapors, but these reports have not identified exposure levels
associated with these effects. A relationship appears to exist between an inherited deficiency in the enzyme, glucose 6-
phosphate dehydrogenase (G6PD), and susceptibility to naphthalene-induced hemolysis. Newborn infants also appear to
The INDEX project Final report
126
be susceptible to naphthalene-induced hemolysis presumably due to a decreased ability to conjugate and excrete
naphthalene metabolites.
Individuals with a G6PD genetic defect are prone to hemolysis after exposure to a variety of chemical oxidizing agents
including nitrates, nitrites, aniline, phenols (Dean et al. 1992), and naphthalene. Valaes et al. (1963) reported adverse
effects in 21 Greek infants exposed to naphthalene from clothing, diapers, blankets, and other items that had been stored
in contact with mothballs. Durations of exposure ranged from 1 to 7 days. Inhalation was identified as the primary route
of exposure because 19 of the 21 infants did not have dermal contact with the naphthalene-contaminated materials. A
total of 21 infants developed hemolytic anemia and two infants died from kernicterus, a severe neurological condition
that was thought to be a consequence of massive hemolysis. Ten of the 21 anemic children and 1 of the 2 infants that
died from naphthalene exposure had a genetic polymorphism that resulted in a deficiency in glucose-6-phosphate
dehydrogenase (G6PD). This enzyme helps to protect red blood cells from oxidative damage, and G6PD deficiency
makes the cells more sensitive to a wide variety of toxicants, including naphthalene. Eight adults and one child reported
gastrointestinal (nausea, vomiting, abdominal pain) and neurological (headache, malaise, confusion) symptoms after
exposure to large numbers of mothballs in their homes (Linick, 1983). The duration of exposure was not specified.
Testing at one home following the incident indicated an airborne naphthalene concentration of 105 mg/m
3
(20 ppb).
Symptoms abated after removal of the mothballs.
There are no data available on skin or respiratory sensitisation in humans. Acute (4-hour) inhalation exposure to
naphthalene induced necrosis of Clara cells in the epithelium of the proximal airways of the lungs of mice at exposure
levels as low as 53 mg/m
3
(10 ppm), but did not affect lung tissue in rats at concentrations as high as 526 mg/m
3
(100
ppm) (West et al. 2001). These results, and those from the chronic inhalation studies, show that mice are more
susceptible than rats to lung damage from inhaled naphthalene. However, there are no studies that have examined nasal
tissues for the development of lesions following acute inhalation exposure. A change to mouth breathing occurred in
rats during exposure to 410 mg/m
3
naphthalene, but no other effects on respiration were noted (Fait and Nachreiner
1985).
Hemolytic anaemia (potentially sensitive populations)
Increased sensitivity to naphthalene-induced hemolysis has been associated with reduced levels of glucose-6-phosphate
dehydrogenase (G6PD). This enzyme helps to protect red blood cells from oxidative damage, and G6PD enzyme
deficiency makes the cells more sensitive to a wide variety of toxicants, including naphthalene. Higher rates of inherited
G6PD deficiencies are found more often in defined subpopulations of males from Asian, Arab, Caucasian (of Latin
ancestry), African, and African-American ancestry than in other groups (U.S. EPA, 1987). Multiple forms of G6PD
deficiency have been identified in these subpopulations. The mildest forms are totally asymptomatic, while moderate
forms are associated with an adverse response to chemical stressors, including naphthalene. The most severe forms of
G6PD deficiency are associated with hemolytic anemia, even in the absence of external stressors (Beutler, 1991). The
overall prevalence of G6PD-deficiency in the United States is reported to be 5.2 to 11.5% (Luzzatto and Mehta, 1989).
There is no evidence of haemolytic anaemia in rodents.
Prevalence of G6PD enzyme deficiency in the EU: Polymorphic G6PD variants are those that have achieved a high
frequency in some populations. They represent balanced polymorphisms in which the benefit of inheriting the mutation
(probably resistance to malaria) counterbalances the disadvantage (susceptibility to hemolysis and neonatal icterus).
Generally, each population has its own characteristics mutations, although, as noted below, there are occasional
exceptions to this rule.
- Mediterranean Variants: G6PD deficiency is very prevalent in some Mediterranean countries. A gene frequency of
about 0.7 has been documented among Kurdish Jews. This is the highest incidence known in any group. Among
Greeks, Turks, Sardinians, Sephardic Jews and Italians, G6PD deficiency is also quite prevalent, but more commonly
with gene frequencies that range from 0.02 to 0.20. The situation regarding Mediterranean variants is in one respect
the reverse of the situation of the African variants. Here several different variants (e.g., G6PD “Sassari” and
“Cagliari”) were believed to be different on the basis of biochemical characteristics, but all seem to share the same
mutation at nt 563. Variants from other parts the world, thought to be unique - G6PD Dallas, Birmingham, and
Panama - proved to be G6PD Mediterranean 563T. Other mutations are found in the Mediterranean region as well.
G6PD Seattle844T (also described as G6PD Lodi and as G6PD “Modena”) and, as noted above, G6PD A- are
relatively common.
- African Variants: G6PD deficiency among Africans is relatively mild; red cells contain 10-15% residual enzyme that
is electrophoretically rapid. Accordingly it is designated G6PD A- to distinguish its mobility from the normal
enzyme, which is designated B. Among Afro- Americans the gene frequency of G6PD A- is about 11%.(11) It is
considerably higher in some parts of Africa.(12) Some Africans have an enzyme with the same rapid mobility
encountered among deficient individuals, but the activity of the enzyme is normal. This African enzyme is designated
The INDEX project Final report
127
G6PD A+, and it is known to be due to an A®G transition at nt 376 that predicts substitution of a negatively charged
aspartic acid for asparagine. Its gene frequency is in the range of 20-30%
Polymorphic G6PD variants that have been characterized at the DNA level have been summarized in Beutler, 1994,
Vulliamy et al., 1993 and Beutler et al., 1995.
Summary of short-term exposure effect levels
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
11
EXP
53
EXP
Respiratory: Clara cell necrosis and decreased
Clara cell mass
(volume/surface area) in proximal
airways
Mouse; 4h West et al. 2001
ATSDR 2003;
no MRL
ECB (2003): In relation to haemolytic anaemia, the available data do not allow the identification of a NOAEL.
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
ATSDR (2003)
Agency for Toxic Substances and Disease Registry
Data are inadequate for deriving an acute-duration inhalation MRL for naphthalene. Data are restricted to a 14-day (6
hours/day, 5 days/week) range-finding study in B6C3F1 mice (NTP 1992a), which only examined hematologic end
points and did not histologically examine expected critical toxicity targets (lung and nasal cavity epithelial tissue) (NTP
1992a), and a study (West et al. 2001) with Swiss Webster mice and Sprague-Dawley rats, which involved single 4-
hour exposure periods. The more recent study, however, only histologically examined the lung and did not examine
nasal tissue. A comprehensive inhalation study involving an acute repeated exposure scenario and examining the other
critical target (the nose, based on the findings from chronic mouse and rat bioassays) is not currently available. Results
from such a study may be useful for deriving an acute-duration inhalation MRL for naphthalene.
Hemolysis is the best documented effect of acute naphthalene exposures in humans, but it has not been observed in
studied strains of rats (F344) or mice (CD-1, B6C3F1). Dose-response data for hemolysis from a susceptible animal
species (such as dogs or the Jackson Laboratory hemolytic anemia mouse) may be useful to obtain data that could be
used for considering changes to the acute-duration oral MRL. Data from both inhalation and oral exposure protocols
would be useful.
ECB (2003)
European Chemical Bureau - Institute for Health and Consumer Protection - European Union Risk Assessment Report
In relation to hemolytic anaemia, the available data do not allow the identification of a NOAEL nor the dose-response
characteristics for this endpoint. In the absence of such information, any significant body burden is considered to give
rise to concerns for human health. Exposure of infants to textiles (clothing/bedding) which have been stored for long
periods with naphthalene moth repellent raises significant concern. There is documented evidence for the development
of severe hemolytic anaemia resulting from such use, although there is no quantitative information available on the
level or duration of exposure to naphthalene in these cases.
Effects of long-term exposure (noncancer)
There are no epidemiological studies on the human health effects of naphthalene, and the only human information
available derives from a limited number of early case-reports which provide no quantitative data on the levels or
duration of exposure. A principal human health effect is haemolytic anaemia (see previous chapters) which in some
cases has been of marked severity in humans exposed by inhalation to naphthalene vapour and by ingestion to solid
naphthalene. Dermal exposure to the solid and vapour was also likely in these cases.
Animal studies reveal species differences in response to naphthalene. Haemolytic anaemia was noted in a dog following
oral dosing of 220 mg/kg/day for 7 days but not in rodents even with high/prolonged exposures. Cataract formation was
the principal effect seen in rats and rabbits following oral exposure to 700 and 1000 mg/kg/day, respectively in studies
ranging from 10-180 days, but this effect was not seen in mice with similar exposures. The lack of reliable reports of
The INDEX project Final report
128
cataracts in humans despite the widespread use of naphthalene and high dose accidental exposure, suggests that cataract
formation is unlikely to be a significant health effect in humans (ECB, 2003).
The few chronic animal studies that are available for naphthalene were conducted primarily to characterize its
carcinogenic potential effects. Noncancer endpoints reported in these studies are summarized below.
Adkins et al. (1986) investigated the impacts of less-than-lifetime inhalation exposures to naphthalene on rats. Groups
of 30 female A/J strain rats were exposed to 0, 53, or 158 mg/m
3
(0, 10, or 30 ppm) naphthalene vapors for 6 hours per
day, 5 days per week, for 6 months. All animals were sacrificed at the end of the exposure period and their lungs
excised and examined for tumors. No adverse noncancer effects on the lung were reported (U.S. EPA, 1998). Other
organs were not examined in study.
A chronic inhalation study of naphthalene toxicity was conducted in B6C3F
1 mice by NTP (1992a). Naphthalene
exposure concentrations in air were 0, 53, or 158 mg/m
3
(0, 10 ppm, or 30 ppm). The concentration of 53 mg/m
3
was
chosen because it was equal to the ACGIH TLV® for naphthalene, while the 158 mg/m
3
concentration was chosen
because it was one-half the air saturation concentration. The control and low-exposure groups consisted of 75 mice of
each sex, while the high-exposure group consisted of 150 mice of each sex. Exposure was for 6 hours per day, 5 days
per week, for 2 years. Comprehensive histopathological evaluations were performed on all control and high-exposure
mice, and on all low-exposure mice that died or were sacrificed during the first 21 months of exposure. The original
study plan called for 50 animals per sex to be exposed for 2 years, and 5 animals per sex to be sacrificed for
hematological evaluations at 14 days, 3, 6, 12, and 18 months. However, as a result of excessive mortality in the control
males, only the 14-day hematological evaluation was conducted. All of the remaining animals were incorporated into
the two-year study.
A statistically significant decrease in survival was noted in the male control group. This phenomenon was attributed to
the frequent fighting that occurred among the control group mice. In contrast, the exposed groups tended to huddle
together during exposure periods, and fought less. Statistically significant increases were seen in several types of
noncancer respiratory tract lesions in both exposed groups. The observed responses included chronic lung
inflammation, chronic nasal irritation with hyperplasia of the nasal epithelium, and metaplasia of the olfactory
epithelium. The authors of the study described the lung lesions as a chronic inflammatory response with granuloma.
These lesions consisted of “focal intra-alveolar mixed inflammatory cell exudates and interstitial fibrosis.” The more
advanced lesions consisted primarily of “large foamy macrophages sometimes accompanied by giant cells.”
No changes in hematological parameters were seen among the exposed animals at 14 days. No cataract formation was
observed after 2 years of exposure. Histopathological examination did not reveal treatment-related effects on the liver,
gastrointestinal system, reproductive system, brain, or any other organs. The results of this study have been interpreted
by U.S. EPA (1998) to support a chronic LOAEL for nasal and respiratory irritation of 53 mg/m
3
.
NTP (2000) conducted a chronic inhalation study in F344/N rats. Male and female rats (49/sex/dose) were exposed to
naphthalene vapor concentrations of 0, 53, 158, and 316 mg/m
3
(0, 10, 30, and 60 ppm) for 6 hours per day plus T
90
(the
theoretical time to achieve 90% of the target concentration in the vapor chamber: 12 minutes), 5 days per week for 105
weeks. Additional groups of rats were similarly exposed for up to 18 months for evaluation of toxicokinetic parameters.
Dose calculations were based upon model estimates of the amount of naphthalene inhaled by rats at the exposure
concentrations used in the two-year study, the total amount of naphthalene metabolized following a six-hour exposure
period (21% to 31% of inhaled naphthalene), and average weights of 125 grams (male rats) and 100 grams (female
rats). Because essentially all of the naphthalene that is absorbed into the bloodstream is metabolized, the total amount of
naphthalene metabolized was assumed to represent the internalized dose to rats from the exposure concentrations used
in this two-year study. The estimated daily doses determined by this method were 0, 3.6, 10.7, 20.1 mg/kg-day for
males, and 0, 3.9, 11.4, and 20.6 mg/kg-day for females.
Rats were clinically examined twice daily and findings were recorded every four weeks beginning at week 4 and every
two weeks beginning at week 92. Body weights were recorded at study initiation, every four weeks beginning at week
4, and every two weeks beginning at week 92. Full necropsies and complete histopathologies were performed on all
core study animals.
There were no clinical findings related to naphthalene exposure from the two-year inhalation study. The mean body
weights all exposed groups of male and female rats were similar to those observed in the appropriate control chamber
group. No significant difference in survival rate was observed for any exposed group when compared to the chamber
control. The mean body weights of female rats were generally similar to the body weights of the control group, while
the mean body weights of naphthalene-exposed male rats were generally less than the chamber control for all exposed
groups.
The INDEX project Final report
129
Although naphthalene is a known cataractogen and ocular irritant, no naphthalene-related cataractogenic effects or
ocular abnormalities were observed in rats during this study. Treatment-related non-neoplastic lesions were observed in
the nose and lungs of male and female rats. The incidence and average severity of nasal lesions (glands, goblet cells,
respiratory epithelium and olfactory epithelium) are summarized in Table 5.1. The incidences of these lesions were
significantly greater than those in the chamber controls for all male and female exposed groups, with the exception of
squamous metaplasia of glands in male and female rats in the 53 mg/m
3
exposure groups (NTP, 2000). In general, the
severities of olfactory epithelial and glandular lesions increased with increasing exposure concentrations.
Two noteworthy type of lesions occurred in the lungs of exposed rats: alveolar epithelial hyperplasia and minimal
chronic inflammation. Female rats in all exposure groups had increased incidences of alveolar epithelial hyperplasia
when compared to the chamber control (chamber control: 4/49, 53 mg/m
3
10ppm: 11/49, 158 mg/m
3
: 11/49, 316 mg/m
3
:
9/49). This effect reached statistical significance in the 53 and 158 mg/m
3
exposure groups. The incidences of alveolar
epithelial hyperplasia in male rats (chamber control: 23/49, 53 mg/m
3
: 12/49, 158 mg/m
3
: 9/48, 316 mg/m
3
: 16/49) were
significantly decreased in the 53 and 158 mg/m
3
exposure groups. The incidences of minimal chronic inflammation of
the lung were increased in males and females exposed to naphthalene. This lesion is characterized by small focal
interstitial and intra-alveolar collections of macrophages, neutrophils, and lymphocytes and minimal interstitial fibrosis.
As noted by the NTP study authors, foci of minimal inflammation are common in chamber control rats (as evident in
this study). Therefore, this change could not be confidently related to naphthalene exposure.
The study conducted by NTP (2000) identified an estimated inhalation LOAEL of 3.6 mg/kg-day based on the
occurrence of nasal lesions in male rats in the 53 mg/m
3
exposure group. A NOAEL was not identified in this study.
The 53 mg/m
3
concentration associated with the LOAEL corresponds to the threshold limit value for naphthalene
(ACGIH, 2000).
Table 5.1: Nonneoplastic and Neoplastic Lesions of the Nose in Male and Female F344/N Rats
Exposed to Naphthalene 6 Hours/Day, 5 Days/Week for 105 Weeks (NTP, 2000)
Concentration 0 mg/m
3
53 mg/m
3
(10 ppm)
158 mg/m
3
(30 ppm)
316 mg/m
3
(60 ppm)
Lesion M F M F M F M F
Nonneoplastic lesions
Olfactory epithelium
Hyperplasia 0/49 0/49 48/49 48/49 45/48 48/49 46/48 43/49
Atrophy 0/49 0/49 49/49 49/49 48/48 49/49 47/48 47/49
Chronic inflammation 0/49 0/49 49/49 47/49 48/48 47/49 48/48 45/49
Hyaline degeneration 0/49 13/49 46/49 46/49 40/48 49/49 38/48 45/49
Respiratory epithelium
Hyperplasia 0/49 0/49 21/49 18/49 29/48 22/49 29/48 23/49
Squamous metaplasia 0/49 0/49 15/49 21/49 23/48 17/49 18/48 15/49
Hyaline degeneration 0/49 8/49 20/49 33/49 19/48 34/49 19/48 28/49
Goblet cell hyperplasia 0/49 0/49 25/49 16/49 29/48 29/49 26/48 20/49
Gland hyperplasia 0/49 0/49 49/49 48/49 48/48 48/49 48/48 42/49
Gland squamous metaplasia 0/49 0/49 3/49 2/49 14/48 20/49 26/48 20/49
Neoplastic lesions
Respiratory epithelial adenoma 0/49 0/49 6/49 0/49 8/48 4/49 15/48 2/49
Olfactory epithelial
neuroblastoma
0/49 0/49 0/49 2/49 4/48 3/49 3/48 12/49
Source: Adapted from NTP Technical Report on the Toxicology and Carcinogenesis Studies of Naphthalene in Rats (Inhalation Studies), Table 6
(NTP, 2000) (M=male, F=female)
Naphthalene administered by inhalation at concentrations of 10, 30, or 60 ppm for 6 hours per day, 5 days per week for
105 weeks caused nonneoplastic and neoplastic effects in nasal respiratory and olfactory regions of male and female
F344/N rats. Non-neoplastic nasal effects were characterized by an increase in the incidence and severity of a complex
group of lesions, including atypical hyperplasia, atrophy, chronic inflammation, and hyaline degeneration of olfactory
epithelium; hyperplasia, squamous metaplasia, hyaline degeneration, and goblet cell hyperplasia of the respiratory
epithelium; and hyperplasia and squamous metaplasia of mucosal glands. Neoplastic effects were characterized by the
induction of two types of rare primary nasal tumors, olfactory neuroblastomas and respiratory epithelial adenomas. The
incidences of olfactory neuroblastomas in males at 0 ppm, 10 ppm, 30 ppm, and 60 ppm were, respectively, 0%, 0%,
8%, and 6%, whereas in females they were 0%, 4%, 6%, and 24%. The incidences of respiratory epithelial adenomas in
The INDEX project Final report
130
males at 0 ppm, 10 ppm, 30 ppm, and 60 ppm were, respectively, 0%, 12%, 17%, and 31% and in females 0%, 0%, 8%,
and 4%. The olfactory neuroblastomas and respiratory epithelial adenomas were considered carcinogenic effects related
to naphthalene exposure based on their relatively high incidence in exposed rats, their absence in concurrent control rats
and NTP historical controls, and their rare spontaneous occurrence in rats of any strain (Abdo et al., 2001; Long et al.,
2003).
In a well conducted unpublished study, groups of 10 male and 10 female rats were exposed nose only for 6 hours/day, 5
days a week for 13 weeks to 0, 11, 53 or 305 mg/m
3
(0, 2, 10 or 58 ppm) vapourised naphthalene (Huntingdon Research
Centre, 1993a). A gross pathological examination was carried out on a wide range of tissues and a microscopic
examination was carried out on a range of tissues including the lungs, liver, kidneys, adrenals, testes, eyes and optic
nerve. Prior to terminal sacrifice, samples of blood were taken from all rats for haematological and clinical chemistry
evaluation. In high dose animals body weight gain was reduced by 43% and 34% in males and females, respectively and
was associated with reduced food consumption. There were no toxicologically significant haematological or clinical
chemistry findings observed. Similarly, no significant changes were noted in organ weight or gross pathology.
Microscopic analysis of the nasal epithelium revealed treatment-related effects at all dose levels. The severity of the
effects was dose-related. At the highest exposure level (305 mg/m
3
) changes included erosion of the olfactory
epithelium, hyperplasia of basal cells in the olfactory epithelium and loss of Bowmans' glands. At the lowest exposure
level (11 mg/m
3
) changes in olfactory epithelium were less marked but included slight disorganisation, mild erosion (in
one rat), minimal atrophy, rosette formation (an attempt at proliferative repair by the olfactory neuroepithelium),
occasional degenerate cells, loss of Bowmans' glands and minimal hyperplasia. There were no treatment related effects
observed in the lungs or nasal respiratory epithelium at this dose. There were no observed changes in the nasal passages
of control animals. In one low dose rat there was evidence of squamous metaplasia of the respiratory epithelium,
however as this lesion was not seen in the other rats at higher doses this lesion was not considered toxicologically
significant. The effects at 10 mg/m
3
were generally minimal in severity and seen in only small numbers of animals, and
therefore appear to represent the low end of the dose-response curve for nasal effects. Overall, signs of damage to the
olfactory epithelium were seen at all doses down to 11 mg/m
3
(2 ppm), and a NOAEL cannot be identified for local
effects.
In a well conducted unpublished study, groups of 5 male and 5 female rats were exposed nose only for 6 hours/day, 5
days a week for 4 weeks to 0, 5, 16, 53, 153 or 374 mg/m
3
(0, 1, 3, 10, 29 or 71 ppm) vapourised naphthalene
(Huntingdon Research Centre, 1993b). Investigations were similar to the 13-week study performed in the same
laboratory. Results were similar to those observed in the 13-week study. High dose animals showed approximately a
50% reduction in body weight gain associated with reduced food consumption. There was no evidence of systemic
toxicity. Local effects were observed with signs of proliferative repair in the nasal olfactory epithelium changes
observed at all doses down to 5 mg/m
3
(1 ppm), and therefore a NOAEL for local effects cannot be identified.
For both the 4 and 13-week studies the mechanism by which the observed effects in the olfactory nasal epithelium arise
is unclear, although the effects may be mediated by locally produced metabolite(s) of naphthalene. The relevance of
these effects to human health is uncertain, as there may be significant species differences in local metabolism. However,
there is no evidence to indicate that these effects are not relevant to human health.
Reproductive and developmental toxicity
No information is available on both the reproductive and developmental effects of naphthalene in humans, although the
occurrence of hemolytic anemia in the neonates of anemic, naphthalene-exposed mothers demonstrates that naphthalene
and/or its metabolites can cross the placental barrier (Anziulewicz et al. 1959; Zinkham and Childs 1957, 1958).
Animal studies involving naphthalene exposure during gestation reported no reproductive effects in rabbits
administered doses of up to 120 mg/kg/day by gavage or in rats given doses of up to 450 mg/kg/day, although doses of
150 mg/kg/day and greater were maternally toxic to rats. There was a decrease in the number of live mouse pups per
litter with a dose of 300 mg/kg/day given during gestation (Plasterer et al. 1985) and in vitro studies of naphthalene
embryotoxicity in the presence of liver microsomes support the concept that naphthalene metabolites may be harmful to
the developing embryo (Iyer et al. 1991).
No exposure-related lesions in reproductive tissues were found in intermediate-duration oral exposure studies in rats
(NTP 1980b) and mice (NTP 1980a) or in chronic inhalation studies in rats (Abdo et al. 2001; NTP 2000) or mice (NTP
1992a). One- or two-generation reproductive toxicity studies evaluating reproductive performance variables in male and
female animals exposed to naphthalene are not available.
The INDEX project Final report
131
Studies of the developmental effects of orally administered naphthalene in rats (NTP 1991), mice (Plasterer et al. 1985),
and rabbits (NTP 1992b; PRI 1985, 1986) have been negative, except for a slight nonsignificant increase in fused
sternebrae in female rabbit pups from a small number of litters at doses of 80 and 120 mg/kg/day (NTP 1992b). No
developmental toxicity studies involving inhalation or dermal exposure to naphthalene are available.
Summary of long-term exposure effect levels (noncancer)
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
53
EXP
9.4
ADJ
Respiratory; Nasal inflammation, olfactory
epithelial metaplasia, and respiratory epithelial
hyperplasia
Mice, 105w
NTP, 1992a OEHHA 2000;
REL: 0.009;
EPA-IRIS 1998 ;
RfC: 0.003
53
EXP
9.4
ADJ
1.2
HEC
Respiratory; nonneoplastic lesions in nasal
olfactory epithelium and respiratory epithelium
in two species
Combined studies:
Mice 2y - Rats 105w -
Rats 2y
NTP, 1992a;
NTP, 2000;
Abdo et al., 2001
ATSDR 2003;
MRL: 0.004
5
EXP
0.94
ADJ
Respiratory; proliferative repair in the nasal
olfactory epithelium
Rats, 4w Huntingdon Research
Centre, 1993b
(unpublished)
ECB 2003
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
OEHHA (2000)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Derivation of the Chronic Reference Exposure Level (REL):
Study NTP (1992a)
Study population B6C3F1 mice (75 or 150/group/sex)
Exposure method Discontinuous whole-body inhalation exposures to 0, 10, or 30 ppm
naphthalene vapor
Critical effects Nasal inflammation, olfactory epithelial metaplasia, and respiratory
epithelial hyperplasia
LOAEL 10 ppm (96% incidence for males and 100% incidence for females)
NOAEL Not observed
Exposure continuity 6 hours/day for 5 days/week
Average experimental exposure 1.8 ppm (10 ppm x 6/24 x 5/7) for LOAEL group
Exposure duration 104 weeks
Subchronic uncertainty factor 1
LOAEL uncertainty factor 10
Interspecies uncertainty factor 10 (see below)
Intraspecies uncertainty factor 10
Cumulative uncertainty factor 1000
Inhalation reference exposure level 0.002 ppm (2 ppb, 0.009 mg/m
3
, 9 µg/m
3
)
The NTP study was chosen for the REL derivation since it is the only available lifetime animal inhalation bioassay and
because no adequate epidemiological studies of long-term human exposure are available. The study was judged to be of
adequate study design. The complete lack of nasal effects among control animals and the nearly total effect among
animals exposed at 2 different concentrations strongly indicates a causal relationship between naphthalene exposure and
nasal effects. The effects seen are consistent with those reported among exposed workers, who developed
rhinopharyngolaryngitis or laryngeal carcinoma (Wolf, 1978). However, the hematological effects observed in humans
have not been reported in laboratory animals, which raises the possibility that humans may be significantly more
sensitive to naphthalene.
The most important limitation of the study is that the lowest concentration tested caused adverse effects in most (96%)
of the animals tested. Thus the study amply demonstrates the risk of lifetime exposures to 53 mg/m
3
(10 ppm), but is
uninformative regarding the concentration-response relationship at lower concentrations. Only a general assumption can
be drawn on the magnitude of uncertainty factor needed to predict a concentration at which adverse effects would most
likely not be observed. Lacking specific guidance or relevant research for this situation, the default 10-fold factor was
applied. U.S. EPA also used the NTP study to develop its RfC of 3 mg/m
3
with slightly different assumptions and a
cumulative uncertainty factor of 3000 (U.S. EPA, 2000). OEHHA followed the U.S. EPA precedent in using an
The INDEX project Final report
132
intraspecies UF of 10 for naphthalene, rather than using the HEC/RGDR approach. According to U.S. EPA (2000),
because of its low water solubility and low reactivity, naphthalene-related effects on the nasal epithelium are expected
to result following absorption of naphthalene and its metabolism to reactive oxygenated metabolites, not from direct
contact. This is supported by data on naphthalene metabolism indicating that toxic effects on the respiratory tract are
due to a naphthalene metabolite that may be formed either in the liver or in the respiratory tract. Necrosis of bronchial
epithelial (Clara) cells in mice and necrosis of olfactory epithelium in mice, rats, and hamsters occur following
intraperitoneal injection of naphthalene. The nasal effects from inhalation exposure to naphthalene were considered to
be extra-respiratory effects of a category 3 gas (U.S. EPA, 1994). The assumption is made that nasal responses in mice
to inhaled naphthalene are relevant to humans; however, it is uncertain that the RfC for naphthalene based on nasal
effects will be protective for hemolytic anemia and cataracts, the more well-known effects from naphthalene exposure
in humans.
U.S.EPA - IRIS (1998)
Integrated Risk Information System
Determination of the Reference Concentration for Chronic Inhalation Exposure (RfC)
Critical effect Experimental
doses*
UF MF RfC
Nasal effects: hyperplasia and metaplasia in respiratory
and olfactory epithelium, respectively
Chronic mouse inhalation study
NTP, 1992a
NOAEL: None
LOAEL(HEC): 9.3
mg/m
3
3000 1 0.003 mg/m
3
*Conversion Factors and Assumptions -- Following the Category 3 guidance (U.S. EPA, 1994), experimental exposure concentrations of
0, 10, and 30 ppm were converted to 0, 52, and 157 mg/m
3
, respectively; adjusted to a continuous exposure basis in mg/m
3
(6/24 hr ×
5/7 days) equals mg/m
3
× 0.1786: 0, 9.3, and 28 mg/m
3
. Because the blood:gas (air) coefficients for naphthalene were not available, the
default ratio of 1 was used and the values for the LOAEL(HEC) were 0, 9.3, and 28 mg/m
3
. Scenario -- The LOAEL human equivalent
concentration (HEC) was calculated for an extrarespiratory effect for a category 3 gas. Since the b:a lambda for humans (h) is unknown,
a default value of 1.0 is used for this ratio. LOAEL(HEC) × [b:a lambda(animal)/b:a lambda(human)] = 9.3 mg/m
3
.
The NTP (1992a) study was given medium confidence because adequate numbers of animals were used, and the
severity of nasal effects increased at the higher exposure concentration. However, the study produced high mortality, (<
40% survival in the male control group due to wound trauma and secondary lesions resulting from increased fighting).
Also, hematological evaluation was not conducted beyond 14 days. The database was given a low-to-medium
confidence rating because there are no chronic or subchronic inhalation studies in other animal species, and there are no
reproductive or developmental studies for inhalation exposure. In the absence of human or primate toxicity data, the
assumption is made that nasal responses in mice to inhaled naphthalene are relevant to humans; however, it cannot be
said with certainty that this RfC for naphthalene based on nasal effects will be protective for hemolytic anemia
and cataracts, the more well-known human effect from naphthalene exposure. Medium confidence in the RfC
follows.
Dose conversion: Because of its low water solubility and low reactivity, naphthalene-related effects on the nasal
epithelium are expected to result following absorption of naphthalene and metabolism to reactive oxygenated
metabolites, rather than being a result of direct contact. This hypothesis is supported by data on naphthalene metabolism
indicating that toxic effects on the respiratory tract are due to a naphthalene metabolite that may be formed either in the
liver or in the respiratory tract. For example, necrosis of bronchial epithelial (Clara) cells in mice (O'Brien et al., 1985,
1989; Tong et al., 1981) and necrosis of olfactory epithelium in mice, rats, and hamsters (Plopper et al., 1992) occur
following intraperitoneal injection of naphthalene. The nasal effects from inhalation exposure to naphthalene were
considered to be extra-respiratory effects of a category 3 gas, as defined in the U.S. EPA guidance for deriving RfCs
(U.S. EPA, 1994). Following this guidance, experimental exposure concentrations were adjusted to a mg/m
3
basis (0,
52, and 157 mg/m
3
), adjusted to a continuous exposure basis (mg/m
3
× 6h/24h × 5d/7d = mg/m
3
× 0.1786: 0, 9.3, and 28
mg/m
3
), and converted to human equivalent concentrations (HECs) by multiplying the adjusted concentrations by the
ratio of mouse:human blood/gas partition coefficients. Because the blood/gas coefficients for naphthalene were not
available, the default ratio of 1 was used.
Dose-response modeling: Whereas the data from the NTP (1992a) study show nasal effects to be the most sensitive
effects from chronic inhalation exposure to naphthalene, they provide no indication of the shape of the dose-response
curve because the incidence of nasal lesions at the lowest exposure level was 100% in females and nearly 100% in
males. In this case, application of a BMD approach, in which quantal mathematical models are fit to the incidence data
for nasal effects, does not sensibly assist in extrapolating to a NOAEL, and a NOAEL/LOAEL approach was taken for
deriving an RfC for naphthalene.
The INDEX project Final report
133
ATSDR (2003)
Agency for Toxic Substances and Disease Registry
Derivation of the Minimal Risk Level (MRL):
Dose and end point used for MRL derivation: The lowest exposure level in both studies (NTP, 1992a; 2000), 53 mg/m
3
(10 ppm), was a LOAEL in both sexes of both species for nonneoplastic lesions in nasal olfactory epithelium and
respiratory epithelium. Applying EPA inhalation dosimetry, a human equivalent LOAEL of 1.2 mg/m
3
(0.2 ppm), based
on the rat LOAEL, was selected as the point of departure for the chronic inhalation MRL.
Modifying Factors used in MRL derivation:
10 for use of a LOAEL
3 for extrapolation from animals to humans with dosimetric adjustment
10 for human variability
Total Uncertainty Factor = 10x3x10=300
Determination of the human equivalent dose:
10 ppm x 6 hours/24 hours x 5 days/7 days x 128.18/24.45 = 9.4 mg/m
3
(duration-adjusted LOAEL for nasal effects in
rats or mice)
Following EPA (1994) Methods for Derivation of Inhalation Reference Concentrations and Application of Inhalation
Dosimetry, equations for a category 1 gas producing nasal effects were used to derive human equivalent concentrations:
HEC=Animal Concentration x RGDR
ET
;
RGDR
ET
= regional gas dose ratio in the extrathoracic (ET) region
= (Dose
ET
)A/(Dose
ET
)H = [minute volume/ETsurface area]A÷[minute volume/ETsurface area]H;
Reference minute volumes (L/min): 13.8 human, 0.137 rat, 0.0368 mouse;
Reference ET surface area (cm2 ): 200 human, 15 rat, 3 mouse;
RGDR
ET
(Rat to Human)=[0.137/15]÷[13.8/200]=0.132; LOAEL
HEC
=duration-adjusted LOAEL x 0.132=9.4 mg/m
3
x
0.132=1.2 mg/m
3
RGDR
ET
(Mouse to Human)=[0.0368/3]÷[13.8/200]=0.178; LOAEL
HEC
=duration-adjusted LOAEL x 0.132=9.4 mg/m
3
x 0.178=1.7 mg/m
3
Using public health protection reasoning, the LOAEL
HEC
based on the rat data was selected as the point of departure for
the chronic inhalation MRL.
ECB (2003)
European Chemical Bureau - Institute for Health and Consumer Protection - European Union Risk Assessment Report
The hazardous properties of naphthalene have been evaluated to the extent that the minimum data requirements
according to Article 9(2) of Council Regulation EEC No.793/93 have been met. The key health effects of haemolytic
anaemia, repeated inhalation toxicity and carcinogenicity have been identified. For haemolytic anaemia, it is not
possible to identify a NOAEL from the available data. For repeated inhalation toxicity, the key effect of concern is local
damage to the upper respiratory tract. The available data do not allow the identification of a NOAEL; in a 28-day study
in rats (Huntingdon Research Centre, 1993b)., damage to nasal olfactory tissue occurred at 5 mg/m
3
, the lowest
concentration level used. Therefore, the LOAEL of 5 mg/m
3
from this study will be used in the risk characterisation for
repeated inhalation toxicity, including carcinogenicity. This experimental exposure concentration has been further
adjusted by the authors to a continuous exposure basis: 5 mg/m
3
× 6h/24h × 5d/7d = 0.9 mg/m
3
.
Genotoxicity
Naphthalene has given reproducible negative results in bacterial mutation assays, and was negative in an in vitro UDS
assay. It was however found to be clastogenic in CHO cells in the presence but not the absence of S9. Two in vitro
studies using CHO cells and human peripheral lymphocytes were negative for induction of SCE. Naphthalene was
found to be negative in two in vivo bone-marrow micronucleus tests and an in vivo rat liver UDS study. Overall, the
balance of evidence indicates that naphthalene is not genotoxic (ECB, 2003).
The available data suggest that genotoxic action by the naphthalene metabolite, 1,2-naphthoquinone, is plausible and
that the mutagenic/genotoxic potential of naphthalene and its metabolites may be weak. Assays of possible genotoxic
The INDEX project Final report
134
action in sensitive target tissues of naphthalene in rodents (lung and nasal epithelial tissue), however, are not available
(ATSDR, 2003).
Carcinogenic potential
The only studies of cancer in humans exposed to naphthalene are two case series reports of cancer; one report of four
laryngeal cancer cases (all of whom were smokers) among workers in a naphthalene purification plant in East Germany,
and another report of 23 cases of colorectal carcinoma admitted to a hospital in Nigeria. NTP (2002), EPA (2002), and
IARC (2002) concur that these studies provide inadequate evidence of naphthalene carcinogenicity in humans. No
cohort mortality or morbidity studies or case-control studies examining possible associations between naphthalene
exposure and increased risk of cancer (or other health effects) are available.
There are two comprehensive chronic-duration inhalation toxicology and carcinogenicity studies of naphthalene in
animals, one in rats (Abdo et al. 2001; NTP 2000) and one in mice (NTP 1992a). These studies identify respiratory
tissues as the most sensitive toxicity targets of chronic-duration exposure to inhaled naphthalene in animals: in
particular neuroblastoma of the nasal olfactory epithelium was observed in rats and neoplastic lesions in the lungs of
mice. Exposure related lesions in other tissues were not found in these studies. NTP (2002) and IARC (2002) concurred
that these studies provide sufficient evidence of naphthalene carcinogenicity in animals. Overall, the proposed
mechanism of carcinogenic action IARC (2002) is that the higher rates of metabolism of naphthalene in mice lead to
cytotoxic metabolites in the lung, causing increased cell turnover and tumours. The absence of lung tumours in rats is
entirely consistent with this mechanism. The maximal rates of metabolism measured in human lung microsomes are
about 10–100 times lower than those in mice.
The Office of Environmental Health Hazard Assessment (OEHHA) on march 2004 adopted a unit risk value for
naphthalene of 3.4 x 10
-5
(µg/m
3
)
-1
and slope factor of 1.2 x 10
-1
(mg/kg-day)
-1
. These values are based on data for
incidence of nasal respiratory epithelial adenoma and nasal olfactory epithelial neuroblastoma in male rats.
Interactions with other chemicals
Schmeltz et al. (1978) suggested that it is likely that certain naphthalenes compete with benzo[a]pyrene (BaP) for the
same enzyme sites, resulting in alteration of the BaP metabolic pathway and decreased production of the active BaP
metabolite. This hypothesis is consistent with the observation that benzo(a)pyrene hydroxylase is inhibited by
naphthalene (Shopp et al. 1984). Dermal application of the naphthalene mixture did not induce tumors in the absence of
BaP. The results of these studies were not analyzed statistically.
Several studies have been conducted to assess factors that influence the toxicity of naphthalene. For the most part, these
studies have evaluated the effects of mixed function oxidase activity (MFO) and alterations in glutathione levels on
pulmonary and ocular toxicities. The effects of cyclooxygenase activity, antioxidants, and epoxide hydrolase inhibitors
on the cataractogenic effect of naphthalene have also been evaluated. The administration of MFO inhibitors (SKF-
525A, metyrapone) and antioxidants (caffeic acid and vitamin E) decreased ocular toxicity in mice (Wells et al. 1989).
Use of ALO1576, an inhibitor of the enzyme aldose reductase, prevented cataract formation in both in vivo and in vitro
studies (Xu et al. 1992a, 1992b). On the other hand, naphthalene-induced cataracts were enhanced by pretreatment with
a MFO inducer (phenobarbital) and a glutathione depletor (diethyl maleate) (Wells et al. 1989). Pulmonary damage was
decreased by prior treatment with a MFO inhibitor (piperonyl butoxide), but enhanced by prior treatment with a
glutathione depletor (diethyl maleate) (Warren et al. 1982). For the most part, these studies support the role for mixed
function oxidase activity and glutathione conjugation in naphthalene-induced pulmonary and ocular lesions.
Odour perception
Source: Amoore and Hautala (1983)
Odour threshold: 0.42 mg/m3 (0.08 ppm)
Source: Devos et al. (1990)
Odour threshold: 7.5 µg/m3
Source: Canadian Centre for Occupational Health and Safety - CCOHS
Odour is perceptible at 1.6 mg/m3 to 4.7 mg/m3 (0.3 to 0.9 ppm)
The INDEX project Final report
135
Source: New Jersey Department of health and senior services - Hazardous Substance Fact Sheets
Odour threshold: 0.2 mg/m3 (0.038 ppm).
Summary of Naphthalene Dose Response Assessment
________________________________________________________________________________________________
Exposure other than inhalation: Rapid dermal absorption after skin contact
Toxicokinetics: Rapidly absorbed into the blood. Distributes to adipose tissue, breast milk and fetus.
Health effect levels of short- and long-term exposure (noncancer)
NOAEL
mg/m³
LOAEL
mg/m³
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
Short-term exposure
11
EXP
53
EXP
Respiratory: Clara cell necrosis and decreased
Clara cell mass (volume/surface area) in
proximal airways
Mouse; 4h West et al. 2001
ATSDR 2003;
no MRL
Long-term exposure
53
EXP
9.4
ADJ
Respiratory; Nasal inflammation, olfactory
epithelial metaplasia, and respiratory epithelial
hyperplasia
Mice, 105w
NTP, 1992a OEHHA 2000;
REL: 0.009;
EPA-IRIS 1998 ;
RfC: 0.003
53
EXP
9.4
ADJ
1.2
HEC
Respiratory; nonneoplastic lesions in nasal
olfactory epithelium and respiratory epithelium
in two species
Combined studies:
Mice 2y - Rats 105w -
Rats 2y
NTP, 1992a;
NTP, 2000;
Abdo et al., 2001
ATSDR 2003;
MRL: 0.004
5
EXP
0.94
ADJ
Respiratory; proliferative repair in the nasal
olfactory epithelium
Rats, 4w Huntingdon Research
Centre, 1993b
(unpublished)
ECB 2003
Carcinogenicity: IARC: 2B ; U.S.EPA: C ; ACGIH: A4 ; Inadequate evidence in humans
Genotoxicity: plausible for 1,2-naphthoquinone (naphthalene metabolite)
Odour threshold: 0.0075 mg/m³ (Devos), 0.2 mg/m³ (HSFS), 0.42 mg/m³ (Amoore &Hautala)
Susceptible population:
- Newborn infants susceptible to naphthalene-induced anaemia due to poor metabolism.
- Inherited G6PD enzyme deficiency (with mild, moderate, and severe polymorphic variants). G6PD deficiency is very
prevalent in some Mediterranean countries. Among Greeks, Turks, Sardinians, Sephardic Jews and Italians, G6PD
deficiency with gene frequencies that range from 0.02 to 0.20.
Remarks: Acute inhalatory exposure seems to be typically associated with wear of naphthalene contaminated clothing.
The INDEX project Final report
136
6. Risk Characterization
Health hazard evaluation of short-term exposure
Case reports of individuals (primarily infants) exposed to naphthalene via inhalation, dermal contact, or a combination
of both exposure routes (e.g. textiles which have been stored with naphthalene moth repellent) point to hemolytic
anemia and its sequelae as the most commonly manifested effects in humans following exposure at concentrations that
exceed average environmental levels. Sensitive populations include individuals deficient in the enzyme glucose-6-
phosphate dehydrogenase (G6PD). Also, newborns and infants are thought to be more susceptible than older people
because hepatic enzymes involved in conjugation and excretion of naphthalene metabolites are not well developed after
birth. There are no studies that have specifically examined the influence of age on naphthalene toxicokinetic capabilities
in humans. Although the availability of such studies may increase the understanding of the specific physiological basis
for the apparent susceptibility of newborns, they are unlikely to be conducted. Experiments examining the most
sensitive targets in animals are likely surrogates. In addittion, there is no evidence of haemolytic anaemia in rodents.
Consequently, in relation to naphthalene induced anaemia, available data do not allow the identification of a NOAEL
nor the dose-response characteristics for this endpoint. Naphthalene has a distinct odour of mothbolls or coal tar, with a
reported minimum thresholds at 7.5 µg/m³.
Health hazard and cancer risk evaluation of long-term exposure
No reliable human toxicity data were found for subchronic or chronic exposure to naphthalen.
In the NTP study (1992, chosen by U.S.EPA, ATSDR and OEHHA as a key-study for the derivation of exposure limits)
mice were exposed discontinuously to whole-body naphthalene inhalation at 0, 53 and 158 mg/m
3
. The complete lack of
nasal effects among control animals and the nearly total effect among animals exposed at 2 different concentrations
strongly indicates a causal relationship between naphthalene exposure and nasal effects. The effects seen (Toxicological
endpoint: Respiratory; Nasal inflammation, olfactory epithelial metaplasia, and respiratory epithelial hyperplasia) are
consistent with those reported among exposed workers, who developed rhinopharyngolaryngitis or laryngeal carcinoma
and the assumption is made that nasal responses in mice to inhaled naphthalene are relevant to humans.
Taking the lower concentration as a LOAEL and adjusting this value in order to allow for conversion from
discontinuous to a continuous pattern of exposure a concentration of 10 mg/m
3
is obtained. Incorporating factors of 10
for interspecies variability, 10 for interindividual variation and 10 for use of a LOAEL rather than a NOAEL this results
in an Exposure Limit of 0.01 mg/m
3
(see also Table 6.1). However, it cannot be said with certainty that this EL for
naphthalene based on nasal effects will be protective for hemolytic anemia, the more well-known human effect from
naphthalene exposure.
As stated in the ECB risk assessment report, in relation to carcinogenicity, naphthalene is not genotoxic in vivo and thus
the tumours are considered to arise via a non-genotoxic mechanism. The tumours develop only at the sites where non-
neoplastic inflammatory changes also occur. Thus, it is considered that the development of the nasal tumours in the rat
is a consequence of chronic tissue injury, for which an identifiable threshold of effect will exist, although currently not
identified. Given that the underlying mechanism for the development of nasal tumours in the rat is considered to be the
chronic inflammatory damage seen at this site, it follows that prevention of local tissue damage would prevent
subsequent development of tumours.
Result
With regard to the general population a long-term exposure limit has been set at 10 µg/m
3
, according to the assumption
that nasal effects observed in mice are consistent with the health effects reported among exposed workers. Available
exposure data indicate that, on average, the European population is exposed at naphthalene levels 10 times lower than
this EL, although an important exception resulted from a survey held in Athens, were levels exceeding the EL were
measured in nearly all residences. It is assumed that increased residential exposures originate from the use of
naphthalene based moth-repellents, a widespread use occuring in certain countries of the Mediterranean area.
An important source of uncertainty in establishing safe exposure limits is the potentially greater sensitivity of certain
subpopulations to naphthalene toxicity, including infants and children, neonates, fetuses, and individuals deficient in
glucose-6-phosphate dehydrogenase (G6PD), the prevalence of this inherited deficiency reported to be 2 to 20% in
defined Mediterranean subpopulations. In these latter cases manifested effects are hemolytic anemia and its sequelae.
The INDEX project Final report
137
In relation to carcinogenicity, naphthalene is not genototoxic in vivo and thus tumor development, observed in rodents,
is considered to arise via a non-genotoxic mechanism. Also, the underlying mechanism for the development of nasal
tumours in the rat is considered to be the chronic inflammatory damage seen at this site. It follows that prevention of
local tissue damage would prevent subsequent development of tumours.
Relevance of EU-population exposure to naphthalene
Table 6.1: Percentage of population exposed beyond the established exposure limit (EL) and margins of safety
Population based studies
LOAEL
a
LOAEL
10
b
LOAEL
100
c
LOAEL
1000
d
= EL
Margin of
safety
(MOS)
Description (Study, Year) N 10 mg/m
3
1 mg/m
3
0.1 mg/m
3
0.01 mg/m
3
50
th
(90
th
)
Athens (Expolis, 96-98) 42 < < 20% 80% no MOS
Basel (Expolis, 96-98) 47 < < < < 16 (9)
Helsinki (Expolis, 96-98) 188 < < < < 22 (9)
Milan (Expolis, 96-98) 41 < < < 5 % 5 (1)
Oxford (Expolis, 98-00) 40 < < < < 12 (4)
Prague (Expolis, 96-98) 46 < < < < 7 (3)
Avg. MOS:
excluding Athens
10 (4)
a
Animal LOAEL ;
b
not considering a NOAEL (10);
c
interspecies variability (10) ;
d
intraspecies variability (10);
< out of the evaluation range (i.e. <5% of the environments investigated)
The INDEX project Final report
138
Acetaldehyde
Synonyms: ethanal, acetic aldehyde, ethyl aldehyde, methyl formaldehyde
CAS Registry Number: 75-07-0
Molecular Formula: C
2
H
4
O
1. Compound identification
Acetaldehyde is a colourless, volatile liquid and at dilute concentrations has a pungent odour. Acetaldehyde is a highly
flammable and reactive compound that is miscible in water and most common organic solvents like alcohol, acetone,
gasoline, toluene, xylene, benzene, ether, paraldehyde. It is used in the manufacture of acetic acid, perfumes, and
flavours. As a liquid, it is lighter than water but the vapours are heavier than air. It is volatile at ambient temperature
and pressure. It is highly reactive and a strong reducing agent. Acetaldehyde can react violently with acid anhydrides,
alcohols, ketones, phenols, ammonia, hydrogen cyanide, hydrogen sulfide, halogens, phosphorus, isocyanates, strong
alkalis, and amines. It is also an intermediate in the metabolism of alcohol (SIS 2003).
2. Physical and Chemical properties
Molecular weight (g/mol) 44.1
Melting point (°C) -123.5
Boiling point (°C) 20.2
Density (g/l at 20 °C, 1 atm)
Relative density (air =1) 1.52
Explosion limits of mixtures (air vol-% acetaldehyde) 4.5-60.5
Solubility: miscible in water and most common solvents
Conversion factors at 20 °C and 760 mm Hg:
1 ppb = 1.829 µg/m
3
1 µg/m
3
= 0.547 ppb
Sources: Hagemeyer (1978), IPCS/CEC (1990), Verschueren 2001.
3. Indoor Air Exposure assessment
Contribution of inhalation exposure to total exposure
Acetaldehyde is a metabolic product of sugars and ethanol. On the basis of the assumptions that a standard drink
contains 10 g of ethanol and that about 90% of imbibed alcohol is metabolized to acetaldehyde, alcoholic beverages are
generally by far the most significant source of exposure to acetaldehyde for the general population (WHO, 1995).
On the basis of the average dietary intake of food groups in different regions of the world and the contents of
acetaldehyde in foodstuffs and non-alcoholic beverages in the Netherlands (Maarse & Visscher, 1992), food
(particularly fruit juices and vinegar) may be one of the principal sources of exposure to acetaldehyde in the general
population. More representative data on mean concentrations in foodstuffs have not been identified, but, on the basis of
the ranges of concentrations determined in the Dutch survey, intake in food is estimated to range from just less than 10
to several hundred µg/kg body weight per day.
On the basis of a daily inhalation volume for adults of 22 m
3
, a mean body weight for males and females of 64 kg, and
the assumption that mean concentrations are approximately 5 µg/m
3
(range: 2 to 8.6 µg/m
3
; Guicheret & Schulting,
The INDEX project Final report
139
1985; Watanabe, 1987; Grosjean, 1991), the mean intake of acetaldehyde from ambient air for the general population is
estimated to be 1.7 µg/kg body weight per day (WHO, 1995).
Emission sources
Typically, concentrations are higher indoors than outdoors due to combustion sources such as cigarettes, fireplaces, and
woodstoves. Acetaldehyde can also be emitted from cooking hamburgers, coffee roasting and from some building
materials such as rigid polyurethane foams, and some consumer products such as adhesives, coatings, lubricants, inks,
and nail polish remover. It is also used as a fruit and fish preservative, flavoring agent, a denaturant for alcohol, for
hardening gelatin fibers, and as a solvent in the synthetic rubber, paraldehyde, tanning and paper industries, in the
manufacture of perfumes, butanol, aniline dyes, plastics, and silvering mirrors (EPA/Cal 2003). Since acetaldehyde is a
product of human metabolism and it is found in exhaled air, it is probable that increased indoor concentrations may be,
in addition to other sources, related to emissions from humans (Jurvelin 2001).
Residences with smokers have two to eight times higher acetaldehyde concentrations than the outdoor mean
concentration (WHO 1995). Based on limited data collected in the US suggest an estimate of an average acetaldehyde
concentration inside residences of 5.4 to 27.0 µg/m
3
(3.0 to 15 ppb). The acetaldehyde concentrations in offices and
public buildings were similar in magnitude to those inside residences. Average and maximum in-vehicle acetaldehyde
concentrations measured in southern California were similar in magnitude to those inside residences (EPA/Cal 2003).
Acetaldehyde is present in vehicle exhaust and in wastes from various industries. Also open burning and incineration of
gas, fuel oil, and coal produce acetaldehyde. Similarly, degradation of hydrocarbons, sewage, and solid biological
wastes produce acetaldehyde (WHO 1995, EPA/Cal 2003).
Indoor air and exposure concentrations
There is a lack of European population based data for acetaldehyde. In Helsinki average indoor air concentrations of
acetaldehyde are clearly higher than ambient concentrations, 18.2 µg/m
3
and 2.7 µg/m
3
respectively. Similarly, indoor
concentrations were higher than personal exposures suggesting the presence of considerably strong indoor sources.
Cumulative distributions of the indoor and 48-hour personal exposure concentrations in Helsinki are presented in Figure
3.1 and Figure 3.2.
In Paris (Figure 3.1) in recently refurbished ats (a non representative study) indoor concentrations measured in bed
rooms were at the same level than in Helsinki (n= 61, GM = 10.2 µg/m
3
, GSD = 1.8 (Clarisse et al 2003).
The concentration of acetaldehyde in 14 homes and a small office building in northern Italy ranged from 1 to 48 µg/m
3
with a mean value of 17 µg/m
3
(DeBortoli M et al, 1986). The ratio of the minimum, maximum, mean, and median
concentration in indoor versus outdoor air was 0.5, 24, 6.0, and 3.6, respectively. In Sweden Gustafsson et al (2003)
reported mean values ranging from 12 µg/m
3
to 14 µg/m
3
in non-smoking homes. In 1986 - 1987, the mean yearly
exposure to acetaldehyde from air pollution in Sweden was 1.0 µg/m
3
(Bostrom CE et al 1994).
Lee and Wang (2004) studied emissions of burning ten types of incense in chamber (18 m
3
) tests. Acetaldehyde
concentrations of the nine incense types ranged from 50 to 150 µg/m
3
, one type exceeded a level of 450 µg/m
3
. From
these results we can conclude that incense burning may increase dramatically indoor air concentrations of acetaldehyde
compared to the ‘normal’ residential levels. Few short time peak concentrations are presented in Table 3.1.
In Denmark (Granby and Kristensen 1997) and Finland (Viskari et al 2000), ambient acetaldehyde showed considerably
low average (3-8 months periods) ambient concentrations being 1.8 µg/m
3
and 1.1-3.2 µg/m
3
, respectively. In Italy and
Greece, much higher average concentrations were measured, being 8.2 µg/m
3
and 15.1 µg/m
3
, respectively (Possanzini
et al 1996, Bakeas et al 2003).
In the USA ambient concentrations ranged from 2.0 to 5.7 µg/m
3
(Grosejan et al 1993, CARB 1999). The concentration
of acetaldehyde measured in EPA headquarters building in Washington, DC ranged from 3.8 to 11.1 µg/m
3
with a
median value of 5.2 µg/m
3
(EPA 1990). High concentrations were found in Brazil (Grosejan et al 2002), Japan,
(Satsumabayashi et al 1995), and in Mexico (Baez et al 1995), being 10.4 µg/m
3
, 2.3-12 µg/m
3
, and 28.6 µg/m
3
,
respectively. In Chinese studies ambient levels ranged from 1.7 µg/m
3
to 7.6 µg/m
3
. The mean indoor concentration of
acetaldehyde in hotels was tenfold compared to outdoor levels, identified as a consequence of smoking indoors (Ho et al
2002, Feng et al 2004).
The INDEX project Final report
140
0
20
40
60
80
100
0 1020304050
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Hel
Par
Fre
Figure 3.1. Cumulative frequency distributions of indoor air concentrations of acetaldehyde in Helsinki (Hel, n= 15,
Jurvelin et al 2001), in the French Survey (Fre, n= 201, Kirchner 2004) and in a non-representative study in Paris (Par)
(n= 61, Clarisse et al 2003).
0
20
40
60
80
100
010203040
Exposure (µg/m
3
)
Cumulative frequency (%)
Hel
Figure 3.2. Cumulative frequency distribution of 48-hour personal exposure concentrations of acetaldehyde in Helsinki
(Hel) (n= 15, Jurvelin et al 2001).
The INDEX project Final report
141
Table 3.1. Short time acetaldehyde concentrations related to specific microenvironments or emission sources.
Averaging time Reference
A
MGMmax
Insence burning during burning 50-450 Lee and Wang 2004
Tavern tobacco smoke 203 EPA 2003
Houses in US manufactured 30-min 5 - 34 18 Hodgson et al 2000
site-built 30-min 21 - 103 36
Exposure to ETS non-smoker living with a smoke
r
1-da
y
7.6 Nazaroff and Singer (2002)
AM = arithmetic mean, GM = geometric mean
Environment or
emission source
Concentration (µg/m
3
)
4. Toxicokinetics
Absorption
Consistent with effects being observed primarily at the initial site of exposure following inhalation (i.e., in the
respiratory tract), available data indicate that the greatest proportion of inhaled acetaldehyde is retained at the site of
contact, becoming rapidly and irreversibly bound to free protein and non-protein sulphydryl groups (notably, cysteine
and glutathione). The results of pharmacokinetic studies conducted in humans (Dalhamn et al., 1968; Egle, 1970) and
rodents (David and Heck, 1983; Morris, 1997) indicate that the absorption of acetaldehyde into the systemic circulation
is likely not extensive following inhalation.
The percentage of acetaldehyde retained by 8 volunteers inhaling acetaldehyde vapour (100-800 mg/m
3
) from a
recording respirometer ranged from 45 to 70%, at different respiratory rates. Total respiratory tract retention was the
same whether the vapour was inhaled through the nose or the mouth. A direct relationship was found between the
contact time and uptake, independent of rate. Thus, the critical factor in determining acetaldehyde uptake is the duration
of the ventilatory cycle (Egle, 1970).
Distribution
Following inhalation by rats, acetaldehyde is distributed to the blood, liver, kidney, spleen, heart, and other muscle
tissues (Hobara et al., 1985; Watanabe et al., 1986). Low levels were detected in embryos after maternal intraperitoneal
injection of acetaldehyde (mouse) and following maternal exposure to ethanol (mouse and rat) (Blakley and Scott,
1984).
Distribution of acetaldehyde to brain interstitial fluid, but not to brain cells, has been demonstrated following
intraperitoneal injection of ethanol in rats. A high affinity, low Michaelis constant ALDH (aldehyde dehydrogenase)
may be important in maintaining low levels of acetaldehyde in the brain during the metabolism of ethanol (WHO,
1995)
. Acetaldehyde is taken up by red blood cells and, following ethanol consumption in humans and in baboons, in
vivo, intracellular levels can be 10 times higher than plasma levels (Baraona et al., 1987)
Small amounts of acetaldehyde are produced endogenously during the normal intermediary catabolism of deoxyribose
phosphate and various amino acids (Nicholls et al., 1992; Jones, 1995). Consumption of alcoholic beverages is also an
important source of acetaldehyde in the body, formed through the metabolism of ethanol by alcohol dehydrogenase.
Studies using the perfused human placental cotyledon indicated that the human placenta has the potential to produce
acetaldehyde, which can enter the fetal circulation. Furthermore, partial transfer of acetaldehyde from maternal to fetal
blood may occur (Karl et al., 1988).
Metabolism and elimination
Based on the high degree of retention of acetaldehyde in the respiratory tract following inhalation in humans, it is likely
that the predominant pathway for the metabolism of acetaldehyde involves conjugation to thiols (i.e., cysteine and
glutathione) at the site of exposure, subsequent formation of hemimercaptal or thiazolidine intermediates, and
elimination of thioethers and disulphides in the urine (Sprince et al., 1974; Cederbaum and Rubin, 1976; Hemminiki,
1982; Brien and Loomis, 1983; Nicholls et al., 1992).
The INDEX project Final report
142
Inhaled acetaldehyde is also rapidly oxidized to acetate by aldehyde dehydrogenase (ALDH) in human nasal and lung
epithelia (Bogdanffy et al., 1986; Yin et al., 1992; Morris et al., 1996). Acetate enters the citric acid cycle as acetyl-
CoA. ALDH activity has been localized in the respiratory tract epithelium (excluding olfactory epithelium) in rats
(Bogdanffy et al., 1986), in the renal cortex and tubules in the dog, rat, guinea-pig, and baboon (Michoudet and Baverel,
1987a; 1987b), and in the testes in the mouse (Anderson et al.,1985).
There are several isoenzymes of ALDH with different kinetic and binding parameters that influence acetaldehyde
oxidation rates (Marjanen, 1973; Parrilla et al 1974, Teschke et al., 1977). Human liver ALDH consists of at least 4
main isoenzymes, which are also present in many other tissues (Koivula, 1975; Goedde et al., 1979). Subjects
homozygous or heterozygous for a point mutation in the mitochondrial ALDH corresponding gene have low activity of
this enzyme, metabolize acetaldehyde slowly, and are intolerant of ethanol (Yoshida et al., 1984; Goedde and Agarwal,
1986; Hsu et al., 1978).
Acetaldehyde is metabolized by mouse and rat embryonic tissue in vitro (Priscott and Ford, 1985). Though
acetaldehyde is metabolized in a dose-dependent manner in human renal tubules (Michoudet and Baverel, 1987b), the
liver is the most important metabolic site. The metabolism of acetaldehyde can be inhibited by crotonaldehyde,
dimethylmaleate, phorone, disulfiram, and calcium carbamide (WHO, 1995; IARC 1985).
5. Health effects
Effects of short-term exposure
Humans exposed acutely to moderate concentrations of acetaldehyde experience irritation of the eyes and respiratory
tract and altered respiratory function. Animals exposed to moderate to high concentrations exhibit skin and eye irritation
and notable cellular alterations in the respiratory epithelium and hyper- keratosis of the forestomach.
A group of 12 human subjects were exposed to acetalclehycle vapour for 15 min while being shown a movie "to divert
their attention". Most of the subjects developed eye irritation at 90 mg/m
3
(50 ppm), but it took more than 360 mg/m
3
(200 ppm) to cause nose or throat irritation in the majority of the subjects (Silverman et al., 1946). Several subjects
strenuously objected to the vapor at as low as 45 mg/m
3
(25 ppm). Even those who reported no eye irritation at 90
mg/m
3
showed erythematous eyelids and bloodshot eyes when exposed to 360 mg/m
3
of acetalctehyde (Silverman et al.,
1946).
A group of 14 "healthy male" volunteers, aged 18-45 years, were exposed in a 100 m
3
chamber to a measured
concentration of acetaldehyde vapour of 240 mg/m
3
(134 ppm) for 30 min. This concentration was said to be mildly
irritating to the upper respiratory tract. No other clinical signs were reported (Sim and Pattle, 1957).
Intravenous infusion of human subjects with 5% acetaldehyde at a rate of 20.6-82.4 mg/min for up to 36 minutes (the
lowest dose converts to 10.6 mg/kg over 36 minutes) resulted in an increased heart rate, increased ventilation rates and
respiratory dead space, and a decreased alveolar carbon dioxide level (Asmussen et al., 1948; IARC 1985).
Summary of short-term exposure effect levels
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
45
Study
Eye irritation Human volunteers,
15min; poor quality of
data
Silverman et al., 1946 WHO 1995;
TC: 2
45
Study
11.5
1h-ADJ
Eye irritation Human volunteers,
15min; poor quality of
data
Silverman et al., 1946 OEHHA 1999;
HPC: 0.115
Final statement (UNIMI) : Human LOAEL: 5 mg/m³, Animal NOAEL: 50 mg/m³, no evidence in considering acute and chronic effect levels separately
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
WHO (1995)
International Programme on Chemical Safety - Environmental Health Criteria 167
Approach to risk assessment: The following guidance is provided as a potential basis for derivation of limits of
exposure by relevant authorities. Though the principal sources of exposure to acetaldehyde in the general population are
The INDEX project Final report
143
through the metabolism of alcohol, in cigarette smoke, and food, air is believed to be the main route of exposure in the
occupational environment.
On the basis of data on irritancy in humans, a tolerable concentration can be derived as follows:
Tolerable concentration = 45 mg/m
3
: 20 = 2 mg/m
3
(2000 µg/m
3
) where no effects were observed in a limited study on
human volunteers at 45 mg/m
3
(Silverman et al., 1946) and 20 is the uncertainty factor (×10 for intraspecies variation
and × 2 for the poor quality of the data).
OEHHA (1999b)
Office of Environmental Health Hazard Assessment
Derivation of Health Protective Concentration (draft)
Since adopted health assessment values suitable for assessing potential health impacts from short-term inhalation
exposures are not available for acetaldehyde, OEHHA calculated a draft health protective concentration (HPC). In
calculating the HPC, OEHHA followed the risk assessment methodology used for developing the acute reference
exposure levels (RELs) under the Air Toxics Hot Spots Program risk assessment guidelines process (OEHHA, 1999a).
As mandated by state legislation, these guidelines underwent scientific and public peer review, prior to approval by the
Scientific Review Panel (Senate Bill 1731, Statutes of 1992, Ch. 1162 of the California Health and Safety Code) and
adoption by OEHHA.
Silverman et al. (1946) found that a 15-minute exposure to acetaldehyde induced eye irritation in male and female
human volunteers at a concentration of 45 mg/m3 (25 ppm). This concentration was identified as a lowest-observed-
adverse-effect level (LOAEL); a no-observed-adverse-effect level (NOAEL) was not observed in this study.
Application of “Haber’s Law” to extrapolate from a 15 minute exposure to 1 hour results in an adjusted LOAEL of 11.5
mg/m3 (6.5 ppm).
An acute 1-hour draft HPC can be calculated for acetaldehyde using the formula:
Draft HPC = LOAEL / UF = 11.5 mg/m3 / 100 = 115 µg/m3 (65 ppb).
The uncertainty factor (UF) for this calculation is 100, which incorporates uncertainty contributions for extrapolation
from a LOAEL to a NOAEL (10) and for potentially sensitive human subpopulations (10).
Effects of long-term exposure
The critical health effects arising from exposure to airborne acetaldehyde are eye and upper respiratory tract irritation
with the possibility of chronic tissue damage and inflammation in the respiratory tract following long-term exposure.
Such long-term inflammatory changes have been associated with tumour in the nasal passages of experimental animals.
The underlying mechanism for tumour formation is considered to be sustained irritation/inflammation accompanied by
cellular proliferation. Such a mechanism is consistent with a threshold for tumour formation and thus control to prevent
local tissue damage and inflammation would be predicted to control for any risk of tumour formation.
No information is available in humans regarding chronic tissue inflammation, the only data available relate to sensory
irritation. The only objective evidence of eye irritation in humans was with a single exposure to 360 mg/m
3
. There are
reports of subjective irritation symptoms at lower concentrations (down to 45 mg/m
3
), although there is uncertainty
regarding the reliability of these observations.
In short-term inhalation studies (> 4 weeks) conducted in rodents, degenerative changes (including inflammation,
hyperplasia, thinning and disarrangement of epithelial cells, and loss of microvilli and sensory cells) and associated
functional effects were observed in the nasal olfactory epithelium in rats exposed (by inhalation) to 437 mg/m
3
acetaldehyde, while degenerative changes in the nasal respiratory epithelium, larynx, trachea and lungs were observed
at higher concentrations (i.e. 1800 mg/m
3
) (Appelman et al., 1982, 1986; Saldiva et al., 1985; Cassee et al., 1996).
In the only subchronic inhalation study identified, in which hamsters were exposed to acetaldehyde for 13 weeks
(Kruysse et al., 1975), non-neoplastic lesions in the tracheal epithelium (including stratification, keratinization,
inflammation, metaplasia and granulation) were observed at 2412 mg/m
3
acetaldehyde (considered to be the LOAEL),
while histopathological lesions in the nasal cavities, larynx, bronchi and lungs were observed only at 8208 mg/m
3
acetaldehyde; the NOEL in this study was considered to be 702 mg/m
3
acetaldehyde (Kruysse et al., 1975).
In chronic inhalation studies in which rats were exposed to acetaldehyde for up to 28 months, focal basal cell
hyperplasia of the nasal olfactory epithelium was observed at 1350 mg/m
3
acetaldehyde (considered to be the
The INDEX project Final report
144
LOAEL), while non-neoplastic lesions in the nasal respiratory epithelium (squamous metaplasia, papillomatous
hyperplasia, and focal or pseudoepitheliomatous hyperplasia) and larynx (squamous metaplasia and hyperplasia) were
observed at concentrations 2700 mg/m
3
acetaldehyde (Woutersen et al., 1984, 1986; Feron et al., 1985; Woutersen and
Feron, 1987). Similarly, nonneoplastic lesions in the nasal epithelia, larynx and trachea have been observed in hamsters
exposed by inhalation to 2700 mg/m
3
acetaldehyde for 52 weeks (Feron, 1979; Feron et al., 1982).
The studies that provide best characterization of concentration–response for the critical effects in the most sensitive
species (i.e., rats) are the short-term studies of Appelman et al. (1982; 1986). While these studies are short-term in
duration, together they establish a concentration-response for lesions after only 4 weeks of exposure. These same types
of lesions appear at longer exposure times and higher exposure levels in chronic studies (Wouterson et al., 1986;
Wouterson and Feron, 1987; Kruysse et al., 1975). Under other circumstances, studies of short duration may not be
considered appropriate, but for acetaldehyde the observed effects are consistent with pathology seen in long-term
studies.
Appleman et al. (1986) conducted two inhalation studies on male Wistar rats (10/group) exposing them 6 hours/day, 5
days/week for 4 weeks to 0, 273 and 910 mg/m
3
(0, 150, and 500 ppm respectively). Duration-adjusted concentrations
are 0, 48.75, and 162.5 mg/m
3
, respectively. One group was exposed without interruption, a second group was
interrupted for 1.5 hours between the first and second 3-hour period, and a third group was interrupted as described with
a superimposed peak exposure profile of 4 peaks at 6-fold the basic concentration per 3-hour period. The purpose was
to test intermittent and peak exposure effects. Urine samples were collected from all rats and lung lavage performed on
4-5 per group at the end of the experiment. Cell density, viability, number of phagocytosing cells, and phagocytic index
were determined on the lavage fluid. Microscopic examination was performed on the nasal cavity, larynx, trachea with
bifurcation and pulmonary lobes of all rats of all groups.
Continuous and interrupted exposure to 910 mg/m
3
did not induce any visible effect on general condition or behavior,
but peak exposures at this level caused irritation. No behavioral differences were noted in the other groups. Mean body
weights of the group exposed to 910 mg/m
3
with interruption and with peak exposures were statistically significantly
lower than those of the controls. Body weights were similar to controls in the other exposure groups. Mean cell density
and cell viability were significantly decreased in the group exposed to 910 mg/m
3
with or without peak exposures. The
mean percentage of phagocytosing cells and the phagocytic index were significantly lower than controls in all groups
exposed to 910 mg/m
3
, especially the group exposed to superimposed peaks. Histopathological changes attributable to
exposure were found only in the nasal cavity. Degeneration of the olfactory epithelium was observed in rats exposed to
910 mg/m
3
. Interruption of the exposure or interruption combined with peak exposure did not visibly influence this
adverse effect. No compound-related effects were observed in rats interruptedly or uninterruptedly exposed to 273
mg/m
3
during the 4-week exposure period; therefore, the NOAEL is 273 mg/m
3
.
Appelman et al. (1982) exposed Wistar rats (10/sex/group) for 6 hours/day, 5 days/week for 4 weeks to 0, 728, 1820,
4004 and 9100 mg/m
3
acetaldehyde (0, 400, 1000, 2200, or 5000 ppm, respectively). Duration-adjusted concentrations
are 0, 130, 325, 715 and 1625 mg/m
3
, respectively. The general condition and behavior of the rats were checked daily.
Blood picture (Hb, Hct, RBC, total and differential WBC, and plasma protein) and chemistry were examined at the end
of the treatment period. Activities of plasma glutamic-oxalacetic transaminase, glutamic-pyruvic transaminase, and
alkaline phosphatase were also determined. Urine was analyzed for density, volume, pH, protein, glucose, occult blood,
ketones, and appearance. The kidneys, lungs, liver, and spleen were weighed. Microscopic examination was performed
on the lungs, trachea, larynx, and nasal cavity (3 transverse sections) of all animals and on the kidneys, liver, and spleen
of all control and high- concentration groups.
During the first 30 minutes of each exposure at the 9100 mg/m
3
level, rats exhibited severe dyspnea that gradually
became less severe during the subsequent exposure period. Two animals died at this level (1 female, 1 male) and one
male died at the 4004 mg/m
3
level, but the cause of death could not be determined due to autolysis or cannibalism.
Growth was retarded in males at the three highest exposure concentrations and in females at the 9100 mg/m
3
level. The
percentage of lymphocytes in the blood was lower and the percentage of neutrophilic leukocytes higher in males and
females of the 9100 mg/m
3
group than in controls. There were a few statistically significant differences in several blood
chemistry parameters between the exposure groups and the control group but none of them were concentration-related.
Statistically significant changes in organ-to-body weight ratios included decreased liver weights in both sexes and
increased lung weights in males at the 9100 mg/m
3
level. Males in the 9100 mg/m
3
level produced less urine, but it was
of higher density. Compound-related histopathological changes were observed only in the respiratory system. The nasal
cavity was most severely affected and exhibited a concentration-response relationship. At the 728 mg/m
3
level,
compound-related changes included: slight to severe degeneration of the nasal olfactory epithelium, without hyper- and
metaplasia, and disarrangement of epithelial cells. At the 1820- and 4004 mg/m
3
levels, more severe degenerative
changes occurred, with hyperplastic and metaplastic changes in the olfactory and respiratory epithelium of the nasal
cavity. Degeneration with hyperplasia/metaplasia also occurred in the laryngeal and tracheal epithelium at these levels.
The INDEX project Final report
145
At 9100 mg/m
3
changes included severe degenerative hyperplastic and metaplastic changes of the nasal, laryngeal, and
tracheal epithelium. Based on the degenerative changes observed in the olfactory epithelium, the 728 mg/m
3
level is
designated as a LOAEL.
Available data are inadequate to assess the potential reproductive, developmental, neurological or immunological
effects of direct exposure to acetaldehyde. Based on the limited number of investigations conducted to date, however,
reproductive, developmental, neurological and immunological effects have not been observed at concentrations below
those that induce damage in the upper respiratory tract (Ortiz et al., 1974; Kruysse et al., 1975; Shiohara et al., 1985;
Aranyi et al., 1986; Roumec et al., 1988).
Summary of long-term exposure effect levels
Health Canada and U.S. EPA have evaluated the noncancer inhalation toxicity data for acetaldehyde. Health Canada
derived a tolerable concentration of 0.39 mg/m
3
. EPA derived a reference concentration (RfC) of 0.009 mg/m
3
, which is
40-fold lower than the Health Canada value. Both agencies used the same studies, but differed in their estimation. While
EPA used the NOAEL of 273 mg/m
3
from Appleman et al. (1982), Health Canada used the lower 95% confidence limit
on a benchmark dose (BMD) for the concentration associated with a 5% increase in the incidence of nasal olfactory
epithelial lesions for male rats [from data in the studies by Appleman et al. (1982, 1986)]. Both agencies adjusted for
the intermittent exposure in the laboratory, but EPA also adjusted to a Human Equivalent Concentration (HEC) by
calculating for a gas:respiratory effect in the extrathoracic region. The agencies also differed in choice of uncertainty
factor, with EPA utilizing three factors of ten (intraspecies, use of a subchronic study, and interspecies and database
combined for a third ten). Health Canada did not use a factor for limitations in the database due to the fact that a TC that
is based on critical effects at the site of entry is likely to be protective for systemic effects. A factor for use of a shorter-
term study was not deemed appropriate because there was no indication that severity of the critical effects increases
with duration of exposure. Click on the green circle(s) for more information.
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
273
EXP
49
ADJ
8.7
HEC
728
EXP
130
ADJ
16.9
HEC
Degeneration of
olfactory epithelium
Rat, 4w Appleman et al.,
1986;1982
EPA-IRIS 1991;
RfC: 0.003
218
BC05
39
ADJ
Degeneration of
olfactory epithelium
Rat, 4w Appleman et al.,
1986;1982
Health Canada
2000; TC: 0.39
- - Respiratory system not specified OEHHA 1995;
REL: 0.009
Although the studies cited establish a concentration-response for lesions after only 4 weeks of exposure, same types of lesions appear at longer exposure
times and higher exposure levels, and are consistent with pathology seen in chronic studies.
Final statement (UNIMI) : Human LOAEL: 5 mg/m³, Animal NOAEL: 50 mg/m³, no evidence in considering acute and chronic effect levels separately
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
U.S.EPA - IRIS (1991)
Integrated Risk Information System
Determination of the Reference Concentration for Chronic Inhalation Exposure (RfC)
Critical effect Exposures* UF MF RfC
Degenration of
olfactory epithelium
Short-term Rat
Inhalation Studies
Appleman et al.,
1986;1982
NOAEL: 273 mg/m
3
(150 ppm)
NOAEL:(ADJ): 48.75 mg/m
3
NOAEL(HEC): 8.7 mg/m
3
LOAEL: 728 mg/m
3
(400 ppm)
LOAEL(ADJ): 130 mg/m
3
LOAEL(HEC): 16.9 mg/m
3
1000 1
0.003 mg/m
3
*Conversion Factors -- MW = 44.5. Appleman et al., 1986: Assuming 25C and 760 mmHg, NOAEL(mg/cu.m) = 150 ppm x 44.5/24.45 = 273.
NOAEL(ADJ) = 273 mg/cu.m x 6 hours/day x 5 days/7 days = 48.75 mg/cu.m. The NOAEL(HEC) was calculated for a gas:respiratory effect in the
ExtraThoracic region. MVa = 0.23 cu.m/day, MVh = 20 cu.m/day, Sa(ET) = 11.6 sq. cm, Sh(ET) = 177 sq. cm. RGDR(ET) = (MVa/Sa) / (MVh/Sh) =
0.18. NOAEL(HEC) = NOAEL(ADJ) x RGDR = 8.7 mg/cu.m.
Appleman et al., 1982: Assuming 25C and 760 mmHg, LOAEL(mg/cu.m) = 400 ppm x 44.5/24.45 = 130. LOAEL(ADJ) = 728 mg/cu. m x 6 hours/day
x 5 days/7days = 130 mg/cu.m. The LOAEL(HEC) was calculated for a gas:respiratory effect in the ExtraThoracic region. MVa = 0.17 cu.m/day,
The INDEX project Final report
146
MVh = 20 cu.m/day, Sa(ET) = 11.6 sq. cm., Sh(ET) = 177 sq.cm. RGDR(ET) = (MVa/Sa) / (MVh/Sh) = 0.13. LOAEL(HEC) = LOAEL(ADJ) x RGDR
= 16.9 mg/cu.m.
Two short-term studies conducted by the same research group are the principal studies used. While these studies are
short-term in duration, together they establish a concentration-response for lesions after only 4 weeks of exposure.
These same types of lesions appear at longer exposure times and higher exposure levels in chronic studies (Wouterson
et al., 1986; Wouterson and Feron, 1987; Kruysse et al., 1975). Under other circumstances, studies of short duration
may not be considered appropriate, but for this chemical the observed effects are consistent with pathology seen in
long-term studies. The 150-ppm exposure level was therefore established as the NOAEL from the Appleman et al.
(1986) study and the LOAEL from the Appleman et al. (1982) study.
Uncertainty factors (UF) for the determination of the Inhalation RfC: An uncertainty factor of 10 was applied to account
for sensitive human populations. A factor of 10 was applied for both uncertainty in the interspecies extrapolation using
dosimetric adjustments and to account for the incompleteness of the data base. A factor of 10 was applied to account for
subchronic to chronic extrapolation.
Confidence in the Inhalation RfC: Confidence in the principal studies (Appleman et al., 1982; 1986) is medium since
appropriate histopathology was performed on an adequate number of animals and a NOAEL and LOAEL were
identified, but the duration was short and only one species was tested. Confidence in the data base is low due to the lack
of chronic data establishing NOAELs and due to the lack of reproductive and developmental toxicity data. Low
confidence in the RfC results.
Health Canada (2000)
Canadian Environmental Protection Act (CEPA).
Derivation of a Tolerable Concentration (TC):
A tolerable concentration (TC) has been developed on the basis of Benchmark Concentration (BMC) for degeneration
in the nasal olfactory epithelium of Wistar rats exposed to acetaldehyde (by inhalation) for four weeks, using combined
data from Appelman et al. (1982, 1986).
For many types of effects, studies of short duration are not preferred as the basis for development of a TC. While the
data were derived from short-term studies, the incidence of degenerative changes in the olfactory epithelium was not
dissimilar to that observed in the same strain of rats in the long-term carcinogenesis bioassay at similar concentrations,
conducted by Woutersen et al. (1986). Although group sizes were larger in the long-term bioassay, concentration-
response for these lesions was not well characterized because of the small number of dose groups exposed to higher
concentrations compared with the short-term study and early mortality among animals at the highest concentration.
Data in the Woutersen et al. (1986) study are insufficient to serve as a basis for development of a meaningful BMC for
acetaldehyde, even simply for purposes of comparison.
Using the THRESH program (Howe, 1995), the BMC
05
(the concentration associated with a 5% increase in the
incidence of nasal olfactory epithelial lesions) for male Wistar rats is 357 mg/m
3
; the lower 95% confidence limit for
this value (BMCL
05
) is 218 mg/m
3
. For female Wistar rats, the BMC
05
and BMCL
05
are 445 mg/m
3
and 17 mg/m
3
,
respectively.
The BMCL
05
of 218 mg/m
3
was adjusted from an intermittent to a continuous exposure by multiplying the value by 6/24
and 5/7. There are no data that provide direct evidence and few related data to serve as a basis for whether or not such
an adjustment is appropriate for acetaldehyde. In short-term studies in the same strain of rats, effects seemed slightly
more severe following exposure for eight hours per day (Saldiva et al., 1985) versus six hours per day for four weeks
(Appelman et al., 1986). Interruption of daily exposure by 1.5-hour exposure-free periods or by the superimposition of
eight five-minute peak exposure periods did not appreciably influence the cytotoxic potency of acetaldehyde in short-
term studies in rats compared with uninterrupted exposure to a fixed concentration (Appelman et al., 1986).
An uncertainty factor of 100 was used (10 for interspecies variation, 10 for intraspecies variation). Available data are
inadequate to further address toxicokinetic and toxicodynamic aspects of components of uncertainty with data-derived
values. The value for interspecies variation is considered to be conservative since, due to greater penetration of inhaled
gases into the lower airways of rodents versus humans, the compound is distributed over a larger surface area for the
latter; available data are inadequate, however, to quantitatively account for this variation. No additional quantitative
element has been included to address limitations of the database such as lack of adequate developmental or reproductive
studies by a relevant route of exposure, due to the fact that a TC that is based on critical effects at the site of entry is
likely to be protective for systemic effects. Also, in view of the fact that there is no indication that severity of the critical
The INDEX project Final report
147
effects increases with duration of exposure, an additional quantitative element to address the use of a shorter-term study
as the basis for the TC is considered inappropriate.
The resulting Tolerable Concentration (TC) is 0.39 mg/m
3
. This TC is similar to that derived from the NOEL for
irritation in the study of Appelman et al. (1986) (TC of 0.49 mg/m
3
). On the basis of limited available data in human
studies, the TCs derived above (0.39 and 0.49 mg/m
3
) are two orders of magnitude lower than the threshold for sensory
irritation (i.e., 45 mg/m
3
[25 ppm]) (Silverman et al., 1946).
The degree of confidence in the database on toxicity that serves as the basis for the development of the tolerable
concentration (TC) for inhalation is moderate, although there is a relatively high degree of confidence that critical
effects occur at the initial site of exposure. Despite differences in the anatomy and physiology of the respiratory tract in
rats and humans, respiratory tract defense mechanisms are similar. Thus, it is reasonable to assume that the response of
the human respiratory tract mucosa to acetaldehyde will be qualitatively similar to that of experimental species,
although the likely site of development of lesions may vary due to oro-nasal breathing patterns in humans, which result
in greater potential to deliver acetaldehyde to the lower respiratory tract.
WHO (1995)
Environmental Health Criteria 167
Data suggest that acetaldehyde causes genetic damage to somatic cells in vivo. The irritancy of acetaldehyde may also
play an important role in the development of tumours in the nose and larynx of rats and hamsters, respectively, exposed
by inhalation, though all concentrations of acetaldehyde administered in carcinogenesis bioassays induced both irritancy
and nasal tumours. Therefore, two approaches were adopted for the provision of guidance with respect to the potential
carcinogenicity of acetaldehyde.
1.) In the first, a tolerable concentration (TC) was derived on the basis of division of an effect level for irritancy in the
respiratory tract of rodents by an uncertainty factor, based on the principles outlined in WHO and the assumption that
there is a threshold for acetaldehyde-induced cancer of the respiratory tract in rodents exposed via inhalation. There is
some support for this approach on the basis of relevant data on the analogues of acetaldehyde, i.e., formaldehyde and
glutaraldehyde, which have similar spectra of in vitro mutagenic effects, but are clearly not mutagenic in vivo (IARC,
1985; WHO, 1989). Thus,
Tolerable concentration = 275 mg/m
3
: 1000 = 0.3 mg/m
3
(300 µg/m3)
where: 275 mg/m
3
was the NOEL for irritation in rats in a 4-week study (Appelman et al., 1986) and 1000 is the
uncertainty factor (×10 for interspecies variation, ×10 for intraspecies variation and ×10 for a less than long-term study
and severity of effect, i.e., carcinogenicity associated with irritation).
2.) Since the mechanism of induction of tumours by acetaldehyde has not been well studied, lifetime cancer risk has
also been estimated on the basis of a default model (i.e., linearized multistage). However, it is very likely that, since
estimated risk is based on tumour incidence at concentrations that induce irritancy in the respiratory tract and no-
observed-effect levels for irritancy are well below these concentrations, the true cancer risk is most likely much lower at
concentrations generally present in the environment and may, indeed, be zero.
Concentrations associated with a 10
-5
excess lifetime risk (lower 95% confidence limits) for nasal tumours
(adenocarcinomas, squamous cell carcinomas, and carcinomas in situ) in male and female rats, in the only
carcinogenicity study in which animals were exposed via inhalation to acetaldehyde over the lifetime (Woutersen et al.,
1986), calculated on the basis of the linearized multistage model (Global 82), are 11-65 µg/m
3
. The high-dose animal
groups were excluded in the derivation of these estimates because of early mortality. A body surface area correction
was not incorporated.
OEHHA (1993)
Office of Environmental Health Hazard Assessment
A chronic non-cancer Reference Exposure Level (REL) of 9.0 µg/m is listed for acetaldehyde in the California Air
Pollution Control Officers Association Air Toxics “Hot Spots” Program, Revised 1992 Risk Assessment Guidelines.
The toxicological endpoint considered for chronic toxicity is the respiratory system.
The INDEX project Final report
148
Carcinogenic potential
Acetaldehyde is reasonably anticipated to be a human carcinogen based on sufficient evidence of carcinogenicity in
experimental animals (IARC 1985, 1987, 1999). When administered by inhalation, acetaldehyde increased the
incidence of squamous cell carcinomas and adenocarcinomas in the nasal mucosa in rats of both sexes and laryngeal
carcinomas in hamsters of both sexes. In another inhalation study using a lower exposure level and in an intratracheal
instillation study, no increased incidence of tumors in hamsters was observed. When administered by inhalation,
acetaldehyde enhanced the incidence of respiratory tract tumors as induced by intratracheal instillation of
benzo[a]pyrene in hamsters of both sexes.
There is inadequate evidence for the carcinogenicity of acetaldehyde in humans (IARC 1985, 1987, 1999). A single
study of workers in an aldehyde plant reported nine cases of cancer, including five cases of bronchial tumors and two
cases of carcinomas of the oral cavity. This study was considered to be inadequate for evaluation because of mixed
exposure, the small number of cases, and the poorly defined population. Three case control studies investigated the risk
of oral, throat, and esophageal cancers following heavy alcohol intake. These studies consistently showed an increased
risk of these cancers in people with genetic polymorphisms; these polymorphisms resulted in higher blood acetaldehyde
concentrations after drinking alcohol (IARC 1999). Overall evaluation (IARC 1999): Acetaldehyde is possibly
carcinogenic to humans (Group 2B; there is inadequate evidence in humans for the carcinogenicity of acetaldehyde.
There is sufficient evidence in experimental animals for the carcinogenicity of acetaldehyde).
Acetaldehyde is considered a probable human carcinogen (Classification: B2; EPA 1998), based on increased incidence
of nasal tumors in male and female rats and laryngeal tumors in male and female hamsters after inhalation exposure.
Health Canada (2000) and EPA (1998) each used the same study (Woutersen et al., 1986; Woutersen and Appelman,
1984) and tumors (nasal adenocarcinomas and squamous cell carcinomas) to calculate cancer potency, but their
approaches differ in that Health Canada does not do low dose extrapolation. Health Canada calculated a tumorigenic
concentration with 5% response (TC
05
) of 86 mg/m
3
, with a lower 95% confidence limit (TCL
05
) of 28 mg/m
3
. EPA
estimated an inhalation unit risk of 2.2 E-6 per µg/m
3
.
Summary of principal study: The carcinogenicity of acetaldehyde was studied in 420 male and 420 female albino SPF
Wistar rats (Woutersen and Appelman, 1984; Woutersen et al., 1985). After an acclimatization period of 3 weeks, these
animals were randomly assigned to four groups of 105 males and 105 females each. The animals were then exposed by
inhalation to atmospheres containing 0, 1351, 2702, or 5403 mg/m
3
(0, 750, 1500, or 3000 ppm) acetaldehydefor 6
hours/day, 5 days/week, for 27 months. The concentration in the highest dose group was gradually reduced from 5403
to 1801 mg/m
3
because of severe growth retardation, occasional loss of body weight and early mortality in this group.
Interim sacrifices were carried out at 13, 26, and 52 weeks. One tumor was observed in the 52 week sacrifice group and
none at earlier times. Exposure toacetaldehyde increased the incidence of tumors in an exposure-related manner in both
male and female rats. In addition, there were exposure-related increases in the incidences of multiple respiratory tract
tumors. Adenocarcinomas were increased significantly in both male and female rats at all exposure levels, whereas
squamous cell carcinomas were increased significantly in male rats at middle and high doses and in female rats only at
the high dose. The squamous cell carcinoma incidences showed a clear dose-response relationship. The incidence of
adenocarcinoma was highest in the mid-exposure group (2702 mg/m
3
) in both male and female rats, but this was
probably due to the high mortality and competing squamous cell carcinomas at the highest exposure level. In the low-
exposure group, the adenocarcinoma incidence was higher in males than in females.
Genotoxicity
Acetaldehyde is genotoxic in vitro, inducing gene mutations, clastogenic effects, and sister-chromatid exchanges
(SCEs) in mammalian cells in the absence of exogenous metabolic activation (He and Lambert, 1990; Badr and
Hussain, 1977; Obe et al., 1978; 1979; 1985; Veghelyi and Osztovics, 1978; Boehlke, 1983).
The results of in vivo studies suggest that acetaldehyde can react directly with DNA and proteins to form stable adducts.
Acetaldehyde produced a concentration-related reduction in the extractability of DNA (suggestive of increased
formation of DNA–protein cross-links) from the respiratory nasal mucosa of Fischer 344 rats exposed (whole body) to
1800 or 5400 mg/m
3
acetaldehyde for six hours or to 1800 mg/m
3
acetaldehyde for six hours per day for five days.
Significant reduction in the extractability of DNA in the nasal olfactory epithelia was observed only following exposure
to 1800 mg/m
3
acetaldehyde for six hours per day for five days (Lam et al., 1986). There is indirect evidence from in
vitro and in vivo studies to suggest that acetaldehyde can induce protein-DNA and DNA-DNA cross-links (Lam et al.,
1986).
Many of the toxicological effects of acetaldehyde may be due to the saturation of protective cellular mechanisms at the
initial site of exposure. As with formaldehyde, the potential for acetaldehyde to react with epithelial DNA (and other
cellular components) in the upper respiratory tract may be dependent upon the levels of intracellular thiols (notably
The INDEX project Final report
149
glutathione and cysteine), which prevent binding of acetaldehyde with critical sulphydryl groups in proteins, peptides
and DNA (Cederbaum and Rubin, 1976; U.S. EPA, 1987; von Wartburg, 1987).
In addition, regional deficiencies in aldehyde dehydrogenase activity in rats correlate with the distribution of nasal
lesions in another strain of rats exposed to acetaldehyde in inhalation studies (Bogdanffy et al., 1986). Observed
decreases in uptake of acetaldehyde at high concentrations >180 mg/m
3
in a range of species may be a function of
exceedance of the metabolic capacity of nasal aldehyde dehydrogenase (Morris, 1997).
The pattern of observed irritancy of acetaldehyde at the site of contact and the results of studies indicating that it can
react directly with DNA and proteins to form stable adducts is similar to that for other aldehydes (such as
formaldehyde) that have been carcinogenic to the respiratory system in sensitive inhalation bioassays. Although the
exact mechanism is unknown, induction of tumours by these aldehydes is considered to be a function of both
regenerative proliferative response and DNA–protein cross-linking at the site of contact.
Similarly, it has been proposed that the genotoxicity of acetaldehyde is based principally upon its ability to interact with
single-stranded DNA during cell division (Feron et al., 1982, 1984; Woutersen et al., 1986; Roe and Wood, 1992;
DECOS, 1993). Thus, a crucial determinant in the carcinogenicity of acetaldehyde in the nasal passages may be the
cytotoxicity of this substance at high concentrations (Feron et al., 1982, 1984; Woutersen et al., 1986; Roe and Wood,
1992); cytotoxic concentrations of acetaldehyde cause recurrent tissue damage (and the presence of single-stranded
DNA) and possess significant initiating activity. Moreover, the increased cell turnover may strongly enhance the
fixation of relevant DNA damage and subsequently increase the progression of pre-cancer (initiated) cells to cancer.
However, the limited available data indicate that the pattern of DNA–protein cross-linking and proliferative response
induced by acetaldehyde varies from that of other aldehydes, such as formaldehyde. For acetaldehyde, at concentrations
at which tumours are observed 1350 mg/m
3
, there are increases in DNA–protein cross-links in the respiratory and
olfactory mucosa of rats but no increase in proliferation (Cassee et al., 1996a).
For formaldehyde, at the lower concentrations at which tumours are observed 7.2 mg/m
3
, there are increases in DNA–
protein cross-links and proliferation in the nasal respiratory (but not olfactory) epithelium (Casanova et al., 1994).
While acetaldehyde is genotoxic in vitro and in vivo, information concerning the potential roles of cytotoxicity, cell
proliferation and DNA–protein cross-links in tumour formation is lacking.
Interactions with other chemicals
Acetaldehyde is a highly reactive molecule that can react with many other large or small molecules by addition,
condensation, or polymerization. These pathways may have little quantitative significance in acetaldehyde metabolism,
but the by-products may have biological significance.
Acetaldehyde can react with various macromolecules in the body, which can lead to marked alterations in the biological
function of these molecules, as evidenced by inhibition of enzyme activity, impaired histone-DNA binding, and
inhibition of polymerization of tubulin. The best characterized nucleophiles able to form adducts with acetaldehyde are
amino groups, notably the alpha-amino terminus of peptides and proteins and the epsilon-amino group on the side-chain
of lysine residues.
The toxicity studies with mixtures of aldehydes showed that histopathological changes and cell proliferation of the nasal
epithelium induced by mixtures of formaldehyde, acetaldehyde and/or acrolein appeared to be more severe and more
extensive, both in the respiratory and the olfactory part of the nose, than those observed after exposure to the individual
aldehydes at comparable exposure levels. However the combined effect of the mixtures was at most the sum of the
individual effects. Neither dose addition nor potentiating interactions occurred upon exposure to combinations of these
aldehydes at exposure levels slightly below or around the minimal-observed-effect level (MOEL) (Flemming, 1995;
Cassee, 1996).
Metronidazole or cotrimoxazole (antimicrobial agents) may enhance accumulation of acetaldehyde in blood induced by
antialcohol drugs like disulfiram or nitrefazole (Heelon and White, 1998; Cina et al., 1996; Suokas et al., 1985).
Odour perception
Acetaldehyde is a volatile liquid with a pungent, suffocating odour that is fruity in dilute concentrations.
The INDEX project Final report
150
Source: Devos et al.(1990):
Odour threshold: 0.025 mg/m
3
,
Source: Amoore and Hautala (1983)
The odour threshold for acetaldehyde is reported to be 0.09 mg/m
3
(0.05 ppm), a geometric average of all available
literature data
Source: AIHA (1989)
A wide range of values has been reported: 0.005 to 1800 mg/m
3
. An acceptable, critiqued value is 0.12 mg/m
3
(detection)
Source: New Jersey Department of Health and Senior Services
Odour threshold: 0.12 mg/m
3
The INDEX project Final report
151
Summary of Acetaldehyde Dose Response Assessment
________________________________________________________________________________________________
Exposure other than inhalation: Acetaldehyde is a metabolic product of ethanol and sugars. Estimate of diatary
intake: 10 to several hundred µg/kg bw., one std alc.drink: 100 mg/kg bw., ambient air: 2 µg/kg bw.. Consequently,
most toxicological studies in the literature consider systemic effects of acetaldehyde.
Toxicokinetics: The greatest proportion (45-70%) of inhaled acetaldehyde is retained at the site of contact, with
irreversible binding to sulphhydryl groups (e.g.cisteine) or oxidation to acetate (acetyl-CoA). No extensive absorption
into the systemic circulation. Critical factor determining the uptake is the duration of the ventilation cycle, no difference
between nose and mouth breathing.
Health effect levels of short- and long-term exposure (noncancer):
NOAEL
mg/m³
LOAEL
mg/m³
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
Short-term exposure
45
Study
Eye irritation Human volunteers,
15min; poor quality of
data
Silverman et al., 1946 WHO 1995;
TC: 2
45
Study
11.5
1h-ADJ
Eye irritation Human volunteers,
15min; poor quality of
data
Silverman et al., 1946 OEHHA 1999;
HPC: 0.115
Long-term exposure
273
EXP
49
ADJ
8.7
HEC
728
EXP
130
ADJ
16.9
HEC
Degeneration of
olfactory epithelium
Rat, 4w Appleman et al.,
1986;1982
EPA-IRIS 1991;
RfC: 0.003
218
BC05
39
ADJ
Degeneration of
olfactory epithelium
Rat, 4w Appleman et al.,
1986;1982
Health Canada
2000; TC: 0.39
- - Respiratory system not specified OEHHA 1995;
REL: 0.009
Remark on Appleman studies: Although the studies cited establish a concentration-response for lesions after only 4 weeks of exposure, same types of
lesions appear at longer exposure times and higher exposure levels, and are consistent with pathology seen in chronic studies.
Carcinogenicity: IARC: 2B; U.S.EPA: B2, Unit risk (EPA-IRIS): 2.2E-06 (µg/m
3
)
-1
; The underlying mechanism for
tumour formation is considered to be sustained irritation/inflammation accompanied by cellular proliferation, a
mechanism consistent with a threshold for acetaldehyde induced cancer.
Genotoxicity: Genotoxic in vitro and in vivo, though information concerning the potential roles of cytotoxicity, cell
proliferation and DNA-protein cross-links in tumour formation is lacking.
Odour threshold: Pungent, suffocating odour that is fruity at dilute concentrations; Detection: 0.12 mg/m
3
(AIHA);
geomean: 0.09 mg/m
3
(Amoore and Hautala); 0.025 mg/m
3
(Devos)
Susceptible population: Systemic: Low activities of the aldehyde dehydrogenase enzyme (ALDH), occuring in
subjects with point mutations in the corresponding gene, reduce acetaldehyde oxidation rate and conduce to intolerance
for ethanol. Acetaldehyde levels in the blood are induced by antialcohol drugs like disulfiram or nitrefazole.
Remarks: RD
50
: 7 g/m
3
; neither dose addition nor potentiating interactions occured upon exposure to aldehyde
mixtures (formaldehyde, acetaldehyde, acrolein) at exposure levels around the MOEL.
The INDEX project Final report
152
6. Risk Characterization
Cancer risk and health hazard evaluation
Based on short-term and long-term inhalation studies conducted in experimental animals, the upper respiratory tract is
the principal target site for effects of inhaled acetaldehyde. In short-term studies acetaldehyde caused degenerative non-
neoplastic effects. Although it is genotoxic both in vitro and in vivo, tumours have been observed following inhalation
only at concentrations that have produced significant cytotoxicity and it is likely that both the genotoxicity and irritancy
of acetaldehyde play a role in its carcinogenicity. At acetaldehyde concentrations for which the prevalence of sensory
irritation is low (i.e., < 45 mg/m
3
), risks of respiratory-tract cancers for the general population are considered
exceedingly low.
An Limit of Exposure (EL) has been here derived for short- and long term effects, based on acute eye irritation
observed in human volunteers and on chronic degeneration of the olfactory epithelium observed in rats. No distinction
has been done between short- and long-term ELs since the observed acute effects and endpoints are consistent with the
pathology seen in long-term studies. Also, two studies were considered for the EL derivation, in order to compensate for
the poor quality of the study on volunteers (Silverman et al., 1946). In this study, among 12 volunteers “several“
subjects after 15 min of exposure strenuously objected to acetaldehyde vapor at as low as 45 mg/m
3
,
with this supposed
threshold for sensory irritation set here as a LOAEL (eye irritation was developed by most of the subjects at 90 mg/m
3
).
This level has been devided by an assessment factor of 200, which incorporates uncertainty contributions for
extrapolation from a LOAEL to a NOAEL (10), intraspecies variation (10) and the poor quality of the data (2), resulting
in an Exposure Limit (EL) of 0.2 mg/m
3
. Also, a NOAEL was derived from a subchronic (4 weeks) animal study at 50
mg/m
3
, with an exposure limit of 0.5 mg/m
3
(AF=100) resulting from the extrapolation from an animal study (10) and
intraspecies variation (10).
In addition to the EL derivation, the present assessment takes into account the possible contribution to inhalatory
exposure of the endogenous generation of acetaldehyde in exhaled air. Acetaldehyde is an endogenous metabolite of
sugars and could also be locally formed from the microflora inhabiting the upper airways and mouth. Concentrations
expelled in breath span from 0.009 to 0.026 mg/m
3
, with higher levels observed in smokers and abstinent alcoholics. It
is the first metabolite of ethanol with reported breath concentration increasing to 0.22 – 2.2 mg/m
3
in European subjects
who drank moderate doses of ethanol (0.4-0.8 g/kg bw) (Jones, 1995).
Percentage of population exposed beyond given threshold levels
human LOAEL
(acute)
LOAEL
10
LOAEL
100
LOAEL
200
45 mg/m
3
4.5 mg/m
3
0.45 mg/m
3
0.2 mg/m
3
Studies available
animal NOAEL
(subchronic)
NOAEL
10
NOAEL
100
-
Description (Study, Year) N 50 mg/m
3
5 mg/m
3
0.5 mg/m
3
Paris – refurbished flats (Clarisse, 03) 61 < < < <
Helsinki (Expolis, 97) 15 < < < <
French National survey 7-d TWA (IAQ
observatory, 2003-04)
201 < < < <
< out of the evaluation range (i.e. <5% of the environments investigated)
Comments
Only three exposure studies were found, enabling the construction of cumulative frequency distributions of indoor
exposure to acetaldehyde. Median (90th percentile) indoor concentrations were 0.010 mg/m
3
(0.022) in Paris, 0.016
mg/m
3
(0.030) in Helsinki, and 0.014 mg/m
3
(0.040) in the French National Survey, respectively. Ambient air
concentrations over Europe range from 1-3 mg/m
3
in northern countries, up to 8 and 15 mg/m
3
recently measured in
Italy and Greece, respectively.
The INDEX project Final report
153
Result
The results from only indoor air monitoring surveys allow a crude estimate of average acetaldehyde concentrations in
European residences. Median concentrations (10-20 µg/m
3
) are one order of magnitude lower than the Exposure Limit
set here at 200 µg/m
3
and are within the same range of concentrations occuring in exhaled breath following its
endogenous production in the general populaton, not taking into account increases resulting from the consumption of
alcoholic beverages. Considering that exogenous acetaldehyde peak exposures are mainly associated with tobacco
smoke, concentrations in the order of the Exposure Limit could be expected following intense sigarette consumption.
Assuming that the available exposure data are indicative of the population residential exposure it is concluded that
people in Europe do not experience increased health hazards associated with acetaldehyde levels in their homes,
although additional work should be warranted for a better characterization of exposure and dose response. Also,
measured indoor levels are lower than a presumed threshold for cytotoxic damage to the nasal mucosa, and hence
considered low enough to avoid any significant risk of upper respiratory tract cancer in humans.
The INDEX project Final report
154
Toluene
Synonyms: Methyl benzene, methyl benzol, phenyl methane, toluol
CAS Registry Numbers: 108-88-3
Molecular Formula: C
7
H
8
1. Compound identification
Toluene is a clear, odorous, colourless, readily volatile liquid at ambient conditions. It is flammable and explosive in
air. The technical toluene may contain small amounts of benzene. Toluene will not react with dilute acids or bases and it
is not corrosive. It is removed from air by reacting with hydroxyl radicals. It is one of the most prevalent hydrocarbons
in the troposphere. The lifetime of toluene range from several days in summer to several months in winter. Toluene is
produced in high volume in industrial processes such as catalytic conversion of petroleum and aromatisation of aliphatic
hydrocarbons, and coke oven operations. It is used in blending gasoline to enhance octane ratings, in leather tanning and
as a common solvent. Occupational exposure to toluene might be high during production and use of toluene-containing
products. Toluene is common in many products such as paints, household aerosols, thinners, cleaning agents, coatings,
rubber, nail polish and other cosmetics, adhesives, resin and printing products (IARC 1999, WHO 1986, WHO 2000,
HSDB 2003)
2. Physical and Chemical properties
Molecular weight (g/mol) 92.14
Melting point (°C) -94.9
Boiling point (°C) 110.6
Density (g/l at 20 °C, 1 atm) 0.86
Relative density (air =1) 3.2
Solubility: Soluble in ethanol, benzene, diethyl ether, acetone,
chloroform, glacial acetic acid and
carbon disulfide, insoluble in water
Conversion factors at 20 °C and 760 mm Hg:
1 ppb = 3.824 µg/m
3
1 µg/m
3
= 0.261 ppb
Sources: WHO (1986), Verschueren 2001, EPA/Cal (2003), HSDB (2003)
3. Indoor Air Exposure assessment
Emission sources
Toluene emissions to the atmosphere result from industrial point sources and gas stations, and from mobile sources such
as traffic. Exposure to toluene in indoor environment occurs due to emissions from a variety of toluene-based household
products, uses of paints and thinners, together with tobacco smoke (WHO 2000).
Indoor air and exposure concentrations
Average indoor concentrations of toluene were clearly higher than respective outdoor concentrations in all latitudes in
Europe (Annex 3). Toluene was the most abundant compound in indoor air accounting for 33-54% of the total sum of
The INDEX project Final report
155
aromatics measured in the European EXPOLIS study (Saarela et al 2003). Average outdoor concentrations ranged from
about 6 µg/m
3
in Helsinki and Basel to 43 µg/m
3
in Milan (Jantunen et al 1999). Average indoor concentrations ranged
from 20 µg/m
3
in Helsinki to 74 µg/m
3
in Prague. Personal exposures were clearly higher than indoor concentrations.
The highest average personal exposures were measured in German study, being 130 µg/m
3
(Hoffman et al 2000), and
lowest in Helsinki, 25 µg/m
3
. In a German population based study of children and teenagers (GerES IV), weekly mean
exposure to toluene was higher than the mean residential indoor concentration being 25 µg/m
3
and 15 µg/m
3
respectively. The highest exposure and indoor concentrations were 280 µg/m
3
and 87 µg/m
3
respectively. (Ullrich et al
2002). Italian study carried out by Carrer et al (2000) determining 24-hour exposures of office workers in Milan,
showed an average of 35 µg/m
3
. Based on these results, it is obvious that at least in Central and Southern Europe the
guideline value of 260 µg/m
3
set by WHO (2000) will be exceeded in certain subpopulations.
Median and 90
th
percentile indoor concentrations, 10 µg/m
3
and 49 µg/m
3
, in Arizona, USA (Gordon et al 1999) were
clearly lower than the respective concentrations measured in EXPOLIS in European cities.
Ambient air concentrations of toluene in Europe have been measured in several studies (Saeger et al 1995, Ballesta et al
1997, Ballesta et al 1998, Kemp et al., 1998). Typical ambient air concentrations of toluene were in the order of 10 to
30 µg/m
3
. The concentrations of toluene near petrol stations were generally around an order of magnitude higher than
the urban air concentrations (EPA 2003).
Ambient toluene concentrations in urban environments in US have been in the same range as in Europe ranging about
17-20 µg/m
3
, the highest level being 62 µg/m
3
in a metropolitan area. In rural sites levels were typically below 1 µg/m
3
(Helmig and Arey 1992, WHO 2000).
Residential indoor air concentrations of toluene in European urban populations measured in the EXPOLIS study and the
National Survey in England are presented in Figure 3.1 and Figure 3.2. Respective exposure concentrations are
presented in Figure 3.3.
Source related short time concentrations of toluene are presented in Figure 3.4. Even higher concentration of toluene,
2.1 mg/m
3
was measured in a kindergarten in Athens in the AIRMEX project (Kotzias 2004). The source of toluene
identified in the kindergarten was a spray that was used when preparing Christmas decorations. Lee and Wang (2004)
studied emissions of incense burning in chamber (18 m
3
) tests. Concentrations ranged from 5 µg/m
3
to 99 µg/m
3
.
Measured concentrations did not exceed the Recommended Indoor Air Quality Objectives for Office Buildings and
Public Places in Hong Kong (HKIAQO) of < 1092 µg/m
3
. Brown (2002) studied short time (30-50 min) concentrations
of toluene in established and new buildings, reporting mean concentrations of 14 µg/m
3
and 250 µg/m
3
respectively.
Toluene concentration in a new building decreased to 18 µg/m
3
and 6.9 µg/m
3
in 72 and 246 days respectively.
Emission sources of toluene were not identified, but VOC emissions in general were related to water-based paints,
adhesives and wood-based panels. Elke at al (1998) measured short time concentrations of toluene inside buildings (60-
min average), inside a train and a car (30-min average), being at the same level, 55 µg/m
3
, in a train and a car, and 21
µg/m
3
in a smoker day room.
Short time exposure scenarios for potential consumer activities were analysed in European Union Risk Assessment
Report (EU 2003). The highest toluene concentration, 1000 mg/m
3
(note the unit, mg/m
3
) were expected during
spraying painting. High peak concentrations were also assessed during carpet laying, car polishing and gluing, 195
mg/m
3
, 10 mg/m
3
and 7.1 mg/m
3
respectively.
The INDEX project Final report
156
0
20
40
60
80
100
0 100 200 300 400 500
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
Hel
Mil
Oxf
Pra
Figure 3.1. Cumulative frequency distributions of 30-hour indoor air concentrations of toluene in Athens (Ath, n=42),
Basel (Bas, n=47), Helsinki (Hel, n=188), Milan (Mil, n=41) Oxford (Oxf, n=40) and Prague (Pra, n=46) (EXPOLIS
2002).
0
20
40
60
80
100
0 1020304050607080
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Toluene 28-day
Figure 3.2. Cumulative frequency distribution of 28-day indoor air concentrations of toluene in England (GM= 15.1
µg/m
3
, max 1784 µg/m
3
,
n=796, Brown et al 2002).
The INDEX project Final report
157
0
20
40
60
80
100
0 100 200 300 400 500
Exposure (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
GeS
Hel
Oxf
Pra
Figure 3.3. Cumulative frequency distributions of 48-hour personal exposure concentrations of toluene in Athens (Ath),
Basel (Bas), Helsinki (Hel), Oxford (Oxf) and Prague (Pra) (EXPOLIS 2002), and 1-week mean concentrations of the
German Survey GerES II (GeS) (Hoffman et al 2000).
0
50
100
150
200
250
300
Incence
burning
New
buildings
In a car Room with
ETS
Train with
ETS
Concentration (µg/m
3
)
Toluene
Figure 3.4. Short time toluene concentrations related to specific microenvironments or emission sources. Sampling
times of ‘New buildings’ (Brown 2002), ‘In a car’, ‘Room with ETS’, and ‘Train with ETS’ (Elke at al 1998) were 30-
50 min, 30-min, 60-min, 30-min respectively. During incense burning grab samples were obtained (Lee and Wang
2004).
The INDEX project Final report
158
4. Toxicokinetics
Absorption
The major uptake of toluene vapour is through the respiratory system. A number of investigations in humans (Carlsson,
1982; Carlsson and Lindqvist, 1977; Nomiyama and Nomiyama, 1974; Åstrand 1975) have shown that at rest a three-
hour exposure to toluene vapour will result in an uptake amounting to approximately 50% of the inhaled toluene.
The blood/air partition coefficient for toluene is 11,2-15,6 at 37°C (Lindqvist, 1977; Sato et al., 1972; Sato and
Nakajima, 1977; Sherwood, 1976; Ulfvarson and Övrum, 1976).
The concentration of toluene in alveolar air and in arterial and venous blood rises quickly during the first 10-15 minutes
of exposure (Carlsson, 1982; Åstrand et al., 1972). After only 10 seconds of exposure toluene can be detected in blood
from brachial arteries (Åstrand et al., 1972).
Data from experimental exposure of voluntary study subjects show that physical work results in increased toluene
uptake (Veulemans and Masschelein, 1978b; Carlsson, 1982). Using a 50 W workload, exposure to 300 mg/m
3
(80
ppm) toluene for 2 hours did not result in steady state of the blood concentration of toluene in 12 study subjects. The
toluene uptake was 2,4 times higher than the uptake at rest. During the work, lung ventilation was increased 2,8 times.
Concentrations of toluene in alveolar air and blood increased with increasing work loads (0-150 W in periods of 30
minutes) (Carlsson, 1982). The amount of toluene absorbed increased with greater amounts of body fat (Carlsson and
Ljungquist, 1982).
In nine male volunteers exposed to 200 mg/m
3
(53 ppm) toluene for 2 hours during a workload of 50 W, the total uptake
of toluene was 50% of that inhaled (Löf et al., 1993).
Distribution
The distribution of toluene in the body is among other factors dependent on the tissue/blood partition coefficients and
the metabolism. In rabbits the following partition coefficients have been found: brain, heart, liver, and intestine: 2,3,
muscle tissue: 1,6, adipose tissue: 74,3, bone, connective tissue, and lung tissue: 1,9 (Sato et al., 1974). In rats
brain/blood ratios of 1,2 (Kishi et al., 1988) and 1,7 (Zahlsen et al., 1992) have been determined. In humans the adipose
tissue/blood partition coefficient for toluene is determined to be 81-83 (Sato et al., 1974; Sherwood, 1976). This high
partition coefficient suggests that toluene can be accumulated in adipose tissue.
In mice, the distribution of toluene and its metabolites was investigated using whole body autoradiography after acute
inhalation of side chain marked
14
C toluene (Bergman, 1979; Bergman, 1983). In adipose tissue, bone marrow, spinal
nerves, spinal cord, and in the white parts of the brain, high concentrations of radioactivity occurred as judged from the
photographs. In blood, liver, and kidneys, radioactivity was also found. One hour after exposure nerve tissue showed no
radioactivity. In adipose tissue nearly all radioactivity had disappeared four hours after exposure, and only traces of
non-volatile radioactivity could be found in the liver. After 24 hours all radioactivity had disappeared from the body.
In rats, subcutaneous injection of toluene (100 or 500 mg/kg) resulted in maximum concentrations of blood toluene
after 2 hours (Benignus et al., 1984).
Toluene passes the placenta. Two hours following exposure of rats via inhalation to 1375 or 2700 mg/ m
3
(367 or 720
ppm) for 24 hours, foetal blood had a toluene concentration of 74% of that found in the dam’s blood. The amniotic
liquid contained a toluene concentration of 5% of that in the dam’s blood. Four and six hours after exposure, similar
relative toluene concentrations were found (Ungváry, 1984; quoted from IPCS 1985).
Groups of four 11, 14 or 17 day pregnant mice were killed 0, 30, 60 and 240 minutes after having inhaled 7500 mg/m
3
(2000 ppm)
14
C-toluene for ten minutes. Radioactivity as volatile and non-volatile was measured in lung, liver, kidney,
brain, cerebellum, fat, plasma, amniotic fluid, placenta, and foetus. It was shown that toluene immediately after
inhalation was taken up in the foetal tissue at a concentration of about 10% of that found in the maternal lungs. At four
hours after exposure the toluene radioactivity was decreased to 2% of the original value (Ghantout and Danielsson,
1986).
Toluene has been found in human breast milk. In 12 pooled samples from four urban areas in the United States, toluene
was identified qualitatively in at least 7 samples (Pellizzari et al., 1982; quoted from Jensen and Slorach, 1991).
The INDEX project Final report
159
Metabolism and elimination
The major pathway of toluene metabolism in both humans and laboratory animals involves sidechain oxidation by
sequential action of cytochrome P-450, alcohol dehydrogenase and aldehyde dehydrogenase leading to benzoic acid
which, upon conjugation with glycine, results in hippuric acid, the major urinary metabolite. Minor metabolites include
ortho- and para-cresol. A minor metabolite that is specific for toluene exposure is S-benzyl-N-acetyl-L-cysteine
(Takahashi et al., 1994). In vitro evidence suggests that the rate of metabolism in humans is greater than that in rats
(Chapman, et al., 1990).
Analysis of blood and urine samples from workers and voluntary study subjects exposed to toluene via inhalation in
concentrations ranging from 375 to 2250 mg/m
3
(100-600 ppm) indicate that of the biotransformed toluene, appr. 99%
is oxidised via benzyl alcohol and benzaldehyde to benzoic acid. The remaining 1% is oxidised in the aromatic ring,
forming ortho-, meta- and para-cresol (Woiwode et al., 1979; Woiwode and Drysch, 1981).
Water solubility of the biooxidation products is achieved through linkage with suitable substances (phase 2 reaction).
Benzoic acid is linked to either glycine or glucuronic acid forming either hippuric acid or benzoylglucuronide. Cresols
and benzyl alcohol are linked to glucuronic acid or sulphate (IPCS, 1985).
The elimination of toluene from adipose tissue is prolonged according to the findings of Periago et al. (1992), who
examined a worker population. The half-time for elimination from adipose tissue was reported to range between 0,5 and
2,7 days, depending on the amount of body fat. Elimination from bone marrow also is prolonged (Chemical and
engineering news, 1995; WHO, 1987).
Toluene or its metabolites may be eliminated via the lungs, the kidneys, or the liver.
Data from experimental inhalation exposure of voluntary subjects show that the toluene concentration in expired air
decreases rapidly during the first 10 to 20 minutes after cessation of exposure to toluene via inhalation (Veulemans and
Masschelein, 1978a; Carlsson, 1982; Echeverria et al., 1989). Two to four hours later, very low toluene concentrations
are found in expired air (Carlsson, 1982). Of the toluene absorbed, 15-20% is exhaled during the first few hours after
exposure has stopped (Nomiyama and Nomiyama, 1974). The cumulative elimination of toluene via the lungs amounts
to 4-8% and 7-14% after 2 and 20 hours, respectively (Carlsson, 1982).
The cumulative elimination (in per cent) of toluene via the lungs appears to increase with increasing amounts of toluene
taken up (Carlsson, 1982).
The majority (80-90%) of absorbed toluene is biotransformed and excreted from the body via the kidneys. At an
exposure level of 750 mg/m
3
(200 ppm), the excretion is mainly as hippuric acid. About 1% of the biotransformed
toluene is excreted as glucuronides or sulphates of o-, m-, or pcresol (IPCS, 1985).
A very small proportion, approximately 0,06% of the toluene absorbed via inhalation, is excreted unchanged in the
urine in humans (Williams, 1959).
A good correlation was found between toluene exposure (air concentration multiplied by time) and concentration of
hippuric acid in post exposure urine. However, a background level of hippuric acid is present in human urine, as a
product of endogenous metabolism, and of metabolism of substances present in food. In the Western part of the world,
at exposure levels below 375 mg/m
3
(100 ppm) hippuric acid in post exposure urine cannot be used to separate an
exposed person from an unexposed one because the difference between the background level and the toluene-generated
level is too small (Lauwerys, 1983). However, hippuric acid background levels in urine vary geographically. In some
Third World countries a low urinary hippuric acid background level is found. Thus, in these parts of the world it is
possible to use this metabolite as a biological marker for toluene exposure even at exposure levels lower than 375
mg/m
3
(Chang et al., 1996; Vrca et al., 1997a; 1997b).
Data from rats exposed to 3750, 6675, or 11250 mg/m
3
(1000, 1780, or 3000 ppm) of toluene via inhalation for 2 hours
showed that elimination could be described by a bi-exponential function with average half-times of 6 and 90 minutes
(Rees et al., 1985).
In rats a small proportion, less than 2%, of the absorbed toluene is excreted via the bile to the intestine. The substances
excreted are reabsorbed in the intestine. Thus very small amounts are excreted in faeces (Abou-El-Makarem et al.,
1967).
The INDEX project Final report
160
5. Health effects
Effects of short-term exposure
Dysfunction of the central nervous system and narcosis are the major effects of acute exposure to toluene (ATSDR,
1989). Irritation of the skin, eye, and respiratory tract can also result. Inhalational abuse of toluene with high level
exposure for long periods of time has produced progressive and irreversible changes in brain structure and function
(Spencer and Schaumberg, 1985).
Reaction time and perceptual speed were studied in 12 young male subjects exposed by inhalation to toluene
concentrations ranging from 400 to 3000 mg/m³ (100 to 700 ppm), each for a 20-minute interval (Gamberale and
Hultengren, 1972). Statistically significant impaired reaction time was apparent following exposure to 1100 mg/m³ (300
ppm) toluene. A statistically significant impairment in perceptual speed was observed at 3000 mg/m³ toluene. No
effects were observed at 400 mg/m³.
Two groups of middle aged workers, one with previous occupational exposure to solvents and one without, were
exposed once to 400 mg/m³ (100 ppm) of toluene for 6,5 hours (Baelum et al., 1985). Fatigue, sleepiness, a feeling of
intoxication, and eye, nose and throat irritation were reported. Decrements in manual dexterity, color discrimination,
and accuracy in visual perception were also observed. Greater sensitivity to toluene was noted for those subjects with
previous solvent exposure.
A random population sample of 32 male and 39 female subjects were allocated into three groups, one exposed to clean
air, one exposed to constant toluene at a concentration of 377 mg/m
3
(100 ppm), and one exposed to varying
concentrations of toluene (Fourteen 30-minute episodes during the exposure period, each episode starting with an
increasing concentration reaching a peak of 1125 mg/m
3
(300 ppm) after 5 min and then decreasing to a stable period of
about 15 min at 188 mg/m
3
(50 ppm), giving a TWA of 375 mg/m
3
for the whole exposure period). An exposure period
comprised a single day (7 hours). Toluene caused throat and respiratory irritation, headache and dizziness. In
performance tests only minimal effects were found, with a tendency towards lower score and more errors but fewer
false reactions in the primary task of the vigilance test. The effects were not statistically significant (p<0,1). There was
no difference between constant exposure and peak exposure (Bælum et al., 1990).
Nasal mucus flow, lung function, psychometric performance, and subjective responses were studied in 16 young
healthy males exposed to toluene concentrations ranging from 38 mg/m³ to 377 mg/m³ (10 to 100 ppm) for 6 hours
(Andersen et al, 1983). Headaches, dizziness, a feeling of intoxication, and slight eye and upper respiratory irritation
were reported at 377 mg/m³. The subjects also reported that it became more difficult to participate in the battery of
psychometric tests and that their reaction time felt impaired at 377 mg/m³. No significant objective changes compared
to control exposures were observed in the performance test results. No symptoms were reported at 38 and 151 mg/m³.
A battery of neurobehavioral and performance tests was conducted among 42 young men and women exposed by
inhalation for 7 hours to 0, 283, and 566 mg/m
3
(0, 75, and 150 ppm) toluene (Echeverria et al., 1989). Statistically
significant decrements in visual short term memory, visual perception, and psychomotor skills were observed at 560
mg/m³ compared to control exposures. A dose-dependent increase in subjective symptoms of headache and eye
irritation was also observed.
Wilson (1943) reported that workers exposed to concentrations of commercial toluene ranging from 200 to 750 mg/m³
(50 to 200 ppm) for periods of 1 to 3 weeks experienced headaches, lassitude, and loss of appetite. At 750 to 2000
mg/m³ (200 to 500 ppm), symptoms of nausea, bad taste in the mouth, slightly impaired coordination and reaction time,
and temporary memory loss were also observed. Exposure to 2000 to 5600 mg/m³ (500 to 1500 ppm) resulted in
palpitations, extreme weakness, pronounced loss of coordination, and impaired reaction time. Red blood cell counts
were decreased and there were 2 cases of aplastic anemia. The hematologic effects were most likely caused by benzene
impurities (ACGIH, 1986).
Three volunteer subjects exposed by inhalation to toluene concentrations ranging from 200 to 400 mg/m³ (50 to 100
ppm), 8 hours per day, 2 times per week over 8 weeks experienced fatigue, drowsiness, and headaches (von Oettingen
et al., 1942). At 750 to 3000 mg/m³ (200 to 800 ppm), symptoms of muscular weakness, confusion, impaired
coordination, paresthesia, and nausea were also reported. After exposure to 3000 mg/m
3
, all 3 subjects reported
considerable aftereffects (severe nervousness, muscular fatigue, and insomnia) lasting several days.
The INDEX project Final report
161
Summary of short-term exposure effect levels
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
151
EXP
369
1h-ADJ
377
EXP
923
1h-ADJ
CNS-mild, eyes, respiratory;
impaired reaction time, headache, dizziness,
feeling of intoxication, slight eye and nose
irritation
volunteers, 6h
Andersen et al. 1983 OEHHA (1999);
REL: 37
ATSDR (2003);
MRL: 3.8
151
EXP
283
EXP
CNS; NOAEL: see above
LOAEL: visual short term memory, visual
perception and psychomotor skills
volunteers, 6-7h
NOAEL: Andersen et
al. 1983
LOAEL: Echeverria et
al. 1989
ECB (2003)
Final statement (UNIMI) : Human NOAEL = 150 mg/m³
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
OEHHA (1999)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Derivation of the Acute Reference Exposure Level (protective against mild adverse effects):
Study Andersen et al., 1983
Study population 16 young, healthy males
Exposure method inhalation
Critical effects impaired reaction time and symptoms of headache, dizziness, a feeling of
intoxication and slight eye and nose irritation
LOAEL 100 ppm
NOAEL 40 ppm
Exposure duration 6 hours
Extrapolated 1 hour concentration 98 ppm (40
2
ppm* 6 h = C
2
* 1 h )
LOAEL uncertainty factor 1
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 10
Cumulative uncertainty factor 10
Reference Exposure Level 9,8 ppm (37 mg/m³; 37.000 µg/m³)
ECB (2003)
European Chemical Bureau - Institute for Health and Consumer Protection - European Union Risk Assessment Report
In the exposure chamber studies: Echeverria et al., (1989) and Andersen et al., (1983), headache, dizziness, feeling of
intoxication, irritation and sleepiness were recorded to occur with significantly increased frequency at exposure levels
from 562 mg/m
3
(150 ppm) down to 283 mg/m
3
(75 ppm). At 151 mg/m
3
(40 ppm) and below the effects have not been
recorded to occur with increased frequency. For these subjective symptoms a LOAL of 283 mg/m
3
(75 ppm) and a
NOAEL of 151 mg/m
3
(40 ppm) can be established.
With respect to function in performance tests, inhalation of 281 mg/m
3
(75 ppm) and 562 mg/m
3
(150 ppm) for 7 hours
have resulted in significantly worse results in a number of performance tests, indicating a LOAEL of 281 mg/m
3
(75
ppm) for function in performance tests while a NOAEL cannot be established.
ATSDR (2003)
Agency for Toxic Substances and Disease Registry
Derivation of the acute Minimal Risk Level (MRL).
There are several human studies for which the central nervous system is the major end point and could have been used
to derive an acute inhalation MRL. However, the Andersen et al. (1983) study was chosen as the basis for the MRL
because this was the only human study which reported a NOAEL. Baelum et al.(1985) also reported a LOAEL of 377
mg/m
3
for neurological effects in humans. In this study, 43 occupationally-exposed subjects and 43 controls were
exposed to either clean air or air containing 377 mg/m
3
toluene for 6.5 hours in a climate chamber. A battery of ten tests
of visuomotor coordination, visual performance, and cortical function were administered during the 6.5 hour period. For
The INDEX project Final report
162
both the controls and toluene exposed subjects, there were complaints of air quality, irritation of the nasal passages, and
increased feelings of fatigue and sleepiness. Subjects also complained of headaches and dizziness. Toluene exposure
decreased performance on four of the neurobehavioral tests; three of these were tests of visual perseverance. The fourth
test affected was the simple peg board test of visuomotor function, where the effect was noted in toluene-exposed
workers to a much greater extent than controls. Escheverria et al. (1991) reported a LOAEL of 283 mg/m
3
for
neurological effects in humans. In this study, two groups of 42 students were exposed to 0, 75, and 150 ppm toluene for
a 7 hour period. A complete battery of 12 tests was administered before and at the end of each exposure. Toluene
caused a dose-related impairment of function on digit span pattern recognition, the one hole test, and pattern memory.
Rahill et al. (1996) reported a LOAEL of 377 mg/m
3
for neurological effects in humans. In this study, six volunteers
were exposed for 6 hours a day to either 377 mg/m
3
toluene or clean air. Three repetitions of two computerized
neuropsychological tests were performed, with the composite score on the multitasking test being significantly lower
with toluene exposure than with clean air.
Dose endpoint used for MRL derivation: 151 mg/m
3
for neurological effects
Uncertainty factors used in MRL derivation:10 (for human variability)
MRL = 151 mg/m
3
x 5 days/7 days x 8 hours/24 hours ÷10 = 3.8 mg/m
3
Effects of long-term exposure
Most human studies reporting adverse effects due to chronic toluene exposures involve either toluene-containing
solvent abuse or occupational exposure to toluene. Solvent abusers are generally exposed to higher levels of toluene
than are workers. A continuum of neurotoxic effects ranging from frank brain damage to degraded performance on
psychometric tests which roughly track exposure levels has been observed.
An extensive database on human exposure to toluene indicates that dysfunction of the CNS is of primary concern.
Deficits in neurobehavioural functioning have been viewed as precursors of more serious indications of CNS toxicity
(Chemical and engineering news, 1995; WHO, 1981). Their measurement is generally held to be more reliable than
subjective symptoms, which may involve situational factors unrelated to a cause–effect relationship with exposure.
Potential confounders such as age, alcohol, drugs and education need to be assessed to ensure correlations of toxicity
with exposure.
Solvent workers exposed to 160 mg/m
3
toluene (estimated as a time-weighted average) for an average duration of 6,8
years reported a significantly greater incidence of sore throat, dizziness and headache than controls; the sore throat and
headache incidence demonstrated a rough dose-response (Yin et al., 1987).
Orbaek and Nise (1989) examined the neurological effects of toluene on 30 rotogravure printers, 33-61 years of age
(mean 50), employed at two Swedish printing shops for 4-43 years (median 29) in 1985. Mean exposure levels at the
two printing shops were 43 and 157 mg/m
3
of toluene, respectively; however, before 1980 the exposure levels had
exceeded 300 mg/m
3
in both shops. The authors noted that rotogravure printing provides an occupational setting with
practically pure toluene exposure. Comparisons were made to a reference group of 72 men aged 27-69 (mean 47). The
alcohol consumption of both the workers and referents was also determined (< 200 g/week or > 200 g/week).
Neurological function in the workers and referents was evaluated using interviews and psychometric testing; the results
from each of the two printing shops were pooled. The printers reported statistically significantly higher occurrences of
fatigue (60%), recent short-term memory problems (60%), concentration difficulties (40%), mood lability (27%), and
other neurasthenic symptoms. The printers also scored significantly worse than referents in a number of psychometric
tests, including synonym, Benton revised visual retention and digit symbol tests, even after adjustment for age. For all
comparisons, tests of interaction between the effects of toluene exposure and alcohol consumption were not statistically
significant.
A battery of neurobehavioral tests was performed in 30 female workers exposed to toluene vapors in an electronic
assembly plant (Foo et al., 1990). The average number of years worked was 5,7 ± 3,2 for the exposed group and 2,5 ±
2,7 years for the controls. Study subjects did not smoke tobacco or drink alcohol, were not taking any medications, and
had no prior history of central or peripheral nervous system illness or psychiatric disorders. The exposed group of
workers inhaled a time-weighted average (TWA) of 332 mg/m
3
(88 ppm) toluene while the control workers inhaled 49
mg/m
3
(13 ppm). A significant decrease in neurobehavioral performance was observed in the exposed workers in 6 out
of 8 tests. Irritant effects were not examined, and concurrent exposures to other chemicals were not addressed. In this
study, 332 mg/m
3
was considered a LOAEL for central nervous system effects. However, the workers designated by the
authors to be controls did not comprise a true control group, since they were exposed to 49 mg/m
3
toluene. This may
have resulted in an underestimation of the effects of exposure to 332 mg/m
3
toluene. Similar effects were noted in a
follow-up study by Boey et al. (1997).
The INDEX project Final report
163
Abbate et al. (1993) evaluated alterations induced in the auditory nervous system by exposure to toluene in a group of
rotogravure workers. A sample of 40 workers of normal hearing ability was selected from a group of 300 workers who
were apparently in good health but were professionally exposed to toluene (12 – 14 years exposure, 343 mg/m
3
average
exposure, exposure assessment not described). They were subjected to an adaptation test utilizing a BAER technique
with 11 and 90 stimulus repetitions a second. The results were compared with an age and sex-matched control group not
professionally exposed to solvents. A statistically significant alteration in the BAER results was noted in the toluene-
exposed workers with both 11 and 90 stimuli repetitions. The authors suggested that these results can be explained as a
tolueneinduced effect on physiologic stimulus conduction mechanisms, even in the absence of any clinical sign of
neuropathy. Furthermore, this effect could be observed in the responses of the entire auditory system, from peripheral
receptors to brainstem nuclei.
A group of 49 printing-press workers occupationally exposed to toluene for approximately 21,6 years was studied by
Vrca et al. (1997). Toluene exposure levels were determined from blood toluene and urinary hippuric acid levels, and
were estimated to range from 151-226 mg/m
3
. No control group was used. Brain evoked auditory potential (BEAP;
similar to BAER) and visual evoked potential (VEP) measurements were performed on a Monday morning after a
nonworking weekend. There was a significant increase in the latencies of all the BEAP waves examined, except for P2
waves, as well as in the interpeak latency (IPL) P3-P4, while IPL P4-P5 decreased significantly with the length of
exposure. No correlation was noted between the amplitude of BEAP waves and the length of exposure. The amplitude
but not the latency of all the VEPs examined decreased significantly with the length of exposure.
The effects of acute and chronic toluene exposure on color vision were studied in a group of eight rotogravure printing
workers (Muttray et al., 1999). The workers had been employed as printers for an average of 9,8 years. The color vision
acuity of the workers before and after an acute toluene exposure (28– 41 minutes in duration, concentration 1115 –
1358 mg/m
3
) was evaluated using the Farnsworth panel D-15 test, the Lanthony desaturated panel D-15 test, and the
Standard Pseudoisochromatic Plates part 2. A control group of 8 unexposed workers was also tested. Acute toluene
exposure had no effect on color vision. Print worker performance prior to acute toluene exposure (chronic effects) was
similar to controls on the Farnsworth panel D-15 and Standard Pseudoisochromatic Plates part 2 tests. Print worker
performance on the Lanthony desaturated panel D-15 test was worse than that of controls (median scores of 1,18 and
1,05 for exposed and controls (higher number indicates degraded performance), respectively, but not significantly (p =
0,06). The authors noted that the small number of subjects limited the statistical power of the study.
Three groups of Croatian workers were examined by means of interviews, medical examination, and color vision testing
using the Lanthony 15 Hue desaturated panel in standard conditions (Zavalic et al., 1998a). Workers were excluded
from the study if they met any of the following criteria: less than 6 months employment, congenital color vision loss, a
medical condition which can affect color vision, visual acuity below 6/10, use of medications which can affect color
vision or a hobby that involved solvent exposure. Alcohol intake and smoking were also assessed for each individual.
The first group consisted of 46 workers (43 women and 3 men) employed in manually glueing shoe soles and exposed
to median levels of 121 mg/m
3
and geometric mean levels of 132 mg/m
3
toluene. The second group consisted of 37
workers (34 men and 3 women) employed in a rotogravure printing press and exposed to median levels of 498 mg/m
3
and geometric mean levels of 588 mg/m
3
toluene. The third group consisted of 90 workers (61 men and 29 women) not
occupationally exposed to any solvents or known neurotoxic agents. The study demonstrated a statistically significant
impairment of color vision in workers chronically exposed to 588 mg/m
3
toluene compared with controls. When the
data were adjusted to allow for the confounding effects of alcohol consumption and age, a significant difference due to
toluene exposure was also reported for workers exposed to 132 mg/m
3
toluene compared with controls.
Zavalic et al. (1998b) examined the effects of chronic occupational toluene exposure on color vision using a further
group of 45 exposed workers (mean toluene exposure concentration = 452 mg/m
3
) and 53 controls. Color vision was
evaluated using the Lanthony desaturated panel D-15 test; test scores were age and alcohol consumption-adjusted.
Color vision was significantly impaired in toluene-exposed workers (p < 0,0001) compared to controls. It was also
observed that there was no significant difference between test scores on Monday morning (prework) and Wednesday
morning. The authors stated that the effect of toluene on color vision can be chronic and that the possible recovery
period is longer than 64 hours.
Effects on kidney and liver
Among 24 toluene abusers examined on the day of admission to hospital, alkaline
phosphatase was elevated in 13 patients, while SGOT was elevated in 7. The elevated enzyme levels returned to normal
after 2 weeks of abstinence (Fornazzari et al., 1983).
No increase in levels of the enzymes serum aspartate aminotransferase and alanine aminotransferase was found, in 59
men with occupational exposure to toluene (recorded level 375 mg/m
3
) for more than one year (1-5 years, 22 men; 6-10
The INDEX project Final report
164
years, 18 men; more than 10 years, 19 men) when compared to an unexposed control group of equal size (Waldron et
al., 1982).
In 47 toluene-exposed workers a significant increase in S-ALP (20% relative to referents) compared with a referent
group of 46 non-exposed workers was found (Svensson et al., 1992b). The association was still significant when heavy
alcohol consumers were excluded from the analysis. The exposure levels were generally below 300 mg/m
3
(80 ppm).
Other liver functionrelated enzyme levels were unaffected. There was no association with cumulative exposure.
Sniffing of toluene resulted in reversible kidney damage (O'Brian et al., 1971), haematuria (Massengale et al., 1963),
reversible type 1 renal tubular acidosis (Bennett and Forman, 1980; Fischman and Oster, 1979; Kroeger et al., 1980;
Moss et al., 1980; Patel and Benjamin, 1986; Reisin et al., 1975; Streicher et al., 1981; Taher et al., 1974; Weinstein et
al., 1985; Will and McLaren, 1981) and hypokalaemia (Kelly, 1975; Taher et al., 1974). In some cases sniffing resulted
in irreversible damage of the kidneys (Russ et al., 1981).
A workplace accident with massive toluene exposure for 18 hours resulted in renal failure with oligouria probably
caused by dehydration and myoglobinuria (Reisin et al., 1975).
Inhalation of 382 mg/m
3
(100 ppm) toluene for 6,5 hours in an exposure chamber resulted in unchanged excretion of
albumin and beta-2-microglobulin for 43 printers with occupational exposure to toluene as compared to 43 age-matched
controls without occupational exposure to toluene (Nielsen et al., 1985).
No signs of renal damage in 118 painters were found compared with a control group. The painters had an average of 9
years occupational exposure to toluene and xylenes. At the time of investigation, the exposure was approximately 94
mg/m
3
(25 ppm) as determined from metabolites in urine (Franchini et al., 1983).
In 42 printers with an occupational toluene exposure averaging 300 mg/m
3
(80 ppm) (range 100-900 mg/m
3
(30-240
ppm)) compared with 48 unexposed controls, no changes in glomerular filtration rate, renal concentrating ability, beta-
2-microglobulin excretion, and excretion of erythrocytes and leukocytes were found (Askergren, 1982).
In conclusion, massive toluene exposure through abuse or workplace accidents has been associated with kidney damage
and renal failure. Three occupational studies did not show a relation between toluene exposure and kidney damage.
Developmental, reproductive and teratogenic effects
Abuse of toluene by pregnant women through deliberate inhalation of products such as paint thinners, glues and paints
has been associated with a number of developmental and congenital anomalies in infants (Donald et al., 1991). In such
reports it is difficult to determine the degree to which other substances may play a role in the development of adverse
effects. Effects commonly noted postpartum include low birth weight, growth retardation, microencephaly, CNS
dysfunction, renal tubule acidosis, and minor craniofacial and limb abnormalities. Perinatal death has also been
reported.
Two studies suggest an increased risk of spontaneous abortions associated with exposure to toluene in the workplace.
One of the studies provides no data on exposure levels, while the levels were around 332 mg/m
3
(range 189-566 mg/m
3
)
in the other study (Ng et al., 1992b). The Ng et al. (1992) study cannot be used to establish definitively a causal
relationship between late spontaneous abortions and toluene exposure or the magnitude of the LOAEL To establish a
definite relationship, a prospective study including pregnant women exposed to toluene at similar exposure levels with
individually monitored data on toluene exposure and fetal loss would be needed. However, based on the current
evidence suggesting an increased risk for late spontaneous abortions, exposure of pregnant women to such exposure
levels would raise serious ethical concerns. Consequently, the results of the Ng study are used as a basis for the risk
characterisation of developmental toxicity in humans.
Menstrual disorders in workers were reported, but the possible presence of other chemicals in the exposure
environments and unmatched characteristics of the exposed and control groups make these findings difficult to interpret
(WHO, 1987). Possible exposure-related effects upon follicle stimulating hormone and testosterone (Svensson et al.,
1992a), but not serum prolactin levels (Svensson et al., 1992b), have been observed in printers without overt effects of
toluene.
Developmental toxicity of toluene has mainly been studied in rats. Rat inhalation studies provide strong evidence of
developmental toxicity (lower birth weight and long-lasting developmental neurotoxicity) in the absence of maternal
toxicity. The effective dose levels are around or more than 3.77 g/m
3
. The NOAEL for lower birth weight and delayed
The INDEX project Final report
165
postnatal development is 2.26 g/m
3
(Thiel and Chahoud, 1997). A NOAEL for developmental neurotoxicity cannot be
determined from the available studies. The LOAEL for this effect is 4.52 g/m
3
(Hass et al., 1999).
There is no indication that toluene cause malformations in rats, mice or rabbits.
Summary of long-term exposure effect levels
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
151
EXP
27
ADJ
301
EXP
54
ADJ
CNS; decreased brain weight, dopamine receptor
binding
Supporting study: Neurobehavioral deficits
rats, 4w
(supported by
occupational, 5.7y)
Hillefors-Berglund et
al. 1995; supported by
Orbaek and Nise
(1989), Foo et al.
(1990)
OEHHA (1999);
REL: 0.4
332
EXP
118
ADJ
CNS;
neurobehavioural deficits
Occupational, 5.7y
Foo et al. 1990 OEHHA (1999);
WHO (2001);
GV: 0.26;
EPA-IRIS (1992)
; RfC: 0.4
2261
EXP
437
ADJ
79
HEC
Respiratory system;
nasal epithelium degeneration
rats, 2y
NTP, 1990 EPA-IRIS (1992)
132
Study
31
ADJ
CNS;
color vision impairment
Occupational, >6m Zavalic et al. (1998a) ATSDR (2003);
MRL: 0.3
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
OEHHA (1999)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Derivation of the Chronic Reference Exposure Level (REL):
Study Hillefors-Berglund et al. (1995); supported by Orbaek and
Nise (1989), Foo et al. (1990)
Study population Male Sprague-Dawley rats
Exposure method Inhalation
Critical effects Decreased brain (subcortical limbic area) weight, Altered dopamine
receptor (caudate-putamen) binding
LOAEL 80 ppm
NOAEL 40 ppm
Exposure continuity 6 hours/day, 5 days/week
Exposure duration 4 weeks, followed by 29-40 days recovery
Average experimental exposure 7 ppm (40 × 6/24 hours × 5/7 days)
Human equivalent concentration 7 ppm (gas with systemic effects, based on RGDR = 1,0
using default assumption that λa = λh
Subchronic uncertainty factor 10
Interspecies uncertainty factor 1 (see below)
Intraspecies uncertainty factor 10
Cumulative uncertainty factor 100
Inhalation reference exposure level 0,07 ppm (70 ppb; 0,3 mg/m
3
; 300 µg/m
3
)
Supportive human study Foo et al., 1990
Study population 30 female workers in an electronic assembly plant
Exposure method Occupational inhalation
Critical effects Neurobehavioral deficits in 6 out of 8 tests
LOAEL 88 ppm
NOAEL Not observed
Exposure continuity 10 m
3
/day occupational inhalation rate, 5 days/week
Average occupational exposure 31,4 ppm (88 ppm x 10/20 x 5/7)
Exposure duration 5,7 + 3,2 years (exposed group); 2,5 + 2,7 years (controls)
LOAEL uncertainty factor 10
Subchronic uncertainty factor 3
The INDEX project Final report
166
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 10
Cumulative uncertainty factor 300
Inhalation reference exposure level 0,1 ppm (100 ppb; 0,4 mg/m
3
; 400 µg/m
3
)
The critical animal study (Hillefors-Berglund et al., 1995) used to derive a REL for toluene describes adverse
neurological effects in rats after a well characterized inhalation exposure to toluene. The study results contain both a
LOAEL and a NOAEL. Decreased brain (subcortical limbic area) weight and altered dopamine receptor binding
compared to controls were noted at the NOAEL, but the changes were not statistically significant; this suggests that if a
threshold for adverse neurological effects exists in this study, it would be at or below the observed NOAEL. The study
LOAEL for altered dopamine receptor binding agrees qualitatively with results from similar studies (von Euler et al.,
1994).
Additionally, toluene induced neurotoxicity has been described in many studies by a variety of endpoints in both
animals and humans (ATSDR, 1999). The adverse neurotoxic effects associated with toluene exposure in the rat study
by Hillefors-Berglund et al. (1995), decreased brain (subcortical limbic area) weight and altered dopamine receptor
binding, occur in areas of the rat brain that are structurally and functionally similar to brain areas (basal ganglia,
thalami) of some human toluene abusers that demonstrate MRI alterations (T2 hypointensity). The altered MRI
parameters may be the result of the partitioning of toluene into the lipid membranes of brain cells (Unger et al., 1994).
If both human and animal adverse effect data on a chemical are available, OEHHA prefers to use the human data to
develop a REL when possible. However, the study by Hillefors-Berglund et al. (1995) provides data (decreased brain
[subcortical limbic area] weight and altered brain dopamine receptor binding) which are specific and sensitive measures
of neurotoxicity that would not be obtainable in human studies. In contrast, the psychometric tests used to generate the
neurotoxicity data in the human occupational exposure studies described above tend to be less sensitive and suffer from
greater measurement uncertainty. Additionally, the Hillefors-Berglund et al. (1995) study has better exposure
characterization than the human occupational exposure studies. Nonetheless, the human studies are useful in supporting
the derivation of the REL for toluene. Ordinarily, an interspecies uncertainty factor of 3 would be applied, in addition to
the human equivalent concentration calculation, to reflect the uncertainty associated with extrapolating from animals to
humans. However, in this case the uncertainty in the interspecies extrapolation is reduced by the availability of human
epidemiological data with generally consistent effect levels, after appropriate duration corrections. Based on
comparison of the data in both animals and humans, it appears that a REL of 271 µg/m
3
(rounded to 300 µg/m
3
in the
final derivation) would protect exposed humans from experiencing chronic neurotoxic effects.
Further studies considered by OEHHA for the derivation of the Reference Exposure are listed in Table 5.1.
Table 5.1: Supporting Neurotoxicity Studies
Study Duration Effect
NOAEL
mg/m
3
NOAEL
(TWA)
mg/m
3
LOAEL
mg/m
3
LOAEL
(TWA)
mg/m
3
VonEuler et al. (1994) 4 weeks rat: altered brain dopamine
receptor binding
n.o. n.o. 302 54
Orbaek and Nise
b
(1989) 29 years human: impairment on
neuropsychometric tests
n.o. n.o. 42-155 15-55
Korsak (1992) 6 months rat: impaired motor function n.o. n.o. 377 67
WHO (2001)
Air Quality Guidelines for Europe 2000
The lowest-observed-adverse-effect level for effects on the CNS from occupational studies, is approximately 332
mg/m
3
(88 ppm) (Foo et al., 1990). A guideline value of 0.26 mg/m
3
is established from these data adjusting for
continuous exposure (dividing by a factor of 4,2) and dividing by an uncertainty factor of 300 (10 for interindividual
variation, 10 for use of a lowest-observed-adverse-effect level rather than a no-observed-adverse-effect level, and an
additional factor of 3, given the potential effects on the developing CNS). This guideline value should be applied as a
weekly average. This guideline value should also be protective for reproductive effects (spontaneous abortions).
The air quality guideline could also be based on the odour threshold. In this case, the peak concentrations of toluene in
air should be kept below the odour detection threshold level of 1 mg/m
3
as a 30 minutes average.
The INDEX project Final report
167
U.S.EPA - IRIS (1992)
Integrated Risk Information System
Determination of the Reference Concentration for Chronic Inhalation Exposure (RfC)
Critical effect Exposures* UF MF RfC
Neurological effects
Occupational Study
Foo et al., 1990
NOAEL: None
LOAEL: 332 mg/m
3
(88 ppm)
LOAEL(ADJ): 119 mg/m
3
LOAEL(HEC): 119 mg/m
3
300 1
0,4 mg/m
3
Degeneration of
nasal epithelium
2-Year Rat Chronic
Inhalation Study
NTP, 1990
NOAEL: None
LOAEL: 2261 mg/m
3
(600 ppm)
LOAEL(ADJ): 437 mg/m
3
LOAEL(HEC): 79 mg/m
3
*Conversion Factors: MW = 92.15.
Foo et al., 1990: Assuming 25 C and 760 mmHg, LOAEL (mg/m
3
) = 88 ppm x 92.15/24.45 = 332 mg/m
3
. This is an extrarespiratory effect of a
soluble vapor. The LOAEL is based on an 8-hour TWA occupational exposure. MVho = 10 m
3
/day, MVh = 20 m
3
/day. LOAEL(HEC) =
LOAEL(ADJ) = 332 x MVho/MVh x 5 days/7 days = 119 mg/m
3
.
NTP, 1990: Assuming 25 C and 760 mmHg, LOAEL (mg/m
3
) = 600 ppm x 92.15/24.45 = 2261 mg/m
3
. LOAEL(ADJ) = LOAEL (mg/m
3
) x 6.5
hours/24 hours x 5 days/7 days = 437 mg/m
3
. The LOAEL(HEC) was calculated for a gas:respiratory effect in the extrathoracic region. MVa = 0.24
m
3
/day, MVh = 20 m
3
/day, Sa (ET) = 11.6 sq.cm, Sh (ET) = 177 sq.cm. RGDR = (MVa/Sa) / (MVh/Sh) = 0.18. LOAEL(HEC) = 437 x RGDR = 79
mg/m
3
.
In humans, toluene is a known respiratory irritant with central nervous system (CNS) effects. Because available studies
could not provide subthreshold (NOAEL) concentrations for either of these effects, the LOAELs for both effects need
to be considered in developing the RfC. Consequently, the study of Foo et al. (1990) was used for the CNS effects, and
that of the National Toxicology Program (NTP, 1990) for the irritant effects. Because the CNS effect was judged to be a
more severe and relevant endpoint, the LOAEL for this effect was used for deriving the RfC. Further, this effect is
supported by a number of other occupational studies that show effects around 377 mg/m
3
(100 ppm).
Foo et al. (1990) conducted a cross-sectional study involving 30 exposed female workers employed at an electronic
assembly plant where toluene was emitted from glue. Toluene levels reported in the study were from personal sample
monitoring and reported as an 8-hour TWA, although the number of samples taken and the actual sampling period were
not given. No historical exposure values were given. Co-exposure to other solvents was not addressed in the study. The
exposed and control cohorts were matched for age, ethnicity, and use of medications. Members of these cohorts did not
use alcohol and were nonsmokers. Medical histories were taken to eliminate any histories of central or peripheral
nervous system disorders. The average number of years (+/- SD) worked by the exposed population was 5,7 +/- 3,2 and
by the controls was 2,5 +/- 2,7. Exposed workers breathed toluene air levels of 332 mg/m
3
(88 ppm) as a TWA and
control workers 49 mg/m
3
(13 ppm) (TWA); both of which are averages of the individual personal samples. A battery
of eight neurobehavioral tests were administered to all exposed and control workers. The tests were performed
midweek, before the workers reported to their stations for the day. Group means revealed statistically significant
differences in 6/8 tests; all tests showed that the exposed workers performed poorly compared with the control cohort.
When individual test results were linearly regressed against personal exposure concentrations, poor concentration-
response relationships resulted for the six tests, with correlation coefficients ranging from 0,44 to 0,30. Irritation effects
were not evaluated in this study, and no clinical signs or symptoms were reported. The paucity of exposure information,
coupled with the small size of the cohort, limits the interpretation of this study, although the results were essentially
confirmed in a clinical study in which the toluene concentrations were carefully controlled (Echeverria et al., 1989) at
levels bracketing 332 mg/m
3
(88 ppm). Although the data in Echeverria et al. (1989) were generated from short- term
exposures (3-7 hours over a period of 142 days), the results may be considered relevant to longer-term exposures as
several studies indicate the absence of a duration-response relationship in toluene-induced symptomatology. Fornazzari
et al. (1983) noted the absence of a duration-effect relationship among toluene abusers when they were segregated into
neurologically impaired vs. unimpaired (p = 0,65). The human studies of Iregren (1982), Cherry et al. (1985), Baelum et
al. (1985), and the principal study of Foo et al. (1990) all report this lack of a duration-response relationship and
confirm the occurrence of CNS effects. Foo et al. (1990) indicate a LOAEL of 332 mg/m
3
(88 ppm) toluene for
neurobehavioral changes from chronic exposure to toluene.
In a 2-year bioassay, Fischer 344 rats (60/sex/group) were exposed to 0, 2261, or 4523 mg/m
3
(0, 600, or 1200 ppm,
respectively) toluene vapors, 6,5 hours/day, 5 days/week (duration-adjusted to 0, 437, and 875 mg/m
3
, respectively) for
103 weeks (NTP, 1990). To generate toluene vapor, the liquid material was heated, and the vapor diluted with nitrogen
and mixed with the chamber ventilation air. An interim sacrifice was carried out at 15 months on control and 4524
The INDEX project Final report
168
mg/m
3
(1200 ppm) groups (10/sex/group) to conduct hematology and histopathology of the brain, liver, and kidney.
Body weights were measured throughout the study. Gross necropsy and micropathology examinations were performed
at the end of the study on all major organs including the nasal passage tissues (three sections), lungs, and mainstem
bronchi. Mean body weights in both exposed groups were not different from controls for either sex. No exposure-
related clinical signs were reported, and survival rate was similar for all groups. At the interim sacrifice, there was a
mild-to- moderate degeneration in the olfactory and respiratory epithelium of the nasal cavity in 39/40 rats of the 600-
and 1200-ppm groups compared with 7/20 controls. At the end of 2 years, there was a significant (p<0,05) increase in
the incidence of erosion of the olfactory epithelium (males: 0/50, 3/50, and 8/49; females: 2/49, 11/50, and 10/50; at 0,
600, and 1200 ppm, respectively) and of degeneration of the respiratory epithelium (males: 15/50, 37/50, and 31/49;
females: 29/49, 45/50, and 39/50; at 0, 600, and 1200 ppm, respectively) in the exposed animals. The females exposed
to 600 and 1200 ppm also exhibited a significant increase in inflammation of the nasal mucosa (27/49, 42/50, and 41/50
at 0, 600, and 1200 ppm, respectively) and respiratory metaplasia of the olfactory epithelium (0/49, 2/50, and 6/50 at 0,
600, and 1200 ppm, respectively). A LOAEL of 2261 mg/m
3
toluene was determined for the concentration-dependent
increase in erosion of the olfactory epithelium in male rats and the degeneration of the respiratory epithelium in both
sexes. No NOAEL could be derived from this study.
ATSDR (2003)
Agency for Toxic Substances and Disease Registry
Derivation of a Minimal Risk Level (MRL):
Studies (Zavalic et al., 1998a; 1988c) demonstrated a statistically significant impairment of color vision in workers
chronically exposed to 588 mg/m
3
(156 ppm) toluene compared with controls. When the data were adjusted to allow for
the confounding effects of alcohol consumption and age, a significant difference due to toluene exposure was also
reported for workers exposed to 132 mg/m
3
(35 ppm) toluene compared with controls. Uncertainty factors used for
MRL derivation: 10 for use of a minimal LOAEL and 10 for human variability
MRL = 132 mg/m
3
x 5 days/7 days x 8 hours/24 hours ÷ 100 = 0.3 mg/m
3
Carcinogenic potential
There is no information from human studies that suggests that toluene has carcinogenic potential. Svensson et al. (1990)
examined a cohort of 1020 toluene-exposed workers who had been employed for a minimum period of three months
during the period 1925–1985. There were no significant increases in tumours and no cumulative dose–response
relationship in workers with an exposure period of at least five years and a latency period of 10 years. Exposure to
benzene had taken place up to the 1960s. Toluene has been shown, however, to hyperphosphorylate rat liver p53, a
tumour suppressor gene (Dees and Travis, 1994). This observation may be of concern, since a reduced ability of p53 to
suppress genetic errors may result in tumour formation.
Genotoxicity
An unequivocal evaluation of the genetic effects of occupational toluene exposure cannot be made because of the small
numbers of individuals analysed and insufficient information on possible exposure to other chromosome-damaging
agents (WHO, 1985). Recent data indicate that toluene induces clastogenic effects in pokeweed-mitogen-stimulated
peripheral blood lymphocytes of printers (Nise et al., 1991). However, variations in exposure history preclude
identification of an exposure–response relationship. Toluene exposure of printers was also highly correlated with an
excess of chromatid breaks in peripheral lymphocytes compared to controls (Pelclova et al., 1990), although concurrent
exposure to printing dyes as a factor cannot be excluded. No effects on sister chromatid exchange, cell cycle delay or
cell mortality were observed in peripheral blood lymphocytes in volunteers exposed for three consecutive days to 189
mg/m
3
(50 ppm) (Richer et al., 1993). Previously, Bauchinger et al. (1982) found a significant increase in sister
chromatid exchanges and chromosome aberrations in printers (smokers and nonsmokers relative to controls) exposed to
toluene for more than 16 years. Even after two years of exposure cessation a higher incidence of aberrations was
observed in exposed individuals compared to controls (Dudek et al., 1990).
Interactions with other chemicals
Exposure of volunteers to toluene 189 mg/m
3
and xylene 189 mg/m
3
, both being often found together in mixtures
such as paint thinners,
resulted in decrements in reaction time in one of a battery of psychomotor and cognitive tests
The INDEX project Final report
169
administered (Dudek et al., 1990). Toluene alone did not result in deficits in any of the tests. Similar
levels of both
solvents (189
mg/m
3
xylene, 151 mg/m
3
toluene) did not modify the conversion of either substance to its
urinary metabolites (Kawai et al. 1992b; Tardif et al. 1991). At higher concentrations (302 or 566
mg/m
3
xylene, 358 or 566 mg/m
3
toluene), the blood and exhaled air concentrations of both solvents were increased
compared to the controls exposed to either solvent alone, indicating that metabolism of both solvents was
decreased by the coexposure paradigm (Tardif et al. 1991, 1992).
Ethanol ingestion during exposure of volunteers to a toluene level of 302 mg/m
3
(80 ppm) for 4,5 hours was found not
to alter the occurrence or severity of the subjective symptoms associated with toluene alone (Iregren et al., 1986).
A number of studies with laboratory animals have shown that toluene interferes with the metabolism and toxicity of
several chemicals, including ethanol, benzene, xylene, hexane and styrene (Chemical and engineering news, 1995;
Plappert et al., 1994; Nylen et al., 1995)
Odour perception
Source: WHO, 1986
Odour threshold: 9,4 mg/m3
Source: American Industrial Hygiene Association, 1989
Characteristic : sour, burnt.
Odour Thresholds:
Detection: 6 mg/m3 ; range 0,6-140 mg/m3 (1,6 ppm; range 0,16 - 37 ppm)
Recognition: 41 mg/m3 ; range 7-260 mg/m3 (11 ppm; range 1,9 - 69 ppm)
Source: (Hoshika et al., 1993)
Barely perceptible concentration level of toluene: 3.5/4.2 mg/m
3
(in Japan/The Nederlands, respectively)
International comparison of odor threshold values of several odorants in Japan and in The Netherlands, given as the
barely perceptible concentration level revealed striking similarities for hydrogen sulfide (in Japan 0.0005 ppm/in The
Netherlands 0.0003 ppm), phenol (0.012/0.010), styrene (0.033/0.016), toluene (0.92/0.99), and tetrachloroethylene
(1.8/1.2) but not for m-xylene (0.012/0.12). Such a similarity was not found with any other literature sources.
The INDEX project Final report
170
Summary of Toluene Dose Response Assessment
________________________________________________________________________________________________
Exposure other than inhalation: Not relevant
Toxicokinetics: ~50% uptake of the inhaled amount
Health effect levels of short- and long-term exposure
NOAEL
mg/m³
LOAEL
mg/m³
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
Short-term exposure
151
EXP
369
1h-ADJ
377
EXP
923
1h-ADJ
CNS-mild, eyes, respiratory;
impaired reaction time, headache, dizziness,
feeling of intoxication, slight eye and nose
irritation
volunteers, 6h
Andersen et al. 1983 OEHHA (1999);
REL: 37
ATSDR (2003);
MRL: 3.8
151
EXP
283
EXP
CNS; NOAEL: see above
LOAEL: visual short term memory, visual
perception and psychomotor skills
volunteers, 6-7h
NOAEL: Andersen et
al. 1983
LOAEL: Echeverria et
al. 1989
ECB (2003)
Long-term exposure
151
EXP
27
ADJ
301
EXP
54
ADJ
CNS; decreased brain weight, dopamine receptor
binding
Supporting study: Neurobehavioral deficits
rats, 4w
(supported by
occupational, 5.7y)
Hillefors-Berglund et
al. 1995; supported by
Orbaek and Nise
(1989), Foo et al.
(1990)
OEHHA (1999);
REL: 0.4
332
EXP
118
ADJ
CNS;
neurobehavioural deficits
Occupational, 5.7y
Foo et al. 1990 OEHHA (1999);
WHO (2001);
GV: 0.26;
EPA-IRIS (1992)
; RfC: 0.4
2261
EXP
437
ADJ
79
HEC
Respiratory system;
nasal epithelium degeneration
rats, 2y
NTP, 1990 EPA-IRIS (1992)
132
Study
31
ADJ
CNS;
color vision impairment
Occupational, >6m Zavalic et al. (1998a) ATSDR (2003);
MRL: 0.3
Carcinogenicity: IARC: 3 ; U.S.EPA: D ; No indications of carcinogenicity
Genotoxicity: No clear indication of genotoxicity
Odour threshold: Range (detection): 0.6 - 140 mg/m³ , GM: 6 mg/m³ (AIHA); 9.4 mg/m³ (WHO);
Barely perceptible concentration level of toluene: 3.5/4.2 mg/m
3
(in Japan/The Nederlands, respectively;Hoshika et al.,
1993)
Susceptible population: No evidence
Remarks: RD
50
: 19 g/m
3
The INDEX project Final report
171
6. Risk Characterization
Health hazard evaluation of short- and long-term exposure
Toluene has low acute toxicity. In humans experimentally exposed to toluene, concentrations of 283 mg/m
3
and above
caused headache, dizziness, and feeling of intoxication, irritation and sleepiness. Furthermore, toluene causes impaired
neuropsychological function, as demonstrated in performance tests (LOAEL: 283 mg/m
3
). For acute effects a NOAEL
of 150 mg/m
3
has been identified (Andersen et al. 1983) and will be taken forward to the risk characterisation. Dividing
by an assessment factor of 10 for interindividual variation, the limit of exposure results in 15 mg/m
3
(see also Table
6.1), applied as a hourly average.
The LOAEL for long-term effects on the CNS from occupational studies is approximately 30 mg/m
3
after adjustment
from intermittent to continuous exposure. Dividing by an assessment factor of 100 (10 for interindividual variation, 10
for use of a LOAEL rather than a NOAEL) a limit of exposure of 0.3 mg/m
3
is obtained (to be applied as a weekly
average; see also Table 6.1), which should also be protective for reproductive effects (spontaneous abortions).
Limited data in humans indicate an increased risk for late spontaneous abortions at dose levels around 332 mg/m
3
.
Human data as well as studies in rats and limited data in mice provide evidence of similar developmental effects, i.e.
lower birth weight, delayed postnatal development and developmental neurotoxicity. Only very high exposure levels
were investigated in humans. In animals, the NOAEL for lower birth weight and delayed postnatal development is 2.3
g/m
3
. A NOAEL for developmental neurotoxicity cannot be determined from the available studies. The LOAEL for this
effect is 4.5 g/m
3
.
There has been no indication that toluene is carcinogenic in bioassays conducted to date and the weight of available
evidence indicates that it is not genotoxic.
Table 6.1: Derivation of toluene short- and long-term limits of exposure (EL)
Effect level - mg/m
3
Assessment
factor
EL
mg/m
3
Toxicological endpoint
Short-term
Exposure Limit
Human
NOAEL
Volunteers
150 10
b
15
Impaired reaction time, headache,
dizziness, feeling of intoxication, slight eye
and nose irritation
Long-term
Exposure Limit
Human
LOAEL
Occupation
al
30 100
ab
0.3 CNS: color vision impairment
a
not considering a NOAEL (10);
b
intraspecies variability (10)
The INDEX project Final report
172
Relevance for the EU-population exposure
Table 6.2: Percentage of population exposed beyond the derived EL and margins of safety
Available exposure data
EL derived
Margins of
Safety (MOS)
Description (Study, Year) N 0.3 mg/m
3
50
th
(90
th
)
Athens 30-h TWA (Expolis, 96-98) 42 5% 7 (1)
Basel 30-h TWA (Expolis, 96-98) 47 < 19 (9)
Helsinki 30-h TWA (Expolis, 96-98) 188 < 23 (7)
Milan 30-h TWA (Expolis, 96-98) 38 < 5 (2)
Oxford 30-h TWA (Expolis, 98-00) 40 < 26 (6)
Prague 30-h TWA (Expolis, 96-98) 46 < 5 (2)
England 28d-TWA (BRE, 97-99) 796 < 20 (6)
German Survey 48-h PEM (GerEs II,
1990-92)
113 5 % 4 (1)
Germany 4w-TWA (GerES IV pilot
study, spring+summer 2001)
44 < 27 (9)
French National Survey 7d-TWA (IAQ
observatory; 2003-04)
110 < 20 (5)
Avg. MOS: 16 (5)
< out of the evaluation range (i.e. <5% of the environments investigated)
Result
Human effects on the central nervous system are considered as the most sensitive effects in both short- and long-term
inhalatory exposure to toluene. Available exposure data indicate that the European population is not experiencing health
effects of concern resulting from the exposure to toluene in their homes. Results from ten monitoring surveys show that
toluene levels in the order of the established exposure limit of 300 µg/m
3
could be reached under worse-case conditions
and in a limited number of urban residences. On average, median concentrations (90th percentile) were found to be 16
(5) times lower than the EL. Also, short-term exposures associated with human indoor activities are not expected to
exceed the acute EL set here at 15.000 µg/m
3
.
The INDEX project Final report
173
Xylenes (ortho-, meta- and para-)
Synonyms: o-xylene (1,2-dimethylbenzene or 2-xylene),
m-xylene (1,3-dimethylbenzene or 3-xylene),
p-xylene (1,4-dimethylbenzene or 4-xylene, also noted as
methyltoluene, benzene-dimethyl, dimethylbenzene)
CAS Registry Numbers: 95-47-6 o-xylene,
108-38-3 m-xylene,
106-42-3 p-xylene
Molecular Formula: C
8
H
10
1. Compound identification
Xylene is an aromatic hydrocarbon, which exists, in three isomeric forms: meta (m-), para (p-) and ortho (o-). Technical
grade xylene contains a mixture of the three isomers. Xylenes are widely used in the chemical industry as solvents for
products such as paints, inks, dyes, adhesives, pharmaceuticals, and detergents (HSDB, 2003). Approximately 92% of
mixed xylenes is blended into gasoline as antiknock agents. It is also used in a variety of solvent applications,
particularly in the paint and printing ink industries. Xylene is a colourless liquid at room temperature with an aromatic
odour. All three isomers evaporate easily to the air from water. In soil and water, the meta and para isomers are
biodegradable, but the ortho isomer is more persistent. p-Xylene is produced in the highest quantities in the U.S. for use
in manufacture of plastics and polymer fibers including mylar and dacron (WHO 1997).
Acute (short-term) inhalation exposure to mixed xylenes in humans results in irritation of the eyes, nose, and throat,
gastrointestinal effects, eye irritation, and neurological effects. Chronic (long-term) inhalation exposure results
primarily in central nervous system (CNS) effects, such as headache, dizziness, fatigue, tremors, and incoordination;
respiratory, cardiovascular, and kidney effects have also been reported. EPA has classified mixed xylenes as a Group D,
not classifiable as to human carcinogenicity. (EPA 2003)
2. Physical and Chemical properties
o-Xylene m-Xylene p-Xylene
Molecular weight (g/mol) 106.16 106.16 106.16
Melting point (°C; 101.3 kPa) -25.2 -47.9 13.3
Boiling point (°C; 101.3 kPa) 144.4 139.1 138.3
Relative density (25°/4°C) 0.876 0.860 0.857
Solubility in water (mg/litre) 142 146 185
Conversion factors at 20 °C and 760 mm Hg:
1 ppm = 4.406 mg/m
3
1 mg/m
3
= 0.227 ppm
Sources: WHO 1997, Verschueren 2001, EPA/Cal. 2003.
3. Indoor Air Exposure assessment
Emission sources
The products that contain and may emit xylenes to indoor air are such as perfumes, pesticide formulations,
pharmaceuticals, adhesives, paints, printed materials, rubber, plastics, leather, polyester fibres, film and fabricated items
(IARC, 1989, ECETOC, 1986; Fishbein, 1988).
The INDEX project Final report
174
Outdoor sources of xylenes are facilities that produce or use xylenes, and exhaust gases from motor vehicles and
through volatilisation from their use as solvents (EPA/Cal. 2003).
Indoor air and exposure concentrations
Mean indoor concentrations of m&p-Xylenes were more than twofold in Northern Europe (Helsinki), almost threefold
in Central Europe (Basel, Praque) and almost twofold in Southern Europe (Milan), compared to respective ambient
concentrations (Annex 3). Xylenes were the second most abundant compounds in indoor air in the EXPOLIS study,
accounting for 18-27% of the target aromatics (Saarela et al 2003). The mean indoor concentrations tend to increase
from north to south being lowest in Helsinki, 7.8 µg/m
3
, and the highest in Milan, 37 µg/m
3
. Lowest mean exposures to
m&p-Xylenes were found in Helsinki, 25 µg/m
3
and the highest in Germany and Athens, 51 µg/m
3
and 55 µg/m
3
,
respectively. Personal exposures were clearly higher than indoor concentrations. The highest personal/indoor ratios
were found in Basel and Helsinki, being over 5 and 3, respectively. Also in all other cities the personal/indoor ratio
exceeded 1, suggesting the presence of important personal sources for m&p-xylenes.
Similar patterns were found for o-Xylene including the highest concentrations in exposures and also higher indoor
concentrations compared to ambient levels. In general, concentrations of o-Xylene were lower than those of m&p-
Xylenes.
TEAM studies carried out in the USA showed maximum 12-hour day time exposures to m&p-Xylenes as high as 1800
µg/m
3
and 460 µg/m
3
in New Jersey in 1981 (Wallace et al 1985) and in Los Angeles in 1987 (Wallace et al 1991),
respectively. Simultaneous ambient air concentrations were typically 10 to 100 times lower. In California, also
residential indoor air concentrations were measured showing a maximum 12-hour concentration of 170 µg/m
3
. Personal
exposure concentration in California showed similar values compared to European distributions.
Maximum 12-hour exposure, 160 µg/m
3
, measured in the TEAM study in California (Wallace et al 1991) agreed also
with European results (Figure ). Instead maximum residential indoor concentration, 68 µg/m
3
, was clearly higher than in
European homes. In New Jersey, maximum daytime 12-hour exposure was as high as 830 µg/m
3
(Wallace et al 1985).
The cumulative distributions of the indoor and 48-hour personal exposure concentrations of m&p- xylenes are presented
in Figure 3.1 and Figure 3.2, and source related short time concentrations are presented in Figure 3.3. m&p -Xylene
concentrations caused by emissions from incense burning were studied by Lee and Wang (2004). Usually,
concentrations caused by incense burning in an 18-m
3
chamber ranged from 0.5 µg/m
3
to 3.8 µg/m
3
, but one type of
incense caused a concentration of 21 µg/m
3
. Brown (2002) studied short time (30-50 min) concentrations of m&p -
Xylene in established and new buildings, reporting mean concentrations of 6.9 µg/m
3
and 30 µg/m
3
respectively. m&p -
Xylene concentration in a new building decreased to 25 µg/m
3
and 2.8 µg/m
3
in 72 and 246 days respectively. Expected
sources of m&p -Xylene in new buildings were building materials. Elke at al (1998) measured short time concentrations
of m&p -Xylene inside buildings (60-min average), inside a train and a car (30-min average). The highest
concentrations were found in a car, the second highest in a train and the lowest in a smoker day room.
Source related short time concentrations of o-Xylene are presented in Figure 3.4, and indoor air and personal exposure
distributions in Figure 3.5 and Figure 3.6. o-Xylene concentrations caused by incense burning were typically less than 5
µg/m
3
, but one type of incense caused a concentration of 96 µg/m
3
. Short time (30-50 min) concentrations of o-Xylene
in established and new buildings were 8.9 µg/m
3
and 32 µg/m
3
respectively. Concentration in a car was about the same
level with a new building, but lower in a train and in a smoker day room. Due to the similar health outcomes, combined
(m-, p-, and o-) indoor air Xylene concentration distributions are presented in Figure 3.7 to be used in the risk
characterisation.
The INDEX project Final report
175
0
20
40
60
80
100
0 50 100 150 200 250
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
Hel
Mil
Oxf
Pra
Figure 3.1. Cumulative frequency distributions of indoor air concentrations of m&p – xylenes in Athens (Ath), Basel
(Bas), Helsinki (Hel), Milan (Mil) Oxford (Oxf) and Prague (Pra) (EXPOLIS 2002).
0
20
40
60
80
100
0 50 100 150 200 250
Exposure (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
GeS
Hel
Oxf
Pra
Figure 3.2. Cumulative frequency distributions of 48-hour personal exposure concentrations of m&p – xylenes in
Athens (Ath), Basel (Bas), Helsinki (Hel), Oxford (Oxf) and Prague (Pra) (EXPOLIS 2002), and 1-week mean
exposures of the German Survey GerES II (GeS) (Hoffman et al 2000).
The INDEX project Final report
176
0
10
20
30
40
50
60
Incence
burning
New buildings In a car Room with
ETS
Train with
ETS
Concentration (µg/m
3
)
m,p-Xylene
Figure 1. Short time m&p -Xylene concentrations related to specific microenvironments or emission sources.
0
20
40
60
80
100
120
Incence
burning
New
buildings
In a car Room with
ETS
Train with
ETS
Concentration (µg/m
3
)
o-Xylene
Figure 2. Short time o-Xylene concentrations related to specific microenvironments or emission sources.
The INDEX project Final report
177
0
20
40
60
80
100
02040
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
Hel
Mil
Oxf
Pra
Figure 3.5. Cumulative frequency distributions of indoor air concentrations of o –Xylenes in Athens (Ath, n=42), Basel
(Bas, n=47), Helsinki (Hel, n=188), Milan (Mil, n=38) Oxford (Oxf, n=40) and Prague (Pra, n=46) (EXPOLIS 2002).
0
20
40
60
80
100
0 10203040506070
Exposure (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
GeS
Hel
Oxf
Pra
Figure 3.6. Cumulative frequency distributions of 48-hour personal exposure concentrations of o –xylenes in Athens
(Ath), Basel (Bas), Helsinki (Hel), Oxford (Oxf) and Prague (Pra) (EXPOLIS 2002), and 1-week mean exposures of the
German Survey GerES II (GeS) (Hoffman et al 2000).
The INDEX project Final report
178
0
20
40
60
80
100
0 50 100 150
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
Hel
Mil
Oxf
Pra
Figure 3.7. Cumulative frequency distributions of indoor air concentrations of m,p and o –Xylenes in Athens (Ath,
n=42), Basel (Bas, n=47), Helsinki (Hel, n=188), Milan (Mil, n=38) Oxford (Oxf, n=40) and Prague (Pra, n=46)
(EXPOLIS 2002).
The INDEX project Final report
179
4. Toxicokinetics
Absorption
The blood/air partition coefficient for the three isomers range from 26.4 to 37.6 (Sato and Nakajima, 1979), indicating
that xylenes entering the body would be readily absorbed into the blood.
Sedivec and Flek (1976a) made direct measurements of the level of absorption of xylenes by subjecting volunteers to
200 or 400 mg/m
3
of either individual isomers or mixtures of the three isomers of xylenes vapor for 8 hours without
interruption and measuring the difference in the concentration of xylenes in the inspired air relative to the amount
expired. The amount of the individual isomers absorbed over time was consistent for all three isomers and ranged from
62.4 to 64.2% of the inhaled volume, reflecting a high solubility of xylenes in blood.
Riihimäki and Savolainen (1980) conducted studies on human subjects both at rest and during exercise to measure the
kinetics resulting from exposure to mixed xylenes. Healthy male subjects were exposed to xylene for 5 days, 6 hours
per day with a 1-hour break at midday and then for an additional 1 to 3 days after a 2-day weekend break. The exposure
scenarios included either constant exposure to 434 or 868 mg/m
3
(100 or 200 ppm) or fluctuating exposure with peaks
of 868 or 1736 mg/m
3
(200 or 400 ppm) that lasted for 10 minutes. The subjects were either sedentary or exercising on
a stationary bike for short periods of time. Regardless of the exposure scenario (constant or fluctuating or with different
xylene concentrations), retention consistently remained around 60% (i.e., 60% of the inhaled xylene was retained in the
blood and 40% was expired). The results indicate that partitioning of xylene between the tissues and the air occurs, but
it is limited by the solubility in the tissue lipids and the rate of passive diffusion through the matrix. Overall, the lowest
uptake rate was noted with 434 mg/m
3
exposure during sedentary conditions (22 µmol/min) and the highest uptake was
seen with the fluctuating concentrations in which the peaks reached 1.7 mg/m
3
during exercise (266 µmol/min). Given
the constant retention values, the two factors that appeared to control the total uptake of xylenes were the ambient
concentration of xylene and ventilation rates of the subjects.
Further studies on xylene uptake by inhalation have been conducted by Ogata et al., 1970; Astrand et al., 1978; Senczuk
and Orlowski, 1978; David et al., 1979.
Distribution
The Kow (octanol/water distribution coefficient) of xylene indicates that xylene is expected to partition primarily into
tissues containing a higher proportion of neutral lipids, such as adipose, liver, and brain tissue.
In their study on the uptake and distribution of ethylbenzene and xylenes, Riihimäki and Savolainen (1980) found that
10–20% of the xylene was distributed to the adipose tissue. Adipose has the highest concentration of neutral fat and the
highest affinity for xylene of all tissues. Therefore, once sequestered in adipose tissue, xylene is expected to have the
lowest rate of metabolism, the slowest movement to blood, and the longest persistence in the body. The concentration of
xylene in gluteal subcutaneous fat was about 10-fold higher than in venous blood following the last day of exposure (5
days exposure + weekend without exposure + 1 day of exposure).
Additional information on the distribution of xylenes in the body is available from Astrand et al. (1978), Engstrom and
Bjurstrom (1978) and Kumarathasan et al. (1998).
Metabolism and elimination
Proposed metabolic pathways for o-xylene are shown in Figure 4.1 as a model for all xylene isomers. The principal
metabolic fate involves oxidation of one of the methyl groups to a methylbenzoic acid derivative via methylbenzyl
alcohol and methylbenzaldehyde intermediates. The methylbenzoic acid derivative is mostly conjugated to glycine,
producing methylhippuric acid derivatives that can be excreted in urine. Conjugation to glucuronic acid is a minor
pathway. Oxidation of the benzene ring to produce xylenols (i.e., dimethylphenols) is expected to be a negligible
metabolic pathway, based on analysis of urinary metabolites. The liver is expected to be the principal site of metabolism
for xylenes.
The INDEX project Final report
180
* o-xylene used as a model for all isomers of xylene
** significant production of glucuronic derivative under conditions of high levels of administration
Figure 4.1. Metabolic pathways for xylenes (Source: Adapted from Ogata et al., 1970; Riihimaki and
Savolainen, 1980; Riihimaki, 1979; Bray et al., 1949; Sedivec and Flek, 1976a,b; Ogata et al., 1980;
Carlsson, 1981; Senszuk and Orlowski, 1978; David et al., 1979)
Riihimäki and Savolainen (1980), in their study of inhalation exposure to xylene by human subjects under sedentary
and physically active conditions, found that 95% of the eliminated xylene was in the form of methylhippuric acid, with
the remainder lost as unmetabolized xylene in expired air. No deposition sites, such as lipid-rich tissues, were studied.
The excretion rate of xylene from the blood followed biphasic, first-order kinetics, with the initial loss of xylene having
a half-life in the venous blood of 0.5–1 hour, followed by a second phase with a half-life of 20–30 hours. The authors
proposed that the two phases representing the rapid loss of xylene from the blood, mostly through conversion to
methylhippuric acid followed by excretion, indicate that well-perfused organs reach equilibrium within minutes and
muscles reach equilibrium within a few hours, whereas adipose tissues may require several days of continuous exposure
to reach equilibrium.
Riihimäki (1979) evaluated the metabolism and excretion of xylene and toluene derivatives in humans. A volunteer was
administered a single dose of 7.4 mmole m-methylbenzoic acid or 7.8 mmole m-methylhippuric acid. Urine was
analyzed for 30 hours following administration for the presence of metabolites. All of the administered xylene
derivatives appeared in urine as methylhippuric acid, indicating that, under the conditions of this study, once xylene has
been oxidized to methylbenzoic acid, the only route of metabolism was as the glycine conjugate.
Following methylbenzoic acid administration, only methylhippuric acid was detected in urine. The rate of loss (i.e,
excretion rate) was greater with the methylhippuric acid treatment than with the methylbenzoic acid treatment. This
study is limited by the fact that it was conducted on a single individual. The determination that loss of xylene is limited
The INDEX project Final report
181
by the availability of glycine suggests that the rate of utilization of glycine may vary with such factors as age and
nutritional status of the individual.
5. Health effects
Differences among individual xylene isomers
Although differences in the toxicity of individual xylene isomers have been detected, no consistent pattern following
inhalation exposure has been identified.
In rats exposed by inhalation for 30 minutes, EC
50
s (the concentrations producing half-maximal decreases in response
rate) for effects on an operant behavior test showed a relative toxicity order of o-xylene > p-xylene > m-xylene, whereas
EC
50
s for a motor performance test showed a toxicity order of p-xylene > o-xylene = m-xylene. The range of EC
50
values among the isomers was not considered large (Moser et al., 1985). In contrast, p-xylene - but not o-xylene or m-
xylene - altered audiometric variables in rats exposed to 7800 mg/m
3
, 6 hours per day, 5 days per week, for 13 weeks
(Gagnaire et al., 2001). A different order of toxicity has been described for effects on motor coordination (rotarod
performance) in rats following 6 hours exposure to concentrations of 13 g/m
3
xylenes: o-xylene > m-xylene > p-xylene
(Korsak et al., 1990). All three isomers caused decreased fetal body weights in rats exposed for 24 hours per day on
GDs 7–14, but o-xylene caused the effect at a lower concentration (1.5 g/m
3
) than did either p-xylene or m-xylene (3
g/m
3
) (Ungváry et al., 1980). No available studies compare the potency of isomers for affecting neurological endpoints
following subchronic or chronic inhalation exposure. Other animal studies that have examined the toxicity of individual
xylene isomers are: Condie et al. (1988) ; Molnár et al. (1986); Fang et al. (1996).
Humans are most likely exposed to a mixture of xylenes rather than to individual isomers. In commercial mixtures the
m-isomer usually predominates (44–70% of the mixture) (Fishbein, 1988; ATSDR, 1995), the exact composition of the
isomers depending on the source. In technical product ethylbenzene is commonly present at significant levels. Thus,
most of the environmental and occupational exposures and toxicological studies are conducted on this mixture of
xylenes containing ethylbenzene. Other minor contaminants of xylenes include toluene and C
9
aromatic fractions.
Effects of short-term exposure
Despite its structural similarity to benzene, xylene does not influence hematopoiesis. Acute controlled exposure studies
have identified self-reported symptoms of irritation (e.g., watering eyes and sore throat) or neurological impairment
(e.g., mild nausea, headache, altered reaction time, altered balance) as potential effects of xylene following inhalation
exposure in humans.
Levels of 434-868 mg/m
3
(100-200 ppm) are associated with nausea and headache; 868-2170 mg/m
3
(200-500 ppm)
with dizziness, irritability, weakness, vomiting, and slowed reaction time; 3480-43000 mg/m
3
(800-10000 ppm) with
lack of muscle coordination, giddiness, confusion, ringing in the ears, and changes in sense of balance; and 43000
mg/m
3
(>10000 ppm) with loss of consciousness (HESIS, 1986). Other documented neurological effects include
impaired short term memory, impaired reaction time, performance decrements in numerical ability, and impaired
equilibrium (dizziness) and balance (Carpenter et al., 1975; Dudek et al., 1990; Gamberale et al., 1978; Riihimaki and
Savolainen, 1980; Savolainen and Linnavuo, 1979; Savolainen and Riihimaki 1981; Savolainen et al., 1979b; 1984;
1985b).
Results from subchronic animal studies identify neurological impairment and possible developmental effects as
potential health hazards from repeated inhalation exposure. Scattered reports of body weight changes and adaptive liver
changes in animals are available, but the results do not consistently identify these effects as potential health hazards.
Nelson et al. (1943) exposed 10 healthy human volunteers for periods of 3 to 5 minutes to estimated concentrations of
434 or 869 mg/m
3
(100 or 200 ppm) technical grade xylene. The subjects reported eye, nose, and throat irritation at 869
mg/m
3
but not at 434 mg/m
3
. A significant area of uncertainty arising from the Nelson et al. (1943) study is the use of
estimated rather than measured exposure concentrations. Carpenter et al. (1975) evaluated eye irritation in 7 human
volunteers exposed for 15 minutes to 460, 1000, 2000, or 3000 mg/m
3
. One volunteer noted mild throat discomfort at
460 mg/m
3
, but not at 2000 mg/m
3
. No subjects reported eye irritation at 460 mg/m
3
(106 ppm).
Hastings et al. (1984) exposed 50 healthy individuals to 434, 869, or 1738 mg/m
3
(100, 200, or 400 ppm) mixed xylenes
for 30 minutes to evaluate eye, nose, and throat irritation. The percent of subjects reporting eye irritation was 56 for
The INDEX project Final report
182
controls (clean air), 60 at 434 mg/m
3
, 70 at 869 mg/m
3
, and 90 at 1738 mg/m
3
. The authors concluded there was no
effect on eye irritation at 434 mg/m
3
because the incidence of irritation was as low as the control group. The data from
Nelson et al (1943), Carpenter et al. (1975), and Hastings et al. (1984) taken together are consistent with a human
NOAEL for eye irritation of about 434 mg/m
3
(100 ppm) for at least a 30-minute exposure.
Exposure of sedentary or exercising subjects to a 10-minute peak concentration of 1736 mg/m³ (400 ppm) resulted in
significantly increased uncontrolled body sway in these subjects. Exposure to 868 mg/m³ (200 ppm) xylene for up to 5
hours did not result in CNS disturbances measured by increased body sway (Laine et al., 1993). Riihimaki and
Savolainen (1980) reported that a single 5-minute exposure to 1736 mg/m³ xylene (isomeric form unknown) resulted in
lightheadedness and inebriation similar to alcohol intoxication. Deleterious effects on EEG, reaction time, body
balance, and manual dexterity were found in 8 healthy volunteers following exposure to 434 mg/m³ (100 ppm) m-
xylene for 6 hours/day for 6 days (Savolainen et al., 1980a).
Exposure of 15 volunteers to 434 mg/m³ technical xylene mixed with 20% ethylbenzene for 70 minutes, including 30
minutes of exercise, resulted in significant impairments in short-term memory and other CNS performance tests
(Gamberale et al., 1978). Because ethylbenzene may have contributed to the CNS effects, definitive conclusions about
the effects of xylene cannot be drawn from this study.
Nine healthy male volunteers were exposed to 868 mg/m³ m-xylene 4 hours a day, with or without exercise for 10
minutes at the beginning of each session (Savolainen et al., 1985a). There were no changes in reaction times, but
average and maximal body sway were decreased in a concentration dependent manner. Exercise had a sway reducing
effect. Male volunteers were exposed to 868 mg/m³ m-xylene vapor for 4 hours a day, either sedentary or with 10
minutes periods of exercise twice a day (Savolainen et al., 1984). The body balance of the subjects was impaired in the
anteroposterior direction. Nine healthy male students were exposed to 868 mg/m³ m-xylene for 4 hours per day at 6-day
intervals over 6 consecutive weeks (Savolainen et al., 1982a). Body sway tended to decrease with exposure. Only minor
electroencephalographic effects were noted on 4 hour exposures to 868 mg/m³ m-xylene exposure, and no other adverse
effects were noted (Seppalainen et al., 1991).
Five volunteers were exposed to 174 mg/m³ (40 ppm) xylene for 7 hours/day, 3 consecutive days/week in an inhalation
chamber. There was an 11-day break between each 3-day session (Mergler and Beauvais, 1992). Individual differences
in olfactory perception thresholds for toluene were noted, but there was no effect of exposure duration.
Summary of short-term exposure effect levels
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
430
Study
215
1h-ADJ
860
Study
430
1h-ADJ
Eye, nose and throat irritation (subjective reports
of)
Volunteers, 30min Hastings et al., 1984
(supp.from other
studies)
OEHHA (1999);
REL: 22
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
OEHHA (1999)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Derivation of the Acute Reference Exposure Level
Study Hastings et al., 1984 (with support from Carpenter et al., 1975;
Nelson et al., 1943)
Study population 50 healthy human volunteers
Exposure method 30 minute exposures to 430, 860 or 1720 mg/m
3
xylene (technical
grade)
Critical effects subjective reports of eye, nose, and throat irritation
LOAEL 860 mg/m
3
NOAEL 430 mg/m
3
(100 ppm)
Exposure duration 30 minutes
Equivalent 1 hour concentration 50 ppm (C1 * 60 min = 100 ppm * 30 min)
LOAEL uncertainty factor 1
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 10
The INDEX project Final report
183
Cumulative uncertainty factor 10
Reference Exposure Level 5 ppm (22 mg/m³, 22,000 µg/m³)
With the possible exception of inconsistently observed developmental endpoints, irritation is the lowest reported human
health effect for xylene.
Effects of long-term exposure
Information on the toxicity of xylenes to humans is almost exclusively limited to case reports of acute exposures and
studies of occupational exposures in which persons often inhaled a mixture of hydrocarbon solvents 8 hours per day, 5-
6 days per week. These studies often have incomplete information on the airborne concentrations of xylene and other
hydrocarbons. One study examining chronic effects in humans from inhalation of predominantly mixed xylenes was
identified (Uchida et al., 1993) and one 4-week controlled exposure study examining the effects of p-xylene exclusively
was identified (Hake et al., 1981).
Xylene exposure has been associated with effects in a number of organ systems including the lungs, skin and eyes;
neurological system; heart and gastrointestinal system; kidney; and possibly the reproductive system.
Pulmonary effects have been documented in occupational exposures to undetermined concentrations of mixed xylenes
(and other solvents) and include labored breathing and impaired pulmonary function (Hipolito 1980; Roberts et al.,
1988). High levels of xylene exposure for short periods are associated with irritation of the skin, eyes, nose and throat
(ATSDR, 1995). Chronic exposure to xylenes has been associated with eye and nasal irritation (Uchida et al., 1993).
Although the mechanism by which xylenes exert their toxic effects on the nervous system and developing fetus are not
completely understood, some theories exist. The CNS toxicity observed during exposure to high concentrations has
been attributed to the liposolubility of xylenes in the neuronal membrane (Desi et al., 1967; Savolainen and Pfaffli,
1980; Tahti, 1992). It has been suggested that xylenes disturb the actions of proteins essential to normal neuronal
function. Changes in levels of various neurotransmitters and lipid composition have been observed in several brain
areas following acute- and intermediate-duration exposure to xylenes (Andersson et al., 1981; Honma et al., 1983). It is
unclear whether these represent direct effects of xylenes or are secondary changes resulting from nonspecific CNS
depression. Some authors have also suggested that metabolic intermediates such as arene oxides or methylbenzaldehyde
may be responsible for the toxic effects of xylenes (Savolainen and Pfaffli, 1980).
Chronic exposure to xylenes (with other hydrocarbons) has been associated with cardiovascular and gastrointestinal
effects. Heart palpitations, chest pain, and abnormal electrocardiogram were noted (Hipolito, 1980; Kilburn et al., 1985)
as were effects on the gastrointestinal system producing nausea, vomiting and gastric discomfort in exposed workers
(Goldie, 1960; Hipolito, 1980; Uchida et al., 1993; Klaucke et al., 1982; Nersesian et al., 1985).
Results of studies of renal effects of xylene are mixed and come from case reports and occupational studies where
multiple chemical exposures are common. The effects from subchronic exposure documented by Hake et al. (1981) and
from chronic exposure documented by Uchida et al. (1993) did not include renal effects. However, Morley et al. (1970)
found increased BUN and decreased creatinine clearance; Martinez et al. (1989) found distal renal tubular acidemia;
Franchini et al. (1983) found increased levels of urinary β-glucuronidase; and Askergren (1981, 1982) found increased
urinary excretion of albumin, erythrocytes, and leukocytes.
Available data (NTP, 1986; Wolfe et al., 1988a, b; Condie et al., 1988) do not consistently identify the kidney and the
liver as a sensitive target of xylenes in animals.
Reproductive effects were documented by Taskinen et al. (1994) who found increased incidence of spontaneous
abortions in 37 pathology and histology workers exposed to xylene and formaldehyde in the work place. The multiple
chemical exposures and the small number of subjects in this study limit the conclusions that can be drawn as to
reproductive effects of xylene in humans.
No hematological effects have been identified in studies where exposure was to xylene only. Previous studies
identifying hematological effects included known or suspected exposure to benzene (ATSDR, 1995; ECETOC, 1986).
One series of case reports identified lowered white cell counts in two women with chronic occupational exposure to
xylene (Hipolito, 1980; Moszczynsky and Lisiewicz, 1983; 1984), although they may also have had multiple chemical
exposures.
Groups of male volunteers (1 to 4 subjects/group) were exposed to p-xylene in a controlled-environment chamber for
7.5, 3, or 1 hr/day, 5 days/week for 4-weeks (Hake et al., 1981). The p-xylene concentration was changed on a weekly
The INDEX project Final report
184
basis starting at 434 mg/m
3
(100 ppm) the first week, followed by 87 mg/m
3
, 652 mg/m
3
, and 434 mg/m
3
(20, 150, and
100 ppm on average, with a range of 50 to 150 ppm) over subsequent weeks. In addition, groups of female volunteers (2
or 3/group) were exposed to 434 mg/m
3
p-xylene for 7.5, 3, or 1 hr/day for 5 days. The volunteers acted as their own
controls, with exposure to 0 ppm p-xylene occurring for two days (males) or one day (females) the week before and the
week after the xylene exposures. No serious subjective or objective health responses, including neurological tests,
cognitive tests and cardiopulmonary function tests were observed. Odor was noted, but the intensity decreased usually
within the first hour of exposure. The authors concluded that p-xylene may have a weak irritating effect on the soft
tissues starting at 434 mg/m
3
, but overall, the small sample size and high variability among the volunteers made all
results difficult to interpret.
The Uchida et al. (1993) study included a relatively large number of workers studied, exposure for an average of 7
years to xylenes predominately and a comprehensive set of medical examinations to document potential effects. A
survey of 994 Chinese workers involved in the production of rubber boots, plastic coated wire and printing processes
employing xylene solvents was carried out. The survey consisted of fitting individual workers with diffusive samplers
for an 8 hour shift. At the end of the 8 hour shift the samplers were recovered for analysis of solvent exposure, and urine
samples were collected for analysis of xylene metabolites. The following day workers answered a questionnaire
concerning subjective symptoms, and blood and urine were collected for analysis. Out of this group of xylene-exposed
workers, 175 individuals (107 men and 68 women) were selected for further study and analysis based on completion of
their health examinations and on results from diffusive samplers showing that xylene constituted 70% or more of that
individual’s exposure to solvents in the workplace. The control population consisted of 241 (116 men and 125 women)
unexposed workers from the same factories or other factories in the same region, of similar age distribution, of similar
time in this occupation (average of 7 years), and having a similar distribution of alcohol consumption and cigarette
usage. The xylene-exposed and unexposed groups were given health examinations which evaluated hematology (red,
white, and platelet cell counts, and hemoglobin concentration), serum biochemistry (albumin concentration, total
bilirubin concentration, aspartate aminotransferase, alanine aminotransferase, gamma-glutamyl transferase, alkaline
phosphatase, leucine aminopeptidase, lactate dehydrogenase, amylase, blood urea nitrogen, creatinine), and subjective
symptoms (survey of symptoms occurring during work and in the previous three months).
Results of analysis of the diffusive samplers showed that workers were exposed to a geometric mean of 61.7 ± 11.3
mg/m
3
(14.2 ± 2.6 ppm) xylene (arithmetic mean of 92.5 ± 93.8 mg/m
3
). This was broken down into geometric means
of 5.2 mg/m
3
o-xylene, 31.7 mg/m
3
m-xylene, 16.5 mg/m
3
p-xylene, 14.8 mg/m
3
ethyl benzene, and 4.5 mg/m
3
toluene.
n-Hexane was rarely present and no benzene was detected. Analysis of data from the health examinations found no
statistically significant difference (p<0.10) between hematology and serum biochemistry values for xylene-exposed and
unexposed populations. The frequency of an elevated ratio of aspartate aminotransferase to alanine transferase and of
elevated ratio of alkaline phosphatase to leucine aminopeptidase was significantly (p<0.01) higher in exposed men than
in the control population of men.
Results of the survey of subjective symptoms found differences in symptoms occurring during work and during a
similar analysis over the preceding three month period, apparently related to effects on the central nervous system and
to local effects on the eyes, nose and throat. The frequency of five symptoms experienced during work was significantly
(p<0.01) elevated in either xylene-exposed men or women including: dimmed vision, unusual taste, dizziness, heavy
feeling in the head, and headache. The frequency of four symptoms experienced during work were significantly
(p<0.01) elevated in both men and women including irritation in the eyes, nasal irritation, sore throat, and floating
sensation. Ten subjective symptoms occurring in the previous three months were significantly (p<0.01) elevated in
exposed men and women including nausea, nightmare, anxiety, forgetfulness, inability to concentrate, fainting after
suddenly standing up, poor appetite, reduced grasping power, reduced muscle power in the extremities, and rough skin.
Dose dependency appeared to exist for 3 subjective symptoms noted during work: irritation in the eyes, sore throat,
floating sensation, and for one symptom occurring in the last three months, poor appetite.
Although the human evidence for persistent effects on the nervous system or other persistent effects from repeated
inhalation exposure to xylenes is inadequate, results from animal studies more clearly identify potential persistent
neurological impairment and possible developmental effects as potential health hazards from repeated inhalation
exposure.
A number of subchronic studies in animals provide evidence for neurological effects following repeated inhalation
exposure to xylenes (Korsak et al., 1992; Korsak et al., 1994; Gralewicz et al. 1995; Gralewicz and Wiaderna 2001;
Pryor et al. 1987; Nylén and Hagman 1994).
The lowest exposure level that produced changes in a number of neurological endpoints was identified in several studies
of rats exposed to 434 mg/m
3
(100 ppm) m-xylene, 6 hours per day, 5 days per week, for 3 months. These studies
observed statistically significant changes in neurobehavioral tests conducted at least 24 hours following cessation of
The INDEX project Final report
185
exposure: decreased rotarod performance indicative of impaired motor coordination (Korsak et al., 1992, 1994),
decreased spontaneous motor activity (Korsak et al., 1992), impaired radial maze performance indicative of a learning
deficit (Gralewicz et al., 1995), and decreased latency to paw lick in the hot plate test, indicating increased sensitivity to
pain (Korsak et al., 1994).
At a lower exposure level (217 mg/m
3
by the same exposure protocol), rats showed statistically significantly decreased
latency in the paw lick response but no statistically significant effects on rotarod performance (Korsak et al., 1994).
Rats exposed to 434 mg/m
3
m-xylene by the same daily protocol for a shorter duration (4 weeks) displayed no
statistically significant changes in tests of radial maze performance, open-field activity, or active avoidance, but paw
lick latency was increased in the hot plate test and step-down time was shortened in 1/6 trials in the passive avoidance
test (Gralewicz and Wiaderna, 2001).
There are no available studies on the possible developmental toxicity of inhaled xylenes in humans, but a number of
studies have examined standard developmental toxicity endpoints and neurobehavioral endpoints in offspring of
animals exposed to mixed xylenes or individual xylene isomers.
The most significant effects on developmental endpoints were decreased fetal body weight or fetal survival in rats at
xylene isomer doses of 1520 or 3041 mg/m
3
(350 or 700 ppm) 24 hours per day (Ungváry et al., 1980) or at mixed
xylenes concentration of 3388 mg/m
3
(780 ppm) 24 hours per day (Ungváry and Tátrai, 1985) and increased abortions
in rabbits exposed to 1000 mg/m
3
(230 ppm) 24 hours per day (Ungváry and Tátrai, 1985). These effects, although of
concern, occurred at concentrations above those at which neurobehavioral effects were reported in adult animals.
Hass and Jakobsen (1993) exposed groups of 36 pregnant Wistar rats to clean air or 868 mg/m
3
(200 ppm) of xylene for
6 h/day on days 4-20 of gestation. There was no sign of maternal toxicity and no decrease in fetal weights and no
increase in soft-tissue or skeletal malformations. A large increase in the incidence of delayed ossification of the os
maxillare of the skull, however, was observed (53% of experimental fetuses as opposed to 2% of the controls). Potential
neurological/muscular changes measured as performance on a rotorod were also noted upon testing of 2-day-old rat
pups.
Developmental and reproductive effects
There are no available studies on the possible developmental toxicity of inhaled xylenes in humans, but a number of
studies have examined standard developmental toxicity endpoints and neurobehavioral endpoints in offspring of
animals exposed to mixed xylenes or individual xylene isomers.
The most significant effects on developmental endpoints were decreased fetal body weight or fetal survival in rats at
xylene isomer doses of 1500 or 3000 mg/m
3
(350 or 700 ppm) 24 hours per day (Ungváry et al., 1980) or at mixed
xylenes concentration of 3400 mg/m
3
(780 ppm) 24 hours per day (Ungváry and Tátrai, 1985) and increased abortions
in rabbits exposed to 1000 mg/m
3
24 hours per day (Ungváry and Tátrai, 1985). These effects, although of concern,
occurred at concentrations above those at which neurobehavioral effects were reported in adult animals.
Gestational exposure of animals to xylenes has resulted in neurodevelopmental effects (Hass et al., 1995; 1997; Hass
and Jakobsen, 1993) and other possible developmental effects (Ungváry et al., 1980; Ungváry and Tátrai, 1985), but
only at levels above those at which neurobehavioral effects in adult male rats were reported.
Exposure of pregnant rats for 6 hours/day on days 4-20 of gestation to 870 mg/m³ (200 ppm) technical (mixed) xylene
resulted in significantly increased incidence of delayed ossification of the skull in the offspring (Hass and Jakobsen,
1993). The rat pups exposed prenatally to 870 mg/m³ xylene displayed significantly decreased motor performance
during adolescence. However, a study using p-xylene showed no significant embryotoxic or developmental effects on
the CNS as measured by acoustic startle response in rats following exposure to 7000 mg/m³ throughout gestation
(Rosen et al., 1986).
The INDEX project Final report
186
Summary of long-term exposure effect levels
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
61.7
Study
22.1
ADJ
Eye irritation, sore throat, floating sensation,
poor appetite
Occupational, 7y Uchida et al. 1993 OEHHA (1999);
REL: 0.7
ATSDR (1995);
MRL: 0.6
217
EXP
39
HEC
434
EXP
78
HEC
CNS; impaired motor coordination
Rats, 3m Korsak et al., 1994 EPA-IRIS (2003)
; RfC: 0.1
868
EXP
Developmental
Rats Hass and Jakobsen,
1993
RIVM (1999)
250
EXP
177
HEC
Developmental Rats Ungváry and Tátrai,
1985
Health Canada
(1991); TC: 0.18
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
OEHHA (1999)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Derivation of the Chronic Reference Exposure Level (REL):
Study Uchida et al. (1993)
Study population 175 xylene-exposed factory workers and control population of 241
factory workers
Exposure method Inhalation
Critical Effects Dose related increase in the prevalence of eye irritation, sore throat,
floating sensation, and poor appetite.
LOAEL 14.2 ppm (geometric mean of exposure concentrations)
NOAEL Not applicable
Exposure continuity 8 hr/d (10 m3/day occupational inhalation rate), 5 d/wk
Exposure duration Occupational exposure for an average of 7 years
Average occupational exposure 5.1 ppm for LOAEL group (14.2 x 10/20 x 5/7)
Human equivalent concentration 5.1 ppm for LOAEL group
LOAEL uncertainty factor 3
Subchronic uncertainty factor 1
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 10
Cumulative uncertainty factor 30
Inhalation reference exposure level 0.2 ppm (200 ppb; 0.7 mg/m3; 700 µg/m3) for mixed xylenes or for
total of individual isomers
A number of issues are important in considering the uncertainty associated with this REL. For ATSDR (1995), the
animal and human toxicity data suggest that mixed xylenes and the different xylene isomers produce similar effects,
although different isomers are not equal in potency for producing a given effect. Therefore exposure of workers to a
mix of xylenes in the Uchida et al. (1993) study would be expected to generate a similar spectrum of responses as
exposure to single isomers, however the intensity of particular effects could be different.
The use of a neurological endpoint for derivation of a REL is supported by the large number of inhalation and oral
studies, which associate neurological effects with xylene exposure. ATSDR (1995) indicates that neurological effects
are a sensitive endpoint. The observation that floating sensation is apparently related to dose, further supports the
concept that this subjective symptom related to neurological effects was due to xylene exposure.
A UF of 3, rather than 10, was applied for the LOAEL to NOAEL extrapolation due to the generally mild adverse
effects observed and the principally low incidence (<50%) of the effects. A factor of 1 was used for subchronic
uncertainty. Although the average occupational exposure was only 7 years, there were 176 xylene-exposed workers of
average age 29.7 ± 9.0 years (arithmetic mean ± SD) for whom, according to the report, there had been essentially no
change in workplace in their working life. Thus, many workers would likely have been exposed for more than 8.4 years,
the cut-off point for chronic human exposure. Another issue is the use of diffusive samplers in the Uchida et al. (1993)
study. These samplers provide a time weighted average concentration of hydrocarbon and cannot indicate the maximum
concentrations a worker is exposed to. It is unknown whether peak concentrations alter the response to xylenes in
humans.
The INDEX project Final report
187
For comparison with the proposed REL of 0.7 mg/m
3
(200 ppb) based on human studies, (1) the free-standing NOAEL
of 339 mg/m
3
(78 ppm) o-xylene obtained by Jenkins et al. (1970) in rats and guinea pigs continuously exposed for 90
days was used to estimate a REL based on animal data. Use of an RGDR of 1, a subchronic UF of 3, an interspecies UF
of 3, and an intraspecies UF of 10 results in a REL of 3.5 mg/m
3
(800 ppb) for o-xylene for systemic effects. (2) Tatrai
et al. (1981) found a free standing LOAEL of 4761 mg/m
3
(1096 ppm) o-xylene for body weight gain in male rats
exposed every day for 8 hours. Time adjustment to continuous exposure and use of an RGDR of 1, a LOAEL UF of 3
for a mild effect, an interspecies UF of 3, and an intraspecies UF of 10 result in a REL of 17.4 mg/m
3
(4000 ppb). (3)
Ungváry and Tátrai (1985) exposed mice by inhalation continuously to 521 or 1000 mg/m
3
(120 or 230 ppm) xylene for
24 h/day on days 7-15 of gestation. The LOAEL was 1000 mg/m
3
and the NOAEL was 521 mg/m
3
. No time adjustment
is needed. Use of an RGDR of 1, a subchronic UF of 1, an interspecies UF of 3, and an intraspecies UF of 10 results in
a REL of 17.4 mg/m
3
(4000 ppb) for xylene for developmental effects.
ATSDR (2003)
Agency for Toxic Substances and Disease Registry
Derivation of the Minimal Risk Level (MRL):
Using the most well defined Uchida et al. (1993) study, ATSDR estimated a chronic inhalation MRL of 0.6 mg/m
3
from
a LOAEL of 61 mg/m
3
in humans for less serious effects; divided by a total uncertainty factor of 100 (10 for use of a
LOAEL and 10 for human variability).
U.S.EPA - IRIS (2003)
Integrated Risk Information System
Determination of the Reference Concentration for Chronic Inhalation Exposure (RfC)
Critical effect Experimental doses* UF MF RfC
Impaired motor coordination
(decreased
rotarod performance)
Subchronic inhalation study in
male rats (Korsak et al., 1994)
NOAEL: 50 ppm
NOAEL
(HEC)
: 39 mg/m
3
LOAEL: 100 ppm
LOAEL
(HEC)
: 78 mg/m
3
300 1 0.1 mg/m
3
*Conversion Factors and Assumptions - MW = 106.17. Assuming 25°C and 760 mmHg, NOAEL(mg/m
3
) = 50 ppm x 106.17/24.45 = 217 mg/m
3
.
NOAEL
[ADJ]
= 217 mg/m
3
x 6 hrs/day x 5 days/7 days = 39 mg/m
3
. The NOAEL*
[HEC]
was calculated for extrarespiratory effects of a Category 3 gas
(U.S. EPA, 1994). Blood/gas partition coefficients: H(b/g)
rat
= 46.0; H(b/g)
human
=26.4 (Tardif et al., 1995). (H
b/g
)
rat
/(H
b/g
)
human
=1.7; value of 1 is used
when the ratio is >1 (U.S. EPA, 1994). NOAEL*
[HEC]
= NOAEL
[ADJ]
x (H
b/g
)
rat
/(H
b/g
)
human
= 39 mg/m
3
.
The subchronic rat study by Korsak et al. (1994) was selected as the principal study for derivation of the RfC. A
NOAEL of 217 mg/m
3
(50 ppm) and a LOAEL of 434 mg/m
3
(100 ppm) were identified for decreased rotarod
performance (impaired motor coordination). This neurologic test was administered 24 hours after termination of the
exposure period, when xylenes would be expected to have been eliminated from the body. Other subchronic rat studies
provide support for the finding that 434 mg/m
3
exposure produces statistically significant changes in a number of
neurological endpoints: decreased rotarod performance (Korsak et al., 1992), decreased spontaneous motor activity
(Korsak et al., 1992), and impaired radial maze performance indicative of a learning deficit (Gralewicz et al., 1995).
Additional support for 434 mg/m
3
as an exposure level that may produce mild neurologic deficits comes from the report
(Gralewicz and Wiaderna, 2001) that rats exposed to 434 mg/m
3
m-xylene for 4 weeks showed shortened step-down
time in 1/6 trials in the passive avoidance test 50 days postexposure. These studies collectively identify 434 mg/m
3
as
the lowest reliable subchronic animal LOAEL and 217 mg/m
3
as a NOAEL for deficits in neurologic endpoints.
The NOAEL of 217 mg/m
3
was duration adjusted to 39 mg/m
3
, and a NOAEL[HEC] of 39 mg/m
3
was calculated on the
basis of species differences in blood/gas partition coefficients. The NOAEL[HEC] of 39 mg/m
3
was divided by a total UF
of 300 (10
1/2
x 10 x 10
1/2
x 10
1/2
; 3 for animal-to-human extrapolation using default dosimetric adjustments, 10 for
intrahuman variability, 3 for extrapolation from subchronic to chronic duration, and 3 for deficiencies in the data base,
including lack of studies in two species [available studies are predominantly in male rats] and a two-generation
reproductive toxicity study) to give the RfC of 0.1 mg/m
3
. Alternative approaches using available PBPK models to
extrapolate rat exposure concentrations to HECs arrived at similar points of departure for the RfC as the NOAEL[HEC] of
39 mg/m
3
.
The INDEX project Final report
188
Health Canada (1991)
Determination of a critical effect:
In inhalation studies conducted to date, xylenes have not been teratogenic but have induced fetotoxic effects, sometimes
at doses below those that were toxic to the mothers. With the exception of unconfirmed results reported by Mirkova et
al. (1983), the lowest concentration of xylenes reported to induce fetotoxic effects in the absence of maternal toxicity in
available investigations was 500 mg/m
3
. At this concentration, in an incompletely documented study, moderate
embryotoxic effects such as retardation of skeletal development and of body weight increase were observed in the
offspring of rabbits exposed continuously on days 7 to 20 of gestation to xylenes of unspecified composition (Ungváry
and Tátrai, 1985). In rats, maternal toxicity and fetotoxicity were observed following exposure to 250 mg/m
3
on days 7
to 15 of pregnancy, the lowest concentration administered, indicating that rats may be a more sensitive species
(Ungváry and Tátrai, 1985).
Derivation of a tolerable concentration (TC):
Inhalation is considered to be the most important route of exposure to xylenes for the general public. A provisional
tolerable concentration (TC) for xylenes has been derived on the basis of the results of studies in animal species
exposed by inhalation. Based on reported effect levels, developmental toxicity was considered the critical endpoint. The
lowest concentration at which fetotoxic effects in the absence of maternal toxicity have been observed following
exposure by inhalation to xylenes was 500 mg/m
3
in rabbits in a limited study (Ungváry and Tátrai, 1985); maternal and
fetal toxicity were observed in rats at the lowest administered concentration (250 mg/m
3
).
A provisional TC of 0.18 mg/m
3
has been derived, therefore, on the basis of the lowest LOEL (250 mg/m
3
) for
meaningful effects reported in a bioassay of adequate quality (though documentation was incomplete) in the most
sensitive species (Ungváry and Tátrai, 1985). This was adjusted for the ratio of inhalation volume to body weight of rats
[(0.11 m
3
/day)/0.35 kg] to humans aged 5 to 11 years [(12 m
3
/day)/27 kg]. An uncertainty factor of 1000 was applied
[10 for intraspecies variation, 10 for interspecies variation, 10 for LOEL rather than NOEL (although observed effects
at the LOEL were only moderately fetotoxic; documentation was also limited)]. No additional factor was incorporated
for the limited period of exposure since fetotoxic effects occur at doses below those which induce adverse effects in
subchronic and chronic studies.
The lowest concentration of the individual xylene isomers reported to induce adverse effects in studies in animal species
following inhalation is 150 mg/m
3
, in which embryo- and fetotoxic effects were observed in the absence of maternal
toxicity following continuous exposure of pregnant rats to p-xylene on days 7 to 14 of gestation (Ungváry et al., 1980).
This is only slightly less than the LOEL used above in the derivation of a TC for xylenes (177 mg/m
3
).
Carcinogenic potential and genotoxicity
Associations between occupational exposure to xylenes and increased risk of leukemia (Arp et al., 1983; Wilcosky et
al., 1984), non-Hodgkin's lymphoma (Wilcosky et al., 1984), and cancer of the rectum (Gérin et al., 1998), colon (Gérin
et al., 1998), or nervous system (Spirtas et al., 1991) have been reported. However, a number of limitations preclude the
usefulness of these data, including small sample sizes, no quantified exposure concentrations, and/or concurrent
exposures to other solvents.
Adequate human data on the carcinogenicity of xylenes are not available and the available animal data are inconclusive
as to the ability of xylenes to cause a carcinogenic response. Evaluations of the genotoxic effects of xylenes have
consistently given negative results.
The genotoxicity of commercial xylenes and all three individual isomers has been studied and the results are, for the
most part, negative (IARC, 1989). All studies cited in the GENE-TOX database are negative with the exception of one
study for which no conclusion was drawn. Xylenes are not mutagenic in bacterial test systems with Salmonella
typhimurium (Bos et al., 1981; Florin et al., 1980; NTP, 1986) and Escherichia coli (McCarroll et al., 1981) or in
cultured mouse lymphoma cells (Litton Bionetics, 1978). Xylenes do not induce chromosomal aberrations or sister
chromatid exchanges in Chinese hamster ovary cells (Anderson et al., 1990) or cultured human lymphocytes (Gerner-
Smidt and Friedrich, 1978), chromosomal aberrations in rat bone marrow (Litton Bionetics, 1978), micronuclei in
mouse bone marrow (Mohtashamipur et al., 1985), or sperm head abnormalities in rats (Washington et al., 1983).
Technical grade xylenes, but not o- and m-xylene, are weakly mutagenic in Drosophila recessive lethal tests (Donner et
al., 1980). No increase in the frequency of sister chromatid exchanges was observed in peripheral lymphocytes in
individuals exposed to xylenes in an occupational setting (Haglund et al., 1980; Pap and Varga, 1987) or an
experimental setting (Richer et al., 1993).
The INDEX project Final report
189
Interactions with other chemicals
The interaction of xylene with alcohol, drugs (aspirin, phenobarbitol), and various solvents (1,l,ltrichlorethane, benzene,
toluene, ethylbenzene, methyl ethyl ketone) has been evaluated in experimental studies with humans and animals.
Xylene has a high potential to interact with numerous substances because the isomers induce microsomal enzymes in
the liver (Blanchard and Morris 1994; Liira et al. 1991), while microsomal enzymes in the lungs are inhibited by xylene
exposure (Blanchard and Morris 1994; Elovaara et al. 1987; Pate1 et al. 1978; Silverman and Schatz 1991; Toftgard and
Nilsen 1982). Which enzymes will be affected is isomer dependent. For example, m-xylene is a more potent inducer of
P-450 2B enzymes than p-xylene (Backes et al. 1993). The isomer differences, as well as organ differences in effects on
xenobiotic metabolizing enzymes, make it difficult to predict the interaction of xylene with other substances.
Acute inhalation exposure to a mixture of toluene and xylene resulted in more than additive respiratory and central
nervous system toxicity (Korsak et al. 1988, 1992). Elevated blood levels of xylene and toluene and decreased excretion
of the major metabolites of xylene and toluene in the urine (Tardif et al. 1992) suggest mutual metabolic inhibition.
However, simultaneous exposures in humans indicate that a threshold exists for this interaction (Tardif et al. 1991). No
increase in blood levels of these substances was observed during combined exposures to 50 ppm toluene and 40 ppm
xylene over 3 consecutive days, whereas increases in blood levels and levels in exhaled air were observed during a
combined 4-hour exposure to 95 ppm toluene and 80 ppm xylene. Thus, combined exposures at below threshold level
are unlikely to produce greater than additive toxicity (Tardif et al. 1991). A physiologically based toxicokinetic
modeling study using rat data suggests that the interaction between toluene and xylene is competitive, with toluene a
more potent inhibitor of xylene metabolism than xylene is of toluene metabolism (Tardif et al. 1993a, 1993b).
Exposure to xylene combined with benzene or ethylbenzene may also produce mutual inhibition of the metabolism of
both solvents (Engstrom et al. 1984; Nakajima and Sato 1979b). Ethylbenzene is found in commercial xylene. In
contrast, ethyl acetate exposure in combination with exposure to
m-xylene caused a reduction in blood xylene levels
(Freundt et al. 1989).
Possibly because of competition for the enzymes involved in conjugation with glycine during the concurrent
metabolism of
m-xylene and aspirin by human volunteers, saturation of the conjugation pathway occurred that led to
decreases in the metabolism of both aspirin and
m-xylene (Campbell et al. 1988). Administration of aspirin to pregnant
rats during inhalation exposure to xylene caused greater than additive potentiation of maternal and fetal toxic effects
(Ungváry 1985). This was postulated to be due to the interference with metabolism of aspirin by xylene and vice versa.
Inhalation of
m-xylene following pretreatment with phenobarbital was associated with both increased pulmonary
retention of
m-xylene and increased urinary excretion of m-methylbenzoic acid (David et al. 1979). Surprisingly,
inhalation of
m-xylene and l,l,l-trichloroethane has been associated with slight improvements in certain
psychophysiological parameters, including reaction time and equilibrium in humans as compared with pre-exposure
measurements (Savolainen et al. 1982a, 1982b), and impairment in others such as visual evoked potentials and
equilibrium (Savolainen et al. 1982a; Seppalainen et al. 1983).
Ethanol appears to inhibit the metabolism of xylene, resulting in elevated blood levels of xylene and decreased
excretion of methylhippuric acid (Elovaara et al. 1980; Riihimaki et al. 1982a, 1982b; Romer et al. 1986; Savolainen
1980; Savolainen et al. 1978, 1979b, 1980b). A kinetic study in rats (Kaneko et al. 1993) suggests that ethanol
inhibition of xylene metabolism occurs only at high concentrations (500 ppm). Paradoxically, ethanol pretreatment
causes additive effects with xylene in inducing microsomal enzymes in the liver (Wisniewska-Knypl et al. 1989). This
would enhance the metabolic capacity of the liver and modify biological effects of other chemicals that are either
detoxified or converted to toxic metabolites by the microsomal enzymes. In summary, it cannot be stated with certainty
whether alcohol and xylene would interact to produce synergistic or antagonistic effects in humans and animals because
there are reasons why both would occur.
Odour perception
Source: ATSDR (1995)
Odor thresholds in air
Mixture: 4.3 mg/m
3
(1.0 ppm)
m-isomer: 16 mg/m
3
(3.7 ppm)
o-isomer: 0.34-0.74 mg/m
3
(0.08–0.17 ppm)
p-isomer: 2.0 mg/m
3
(0.47 ppm)
The INDEX project Final report
190
Source: Carpenter et al. (1975)
Mixed xylenes: 4.3 mg/m
3
(1 ppm) (detection)
Source: AIHA (1989)
Mixture:: 87 mg/m
3
(20 ppm) (detection);: 174 mg/m
3
(40 ppm) (recognition)
m-Xylene: 1.5 – 4.8 mg/m
3
(0.35 - 1.1 ppm) (detection)
o-Xylene: 23 mg/m
3
(5.4 ppm) (detection)
p-Xylene: 9.1 mg/m
3
(2.1 ppm) (detection)
Summary of Xylenes Dose Response Assessment
________________________________________________________________________________________________
Exposure other than inhalation: not relevant
Toxicokinetics: ~60% uptake of the inhaled amount (no difference among isomers); accumulates in adipose tissue of
chronically exposed
Health effect levels of short- and long-term exposure
NOAEL
mg/m³
LOAEL
mg/m³
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
Short-term exposure
430
Study
215
1h-ADJ
860
Study
430
1h-ADJ
Eye, nose and throat irritation (subjective reports
of)
Volunteers, 30min Hastings et al., 1984
(supp.from other
studies)
OEHHA (1999);
REL: 22
Long-term exposure
61.7
Study
22.1
ADJ
Eye irritation, sore throat, floating sensation,
poor appetite
Occupational, 7y Uchida et al. 1993 OEHHA (1999);
REL: 0.7
ATSDR (1995);
MRL: 0.6
217
EXP
39
HEC
434
EXP
78
HEC
CNS; impaired motor coordination
Rats, 3m Korsak et al., 1994 EPA-IRIS (2003)
; RfC: 0.1
868
EXP
Developmental
Rats Hass and Jakobsen,
1993
RIVM (1999)
250
EXP
177
HEC
Developmental Rats Ungváry and Tátrai,
1985
Health Canada
(1991); TC: 0.18
Carcinogenicity: IARC: 3; inadequate evidence in both humans and experimental animals
Genotoxicity: Studies consistently negative
Odour threshold: Mixture of isomers: 4.3 mg/m³ (ATSDR, Carpenter); 0.25 - 0.3 mg/m³ (Devos)
Susceptible population: No evidence
Remarks: RD
50
: 6.1 g/m
3
; Although differences in the toxicity of individual xylene isomers have been detected, no
consistent pattern following inhalation exposure has been identified; nevertheless, human exposure most likely occurs
to the mixture of isomers
The INDEX project Final report
191
6. Risk Characterization
Health hazard evaluation of short- and long-term exposure
Animal and human toxicity data suggest that mixed xylenes and the different xylene isomers produce similar effects,
although different isomers are not equal in potency for producing a given effect. Therefore exposure to a mix of xylenes
in occupational studies would be expected to generate a similar spectrum of responses as exposure to single isomers,
however the intensity of particular effects could be different.
The use of a neurological endpoint for the derivation of an exposure limit EL is supported by the large number of
inhalation and oral studies, which associate neurological effects with xylene exposure. The observation that floating
sensation is apparently related to dose, further supports the concept that this subjective symptom related to neurological
effects was due to xylene exposure.
Using a well defined occupational study (Uchida et al. (1993), a chronic inhalation EL of 0.2 mg/m
3
was derived from a
LOAEL of 62 mg/m
3
in humans for generally mild adverse effects observed and principally low incidence (<50%). This
LOAEL was adjusted to 22 mg/m
3
accounting for intermittent to continuous exposure and divided by a total assessment
factor of 100 (10 for use of a LOAEL and 10 for human variability). The endpoint considered was: dose related increase
in the prevalence of eye irritation, sore throat, floating sensation, and poor appetite.
An acute EL was derived refering on a study on volunteers and subjective reports of eye, nose, and throat irritation as
shown in Table 6.1.
Table 6.1: Derivation of short- and long-term exposure limits (EL)
Effect level - mg/m
3
Assessment
factor
EL
mg/m
3
Toxicological endpoint
Short-term
Exposure
Limit
Human NOAEL
Volunteers
200 10
b
20
Eye, nose and throat irritation (subjective
reports of)
Long-term
Exposure
Limit
Human LOAEL
Occupational
22 100
ab
0.2
Dose related increase in the prevalence of
eye irritation, sore throat, floating
sensation, and poor appetite
a
not considering a NOAEL (10);
b
intraspecies variability (10)
Relevance of EU-population exposure to xylenes
Table 6.2: Percentage of population exposed beyond the derived EL and margins of safety
Available exposure data
Xylenes EL
Margins of
Safety (MOS)
Description (Study, Year) N 0.2 mg/m
3
50
th
(90
th
)
Athens 30-h TWA (Expolis, 96-98) 42 < 7 (4)
Basel 30-h TWA (Expolis, 96-98) 47 < 25 (10)
Helsinki 30-h TWA (Expolis, 96-98) 188 < 25 (10)
Milan 30-h TWA (Expolis, 96-98) 38 < 6 (2)
Oxford 30-h TWA (Expolis, 98-00) 40 < 32 (7)
Prague 30-h TWA (Expolis, 96-98) 46 < 10 (5)
German Survey 48-h PEM (GerEs II,
1990-1992)
113 < 10 (3)
The INDEX project Final report
192
French National Survey 7d-TWA (IAQ
observatory; 2003-04)
102 < 20 (5)
Avg. MOS: 18 (6)
< out of the evaluation range (i.e. <5% of the environments investigated);
Result
A chronic exposure limit of 200 µg/m
3
has been derived based on generally mild adverse effects associated with CNS
and increase in the prevalence of eye irritation and sore throat. The results of eight monitoring surveys (Table 6.2)
indicate that background levels of xylenes in European residences are of no concern to human health since median (90th
percentile) levels are, on average, 20 (6) times lower than the EL established. Acute exposure data indicate that it is
very unlikely that xylenes emissions associated with human indoor activities would generate levels in the order of the
proposed short-term EL of 20 mg/m
3
, considered protective for irritative effects in the general population. Although
human exposure most likely occurs to the mixture of xylene isomers, animal and human toxicity data suggest that
mixed xylenes and the different xylene isomers produce similar effects.
The INDEX project Final report
193
7. Aromatic compounds: Comparison of relevant data
Short-term exposure : derivation of limits of exposure (EL)
Effect level - mg/m
3
Assessment
factor
EL
mg/m
3
Toxicological endpoint
Toluene
Human NOAEL
Volunteers
150 10
b
15
Impaired reaction time, headache,
dizziness, feeling of intoxication, slight eye
and nose irritation
Styrene
Human NOAEL
Volunteers
217 100
bc
2 Eye and throat irritation
Xylenes
Human NOAEL
Volunteers
200 10
b
20
Eye, nose and throat irritation (subjective
reports of)
a
not considering a NOAEL (10);
b
intraspecies variability (10) ;
c
susceptible population (10)
Long-term exposure : derivation of limits of exposure (EL)
Effect level - mg/m
3
Assessment
factor
EL
mg/m
3
Toxicological endpoint
Toluene
Human LOAEL
Occupational
30 100
ab
0.3 CNS: color vision impairment
Styrene
Human LOAEL
Occupational
25 100
ab
0.25
CNS; neuropsychological tests: reaction
time, s/l term logic memory, s/l term verbal
memory, digit-symbol association, block
design and figure identification.
Xylenes
Human LOAEL
Occupational
22 100
ab
0.2
Dose related increase in the prevalence of
eye irritation, sore throat, floating
sensation, and poor appetite
a
not considering a NOAEL (10);
b
intraspecies variability (10) ;
c
susceptible population (10)
Percentage of population exposed beyond given threshold limits
EXPOSURE DATA
Toluene EL Styrene EL Xylenes EL
Aromatic HC
Mixture
Description (Study, Year) N 0.3 mg/m
3
0.25 mg/m
3
0.2 mg/m
3
refer to text*
Athens 30-h TWA (Expolis, 96-98) 42 5 % < < 20%
Basel 30-h TWA (Expolis, 96-98) 47 < < < <
Helsinki 30-h TWA (Expolis, 96-98) 188 < < < <
Milan 30-h TWA (Expolis, 96-98) 38 < < < 35%
Oxford 30-h TWA (Expolis, 98-00) 40 < < < 5%
Prague 30-h TWA (Expolis, 96-98) 46 < < < 15%
England 28-d TWA (BRE, 2002) 796 < n.a. n.a.
German Survey 48-h PEM (GerEs II,
1992-93)
113 5 % < <
French National Survey 7d-TWA (IAQ
observatory; 2003-04)
102 -
110
< < <
< out of the evaluation range (i.e. <5% of the environments investigated);
* see reciprocal calculation procedure
The reciprocal calculation approach for mixtures
A further element is included, based on a ACGIH approach commonly used in occupational settings to describe
Threshold Limit Values (TLVs) resulting from the exposure to solvent mixtures. Supposing the additivity of substances
having similar toxicologic effects, an exposure limit for aromatic hydrocarbons could be defined as follows :
[Benzene]/EL
Benz
+ [Toluene]/EL
Tol
+ [Styrene]/EL
Styr
+ [Xylenes]/EL
Xyls
1
with EL
i
= single Exposure Limits considering neurologic endpoints
The INDEX project Final report
194
0
10
20
30
40
50
60
70
80
90
100
012345
Neurological Endpoint:
Animal LOAEL
Benz
= 2.5 mg/m
3
TC = 0.025 mg/m
3
Human LOAEL
Tol
= 31 mg/m
3
GL = 0.3 mg/m
3
Human LOAEL
Styr
= 25 mg/m
3
GL = 0.25 mg/m
3
Animal NOAEL
Xyls
= 39 mg/m
3
TC = 0.4 mg/m
3
TC = tolerable concentration
GL = indoor guideline value proposed
Athens
Basel
Helsinki
Milan
Oxford
Prague
concentration relative to proposed guidelines
Percentile
In order to consider overall neurologic endpoints, the following critical effects and limits were chosen for benzene and
the xylenes (toluene and styrene were still classified as neurologic toxicants in the present RA).
Benzene: increased rapid response time, Animal LOAEL: 2.5 mg/m
3
, Assessment factor: 100, EL: 0.025
Xylenes: impaired motor coordination, Animal NOAEL: 39 mg/m
3
, Assessment factor: 100, EL: 0.4
Figure 6.1: Cumulative frequency distribution of EXPOLIS data, applying the
reciprocal calculation approach on a mixture of four aromatic hydrocarbons
The INDEX project Final report
195
Styrene
Synonyms: phenethylene, phenylethene, phenylethylene, styrol, styrole, styrolene,
vinylbenzene, vinylbenzol, cinnamene, cinnamol
CAS Registry Numbers: 100-42-5
Molecular Formula: C
8
H
8
1. Compound identification
Styrene is a colourless, viscous liquid with a pungent odour. It is one of the most important monomers, worldwide
because of its common use in the production of polystyrene, acrylonitrile-butadiene-styrene resins, styrene-butadiene
rubbers and latexes, and in the reinforced plastics industry. Styrene has been produced since the 1920s by catalytic
dehydrogenation of ethylbenzene.
2. Physical and Chemical properties
Molecular weight (g/mol) 104.15
Melting point (°C) -30.6
Boiling point (°C) 145.2
Density (at 20 °C, 1 atm) 0.91
Relative density (air =1) 3.6
Solubility: slightly soluble in water, soluble in ethanol
and very soluble in benzene and petroleum ether
Conversion factors at 20 °C and 760 mm Hg:
1 ppb = 4.323 µg/m
3
1 µg/m
3
= 0.231 ppb
Sources: (WHO 1983, WHO 2000, Verschueren 2001, EPA/Cal 2003)
3. Indoor Air Exposure assessment
Emission sources
Styrene exposures, at levels of milligrams per day, have been measured in the manufacture of styrene-based plastics and
rubbers and fibre glass-reinforced polyester products and at much higher levels in the glass fibre-reinforced plastics
industry. General population is typically exposed to levels of micrograms per day resulted from inhalation of ambient
air and cigarette smoke, emissions from newly installed carpets containing a styrene-butadiene rubber latex adhesive,
and from intake of food that has been in contact with styrene-containing polymers (WHO, 1983, IARC 1994, IARC
2000, EPA/Cal 2003).
Natural styrene sources are such as cinnamic acid containing plants, e.g. balsamic trees, and a by-production of fungal
and microbial metabolism. Ambient air concentration of styrene is relatively low compared with toluene and xylene,
because of its reactivity with ozone to yield irritants such as benzaldehyde and peroxides such as peroxybenzyol nitrate,
which is a potent eye irritant. (WHO 2000).
The INDEX project Final report
196
Indoor air and exposure concentrations
Usually indoor styrene concentrations were low, but higher than respective outdoor concentrations ranging 1-6 µg/m
3
and 1-2 µg/m
3
, respectively (Table 4.0.1). Arithmetic mean in Milan was an exception, being as high as 30.3 µg/m
3
due
to few extremely high data points. Indoor concentrations were at the same level or higher in Helsinki and Prague, but
less than one half in Athens compared to personal exposures. Probably occupational emissions caused this difference in
Athens, because concentrations in workplaces were clearly higher, 7.1 µg/m
3
, than residential indoor concentrations
(Annex 3, Jantunen et al 1999).
In Northern America styrene concentrations in outdoor air have been generally <1 µg/m
3
. In an Canadian study, carried
out in 1988–1990, the mean concentrations at the ambient 18 sites ranged from 0.09 to 2.35 µg/m
3
. In indoor air the
mean concentrations were slightly higher (<1–6 µg/m
3
). Smoking has been found a significant contributor to indoor
concentrations of styrene. The styrene content of cigarette smoke has been reported to be 18–48 µg/cigarette (WHO
1983, WHO 2003). Nazaroff and Singer (2002) assessed average daily residential exposures to styrene in ETS for US
non-smokers who live with smokers. Using exposure–relevant emission factors and assuming the mean consumption of
20 cigarettes smoked per day, they assessed a concentration of 0.6 µg/m
3
being caused by ETS.
Lee and Wang (2004) studied styrene emissions from incense burning in the chamber (18 m
3
) tests. They found
maximum styrene concentrations peaking up to 13 µg/m
3
during incense burning.
Cumulative distributions of the indoor concentrations of styrene in some European cities are presented in Figure 3.1 and
distributions of 48-hour personal exposures in Figure 3.2.
0
20
40
60
80
100
0 5 10 15 20
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
Hel
Mil
Oxf
Pra
Figure 3.1. Cumulative frequency distributions of 30-hour indoor air concentrations of styrene in Athens (Ath, n=42),
Basel (Bas, n=47), Helsinki (Hel, n=188), Milan (Mil, n=40) Oxford (Oxf, n=40) and Prague (Pra, n=46) (EXPOLIS
2002).
The INDEX project Final report
197
0
20
40
60
80
100
01020
Exposure (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
GeS
Hel
Oxf
Pra
Figure 3.2. Cumulative frequency distributions of 48-hour personal exposure concentrations of styrene in Athens (Ath),
Basel (Bas), Helsinki (Hel), Oxford (Oxf) and Prague (Pra) (EXPOLIS 2002), and 1-week mean concentrations of the
German Survey GerES II (Hoffman et al 2000).
4. Toxicokinetics
Absorption
In animal and human controlled studies, the uptake of styrene has been found to be rapid. The principal routes of
exposure are pulmonary and, to a lesser extent, dermal. In several studies with workers and volunteers, the pulmonary
retention of styrene was 60–70% of the inhaled dose. Styrene in ambient air is absorbed through the skin at 2–5 % of
the dose absorbed in the respiratory tract. (IARC, 1994).
Distribution
Styrene is widely distributed throughout the body. The distribution and sequestration of styrene to fat tissue and its
subsequent slow elimination indicate a potential for accumulation in situations of repeated daily exposure. However, in
a study of workers exposed to 160 mg/m
3
(37 ppm; 8-hour TWA) styrene, no evidence of accumulation was found in
monitoring samples during a working week (Pekari et al., 1993).
Metabolism and elimination
Styrene is oxidized to styrene-7,8-oxide by the cytochrome P-450-mediated monooxygenase system. The responsible
isozymes are the ethanol-inducible CYP2E1, CYP2B6 and CYP1A2, and additional isozymes with lower activity for
styrene may be involved (Nakajima et al., 1994). Styrene can also undergo oxidation by other mechanisms and it can be
cooxidized to styrene-7,8-oxide during the lipoxynase-mediated formation of arachidonic acid peroxides (Belvedere, G.
et al., 1983). In vitro, styrene oxidation to styrene-7,8-oxide has been shown to be mediated by oxyhaemoglobin and
myoglobin (Tursi et al., 1983; Ortiz de Montellano and Catalano, 1985).
Enzymatic hydrolysis of styrene-7,8-oxide yields phenylethylene glycol which is further oxidized to mandelic acid and
phenylglyoxylic acid, the principal urinary metabolites of styrene in humans (IARC, 1994). Conjugation of styrene-7,8-
oxide with glutathione leads to urinary excretion of thioethers or mercapturic acid, quantitatively a minor metabolite in
humans. In a field study carried out on workers exposed to 43–311 mg/m
3
of styrene, styrene-7,8-oxide concentration in
blood was found to be linearly correlated with ambient styrene (Korn et al. 1994).
The INDEX project Final report
198
The elimination of styrene is linear at lower concentrations of atmospheric styrene. At concentrations of less than 852
mg/m
3
there is a fast elimination phase with a half-life of 0.5–0.7 hours, and a second phase of slow elimination with a
half-life of 13 hours (IARC, 1994). Several physiologically-based pharmacokinetic models have been developed
(33,34) which simulate the behaviour of styrene and styrene-7,8-oxide in the body. Partially saturated metabolism can
be predicted by the models at high exposures exceeding 1280 mg/m
3
(300 ppm).
Urinary excretion of mandelic acid and phenylglyoxylic acid is biphasic. After an 8-hour exposure of workers to styrene
at concentrations of 26–130 mg/m
3
(6.1–30.5 ppm), both metabolites have a half-life of about 2.5 hour for the first
phase and 30 hours for the second (IARC, 1994). These compounds are used in assessing occupational exposure to
styrene. According to several studies, an exposure to 80 mg/m
3
(20 ppm) styrene corresponds, the following morning, to
a combined mandelic acid and phenylglyoxylic acid level of about 2.9 mmol/litre (Pekari et al., 1993). Coexposure to
ethanol inhibits the metabolism of styrene, resulting in a lag in the excretion of mandelic acid (Wilson et al. 1983).
5. Health effects
Effects of short-term exposure
Styrene may irritate the eyes and mucous membranes and may be toxic to the central nervous system (IARC, 1979).
Immediate eye and throat irritation, increased nasal mucus secretion, listlessness, impairment of balance, and
drowsiness followed by unsteadiness, muscle weakness, and depression were reported in a study of 2 human volunteers
exposed to 3408 mg/m
3
(800 ppm) styrene for 4 hours (Carpenter et al., 1944). Other symptoms include a feeling of
being “lightheaded” or “drunk” (Lorimer et al., 1976).
In an exposure chamber study, volunteer subjects complained of an objectionably strong odor when exposed to 852-
1704 mg/m
3
(200-400 ppm) styrene; exposure to 256 mg/m³ (60 ppm) resulted in detectable odor but no irritation (Wolf
et al., 1956). The duration of exposure and number of subjects were not specified. Investigators at a fiberglass plant
could not withstand more than 1-2 minute exposure to concentrations of 2130-3410 mg/m
3
(500-800 ppm) styrene
(Götell et al., 1972). However, workers exposed to this concentration of styrene for hours complained of only mild to
moderate complaints of irritation, suggesting that tolerance may have developed.
Stewart et al. (1968) found eye and throat irritation in 3 out of 6 volunteers exposed to 422 mg/m
3
(99 ppm) styrene for
20 minutes. No symptoms were reported in 3 subjects after exposure to 217 mg/m
3
for 1 hour. Exposure of these
subjects to 1579 mg/m
3
styrene for 25 minutes resulted in nausea, significant discomfort, and an abnormal Romberg
test, indicative of cerebellar dysfunction. Significant decrements were noted in 3/5 subjects in other tests of
coordination and manual dexterity at 50 minutes. Exposure to 920 mg/m
3
or less for up to 1-hour did not cause
measurable impairment of coordination and balance.
The neurotoxic effects mediated by styrene consist of slowing in sensory, but not motor, nerve conduction velocity and
CNS depression (Cherry and Gautrin, 1990). Reaction time was significantly impaired in 12 males exposed to 1470
mg/m
3
(350 ppm) styrene for 30 minutes, whereas no significant impairment was observed at 1065 mg/m
3
(250 ppm) or
lower (Gamberale and Hultengren, 1974). In this study, no effects on perceptual speed or manual dexterity were
detected. In another study of 12 workers exposed during the workday to 111 mg/m³ (26 ppm), Edling and Ekberg
(1985) measured reaction time before and after work and found no significant differences. Other non-CNS symptoms
were reported in a neuropsychiatric questionnaire completed by the subjects.
The CNS depressant effects of acute exposures to high styrene levels are probably mediated by the direct effect of the
lipophilic, unmetabolized styrene on nerve cell membranes.
Abnormal electroencephalograms were correlated with urinary levels of the styrene metabolite, mandelic acid, of 700
mg/l or higher in workers exposed to styrene (Harkonen et al., 1978).
Consumption of ethanol has been shown to decrease formation of the metabolites mandelic and phenylglyoxylic acid in
human volunteers exposed to 426 mg/m³ (100 ppm) styrene for 8 hours (Cerny et al., 1990). Lowered levels of these
metabolites have been associated with a reduced risk of CNS disturbances in volunteer workers (Cherry and Gautrin,
1990). Co-exposure to inhaled acetone was reported to alter cytochrome-P450 enzymes as measured by altered urinary
steroid metabolites and glucaric acid in workers who consumed moderate amounts of alcohol (Dolara et al., 1983).
However, the clinical significance of the presence of these compounds in the urine is unknown.
Styrene is bioactivated to styrene 7,8-oxide, a reactive metabolite which binds to tissue proteins, acts as a hapten, and
elicits contact allergy in some individuals (Sjoborg et al., 1984). Analyses of styrene oxide adducts bound to human
serum albumin have been used as biomarkers for exposure to styrene (Rappaport et al., 1993). In a study comparing 9
The INDEX project Final report
199
styrene-exposed workers with 24 healthy controls, hematocrit, blood lead levels, and delta-aminolevulinate dehydrase
(ALA-D) levels were measured (Fujita et al., 1987). The workers were exposed to at least 213 mg/m³ (50 ppm) styrene
for 7 days. Styrene oxide was shown to inhibit the formation of ALA-D, an important enzyme in heme biosynthesis, in
these workers. Styrene oxide is also known to bind covalently to DNA in vitro (Hemminki and Hesso, 1984).
Two subjects with occupational asthma due to prior exposure to styrene were exposed to 64 mg/m
3
(15 ppm) styrene in
a chamber (Moscato et al. 1987). Immediate bronchoconstriction was observed in both subjects while a late rash was
also observed in one of the subjects.
Predisposing Conditions for Styrene Toxicity
Medical: Asthmatics may be more sensitive to adverse pulmonary effects from styrene exposure (Moscato et al., 1987).
Chemical: Ethanol consumption and acetone inhalation may inhibit the metabolism and clearance of styrene (Cerny et
al., 1990; Dolara et al., 1983; Elovaara et al., 1990).
Summary of short-term exposure effect levels
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
217
EXP
422
EXP
Eye and throat irritation
Volunteers,
1h (NOAEL),
20m (LOAEL)
Stewart et al., 1968 OEHHA (1999);
REL: 21
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
OEHHA (1999)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Derivation of the Acute Reference Exposure Level (protective against mild adverse effects; for a 1-hour exposure)
Study Stewart et al., 1968
Study population three human volunteers
Exposure method inhalation
Critical effects eye and throat irritation
LOAEL 99 ppm (for 20 minutes)
NOAEL 51 ppm
Exposure duration 1 hour
Extrapolated 1 hour concentration 51 ppm
LOAEL uncertainty factor 1
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 10
Cumulative uncertainty factor 10
Reference Exposure Level 5.1 ppm (21 mg/m³; 21,000 µg/m³)
ATSDR
Agency for Toxic Substances and Disease Registry
Derivation of the acute Minimal Risk Level (MRL).
The possibility for brief human exposure to high concentrations of styrene exists in occupational settings, and might
also exist near major spills. Exposure of the general public to episodic high concentrations of styrene at hazardous waste
sites, in the home, or in the general environment is unlikely.
The respiratory tract and central nervous system are the likely target organ systems for inhaled styrene (Alarie 1973;
Carpenter et al. 1944; DeCeaurriz et al. 1983; Kankaanpaa et al. 1980; Murray et al. 1978; Spencer et al. 1942; Stewart
et al. 1968). The data are not considered sufficient to establish an inhalation acute-duration MRL. Furthermore, ATSDR
acknowledges additional uncertainties inherent in the application of the procedures to derive less than lifetime MRLs.
As an example, acute inhalation MRLs may not be protective for health effects that are delayed in development or are
acquired following repeated acute insults, such as hypersensitivity reactions, asthma, or chronic bronchitis. As these
kinds of health effects data become available and methods to assess levels of significant human exposure improve, these
MRLs will be revised. However, the potential carcinogenicity of styrene prevents the design of controlled laboratory
exposures in humans.
The INDEX project Final report
200
Effects of long-term exposure (noncancer)
Chronic exposures to styrene result in central nervous system (CNS) and peripheral nervous system effects, although
the latter are not as pronounced (ATSDR, 1992; Rebert and Hall, 1994; Murata et al., 1991). Irritation or discomfort of
the upper respiratory tract resulting from styrene exposure has not been reported in long-term occupational studies
(Foureman, 1994). However, sensory irritation and neurological impairment does occur in acute human studies at
concentrations above 426 mg/m
3
(Stewart et al., 1968). The evidence for styrene induced hepatic changes is either
negative or equivocal (ATSDR, 1992). Evidence for nephrotoxicity due to long-term occupational exposure is also
negative or equivocal (ATSDR, 1992; Verplanke and Herber, 1998; Kolstad et al.,1995). Some human studies suggest
that chronic exposure to styrene results in reproductive effects, but the limited data are difficult to interpret because of
the small sample numbers (Brown, 1991; Lindbohm, 1993). Immunologic alterations (e.g., altered phenotypic profiles
among lymphocyte subsets, decreased natural killer cell activity, and decreased chemotaxis) have also been observed,
but the limited data prevent quantitative interpretation (Bergamaschi et al., 1995; Governa et al., 1994).
The CNS depressant effects of acute exposures to high styrene levels are probably mediated by the direct effect of the
lipophilic, unmetabolized styrene on nerve cell membranes. Long-term effects of styrene exposure may result from the
action of one or more metabolites of styrene (Savolainen, 1977; Mutti et al., 1988).
One postulated mechanism for the chronic non-cancer toxicity of styrene is the binding of the highly reactive styrene
oxide to components of nervous tissue. Another postulated mechanism is an alteration in the levels of circulating
catecholamines (e.g., dopamine) due to the binding of PGA to these biogenic amines (Mutti, 1993; Mutti et al., 1984a;
Checkoway, 1994) and the subsequent changes in physiological functions that are under biogenic amine control.
Although long-term exposures to styrene are associated with decrements in physiological functions, the exact
mechanism(s) for these effects have not been clearly established (see reviews by ATSDR, 1992; Mutti, 1993; Rebert
and Hall, 1994).
Kolstad et al. (1995) estimated excess deaths due to four major non-malignant disease groups for 53847 male workers
in the Danish RPI. Low and high styrene exposures were based on companies with less than 50% (low) and those with
50% or more (high) employees involved with reinforced plastics. An internal comparison was made with workers
unexposed to styrene to account for more similar activities and lifestyles. Statistically significant (p < 0.05) excess
deaths due to pancreatitis and degenerative disorders of the myocardium and non-significant excess deaths due to
degenerative diseases of the nervous system were observed. Non-significant excess deaths due to glomerulonephritis
were also observed.
Checkoway et al. (1994) described a cross-sectional study of 59 male boat plant workers exposed to <4,3 to 613 mg/m
3
mean = 158 mg/m
3
styrene. Monoamine oxidase B (MAO-B) activity in platelets was measured as an indicator of
catecholamine metabolism. When the styrene exposed workers were divided into quartile exposures, a dose dependent
decrease in MAO-B activity was observed after adjustments were made for age, smoking, alcohol and medication use.
Female workers employed in the reinforced plastics industry (RPI) were studied for levels of substances associated with
neuroendocrine function (Mutti et al., 1984a). Serum prolactin, thyroid stimulating hormone, human growth hormone,
follicle stimulating hormone, and luteinizing hormone were measured in 30 women who were between the 5th and 15th
day of the menstrual cycle. Exposure was based on the nextmorning MA+PGA, and levels of the neuroendocrine
substances were measured in venous blood samples taken the next morning before the start of work. On the basis of a
relationship (not detailed in the report) between urinary metabolites and styrene air concentration, the authors estimated
that the average styrene TWA/8 hr was about 554 mg/m
3
(130 ppm). Controls consisted of women factory workers
living in the same area as the styrene-exposed women, but not knowingly exposed to styrene. After controlling for age
and exposure time, the increased prolactin and thyroid stimulating hormone levels were correlated with the
concentration of next-morning urinary MA+PGA, although only the increased prolactin levels were statistically
significant.
Numerous occupational studies have noted CNS disturbances in styrene-exposed workers. Decreased manual dexterity,
increased reaction times, and/or abnormal vestibuloocular reflex (ability to track moving objects) were observed by
Götell et al. (1972), Gamberale et al. (1975), Lindstrom et al. (1976), Mackay and Kelman (1986), Flodin et al. (1989),
Moller et al. (1990), and Cherry and Gautrin (1990) for air styrene levels of about 51 mg/m
3
(12 ppm) to more than 426
mg/m
3
(100 ppm). However, in each of these studies, there were difficulties in quantifying the effect. The difficulties
included small sample size, unknown exposure duration, lack of concurrent control group, lack of dose-response data,
and either unknown ethanol consumption or lack of adjustment for ethanol consumption. In the Cherry and Gautrin
(1990) investigation, however, the authors determined that accounting for ethanol consumption did not reduce the
correlation between increased reaction time and exposure.
The INDEX project Final report
201
Decrements in other CNS functions were observed among workers in the well-controlled studies of Fallas et al. (1992),
Chia et al. (1994), and Mutti et al. (1984b). Fallas et al. (1992) studied 60 male workers (average age = 29.5 years,
average air styrene = 104 mg/m
3
). The styrene-exposed population was compared to non-exposed worker controls and
matched for age, intellectual level, and ethnic origin. The results from a standardized test battery showed decrements in
the aiming response and 22/60 styrene exposed workers exhibited increased reaction times compared to 7/60 controls.
Acquired color vision loss (dyschromatopsia) was also observed in the styrene-exposed workers compared to controls.
Chia et al. (1994) also observed decrements in CNS function as defined by altered visual retention, audio-digit
recognition, and digit recognition. However, a dose-response relationship did not exist. These workers also exhibited a
statistically nonsignificant dose-dependent dyschromatopsia.
In the most comprehensive occupational study to date on CNS effects of styrene exposure, Mutti et al.(1984b) assessed
memory and sensory/motor function in a group of 50 male styrene-exposed workers (average exposure = 8.6 years) and
a control group of 50 manual workers. In addition to matching for age, sex, and educational level, a vocabulary test was
included to match for general intelligence. Eligibility criteria included absence of metabolic, neurologic, or psychiatric
disorders, limited ethanol intake, and limited cigarette usage. All subjects were instructed to avoid intake of alcohol and
drugs for two days prior to testing. Styrene exposure was assessed from urinary MA+PGA levels the morning after the
last workday in the week, followed immediately by participation in a battery of 8 neuropsychological tests designed to
measure CNS function. The tests included reaction time, short and long term logic memory, short and long term verbal
memory, digit-symbol association (using a reference code), block design (reproducing a displayed design using colored
blocks), and embedded figures (timed identification of figures in Rey’s table). The mean + 2 SDs of the values found in
the control group was set as the normal range limit for each neuropsychological test. The results were expressed as
continuous and quantal data. Expressed as continuous data, styrene-exposed workers exhibited significantly poorer
performances than controls in all tests, except in the digit-symbol test. Also, urinary metabolite concentration and
duration of exposure were found to be significantly correlated with the scores of several tests. As a subgroup, workers
with metabolite levels of up to 150 mmoles MA+PGA/mole creatinine (mean = 75 mmoles/mole creatinine + 33 [SD],
which is equivalent to a mean styrene concentration of 64 mg/m
3
) appeared to have no significant effects. The authors
state that this level of urinary metabolites corresponds to a mean daily 8-hour exposure to air styrene of 107 mg/m
3
(25
ppm). Based on greater urinary excretion of styrene metabolites, significantly poorer performances in four or more
neuropsychological tests were recorded in the other three subgroups (150-299, 300-450, and > 450 mmoles MA +
PGA/mole creatinine).
Mutti et al. (1984b) expressed the quantal data as the fraction of tested subjects who responded abnormally to 1, 2,
and 3 tests (see Table 5.1). Positive dose-response relationships existed between intensity of styrene exposure
(mmoles MA + PGA/mole creatinine) and abnormal scores, whether it was expressed as abnormal responses in at least
one, at least two, or at least three neuropsychological tests. The chi-square test and validity calculations were performed
by constructing 2 x 2 tables selecting different levels of urinary excretion of MA and PGA as a cut-off point. The
highest values for chi-square and predictive validity were found when the cut-off of 150 mmol/mol creatinine was
chosen, suggesting that the quantal isolation of the low dose subgroup from the next subgroup is appropriate.
When the quantal data for the low dose subgroup were analyzed by OEHHA using the Fisher’s Exact Test, a significant
level of abnormal responses were observed for 1 (p = 0.005) and 3 (p = 0.04) tests. The abnormal responses for 2
tests were statistically marginal (p = 0.06). For each of the remaining exposure groups, the p-values were <0.05. Unlike
the assumptions made concerning the continuous data, quantal data results suggest that the low dose subgroup
represents a LOAEL, and that a NOAEL is not available from the data. Mutti et al. (1984b) also expressed the data in a
quantal three-way representation including prevalence (number of respondents for at least one, two or three abnormal
tests), duration (years at work), and intensity (metabolite level). This representation revealed a positive correlation of
neuropsychological deficits with duration as well as intensity.
Table 5.1: Subjects Classified Positive on Neuropsychological Tests as a Function of Styrene Exposure
a
.
MA+PGA, mmoles per mole
creatinine
b
Total
Subjects
Number of Abnormal Tests
1 2 3
c
Controls 50 4/50 2/50 0/50
< 150 (mean = 75 ± 33)
d
14 6/14 3/14 2/14
150-299 (mean = 216 ± 45) 9 6/9 5/9 3/9
300 - 450 (mean = 367 ± 49) 14 10/14 7/14 5/14
> 450 (mean = 571 ± 108) 13 11/13 8/13 6/13
a
Data from Table IV in Mutti et al. (1984b).
b
“Next-morning” styrene urinary metabolites.
The INDEX project Final report
202
c
The quantal grouping of the number of subjects that performed abnormally in >3 tests based on their styrene urinary metabolite
concentrations, both shown in bold, were used in a benchmark concentration (BMC) analysis for the derivation of the REL (see
Section VI below).
d
Based on Guillemin et al. (1982), a linear relationship exists for converting the urinary metabolite concentrations to ppm air
styrene levels (4.97 mmoles MA+PGA/mole creatinine is equivalent to 1 ppm styrene). Thus, the mean styrene concentrations
per group are 0, 15, 44, 74, and 115 ppm.
In addition to dyschromatopsia observed by Chia et al. (1994), Gobba and Cavalleri (1993) and Campagna et al. (1995)
also reported this visual dysfunction among styrene workers in the reinforced plastics industry. Workers (n=36) exposed
to an average of 16 ppm styrene exhibited significantly greater dyschromatopsia than controls, matched for age, ethanol
consumption and tobacco smoking (Gobba and Cavalleri, 1993). Among the study population, only 1/36 styrene-
exposed workers (compared to 16/36 controls) performed the test with 100 percent accuracy. When a different group of
styrene-exposed workers was tested, those exposed to > 50 ppm styrene exhibited greater dyschromatopsia than those
exposed to 50 ppm, and within this group, a subset exhibited a similar decrement after returning from a one month
vacation. In the Campagna et al. (1995) study, the test for dyschromatopsia was given to 81 reinforced plastics industry
workers (79 male and 2 female) exposed to 4.6, 10.1, and 88.8 ppm styrene (first quartile, median, and third quartile,
respectively). No control group was used in this study. Statistical analysis revealed a correlation of color vision loss
with exposure to styrene (defined as next-morning urinary mandelic acid), age, and ethanol consumption.
Exposure to styrene may affect the peripheral nervous system (PNS). In a case report (Behari et al., 1986), a man
working for 5 years with a photostat process that used styrene was diagnosed with peripheral neuropathy. However, in
occupational studies, the relationship between exposure to styrene and PNS effects has been inconsistent (Triebig et al.,
1985; Cherry and Gautrin, 1990). A major difficulty in understanding the potential for this relationship is the lack of
knowledge about the appropriate surrogate for dose that leads to PNS disturbance (Murata et al., 1991). In one study,
however, chronic exposure indices were developed which included work method, years at work, time spent laminating
(source of high exposure), styrene air concentration, and end-of-shift urinary mandelic acid (Matikainen et al. (1993).
Numbness in the extremities increased with the exposure index, although statistically the effect was marginally
insignificant (p < 0.1). The styrene TWA/8 hr was 136 mg/m
3
for the 100 study subjects.
Female reproductive toxicity has been inconsistently reported among humans (Brown, 1991; Lindbohm, 1993). These
studies are difficult to interpret because of the high background rates of endpoints such as spontaneous abortion and
menstrual disorders in combination with confounding exposures. In those studies that showed no reproductive effects
due to styrene exposure, the power of the studies was low due to the small numbers of women. Hence the evidence for
any adverse effects of exposure to styrene on female reproductive function is inconclusive.
Kishi et al. (1992a, 1992b) reported that Wistar rat offspring exposed to airborne styrene in utero at 256 mg/m
3
(60
ppm) for 6 hour/day during days 7 to 21 of gestation, had significantly reduced pup weights at day 1, delayed
development of righting reflex and auditory startle reflex, and nonsignificantly decreased levels of serotonin and its
metabolites 5-hydroxyindoleacetic acid (5-HT) in the cerebrum. Exposure to 1270 mg/m
3
(293 ppm) significantly
reduced pup body weight and the levels of serotonin and 5-HT, and induced alterations in a wider range of behavioural
measures. Thus, the lowest LOEL for neurotoxic effects in animals following inhalation of styrene in an adequately-
conducted study is 260 mg/m
3
(60 ppm) in Wistar rats exposed to the compound in utero (Kishi et al., 1992a,b).
Male workers employed in the reinforced plastics industry were examined for effects on sperm chromatin structure and
semen quality (Kolstad et al., 1999a) and time to pregnancy (Kolstad et al., 1999b). No indications of an exposure-
response relationship were seen when individual changes in semen quality were related to the postshift urinary mandelic
acid concentrations among 23 exposed workers. A weak increase in sperm DNA-susceptibility to in situ denaturation as
a function of mandelic acid concentration was indicated, but was within the interassay variability. No detrimental effect
of styrene exposure was observed with regard to male fecundity among 188 exposed workers when compared to 353
unexposed workers.
Immune system alterations were reported in a study conducted by Bergamaschi et al. (1995). Reinforced plastics
industry workers (n=32 female/39 male, average age = 32 years, average exposure duration = 7 years) were compared
with non-styrene exposed factory workers and matched for age, sex, tobacco use and ethanol consumption. Air styrene
levels, among the different factories, varied between 43 – 213 mg/m
3
(10-50 ppm), and individual worker exposure was
measured by urinary metabolites the morning after the last shift (15 hours post-exposure). Among all workers in the
study (median exposure = 68 mg/m
3
- according to the data of Guillemin et al. (1982)), the proportion of 12/18
lymphocyte subsets and the prevalence of abnormal values of immunologic phenotypes for 11/18 subsets were
statistically different from the controls (p < 0.001 to < 0.05). When the workers were placed into three exposure groups
(0, < 107 mg/m
3
, and > 107 mg/m
3
styrene), dose-response relationships were observed for prevalences of abnormal
responses for four lymphocyte subsets and, in the case of two subsets, abnormal responses were observed in the group
exposed to < 107 mg/m
3
styrene. Natural killer cell activity (a lymphocyte function), measured in a different group of
The INDEX project Final report
203
workers in the same study, was decreased compared to unexposed worker controls. The median exposure, given in
terms of urinary metabolites, was calculated as 89 mg/m
3
based on the data of Guillemin et al. (1982). The data show
that exposure of these workers to air styrene levels below 213 mg/m
3
, and probably at levels near 107 mg/m
3
, resulted
in alterations of the immune system.
Governa et al. (1994) observed reduced chemotactic responses of polymorphonuclear lymphocytes (PMNs) obtained
from 21 styrene-exposed workers. However, the lack of exposure data prevents a quantitative assessment. In the same
study, 0.1 - 0.6 mM styrene inhibited the chemotaxis of isolated healthy PMNs.
Reproductive and teratogenic effects
The frequency of spontaneous abortions among women with definite or assumed exposure to styrene has been
investigated in a number of studies; the majority do not indicate an increased risk in association with occupational
exposure to styrene (IARC, 1994). A study in Canada (McDonald et al., 1988) found an increased risk for spontaneous
abortions (18 observed; SMR, 158; 90% CI, 102–235) among women employed in polystyrene manufacture. The
expected figures were derived from the experience of 47 316 pregnant women who had worked for 30 hours or more
per week at the start of pregnancy. No styrene concentrations were given, and most of these women had mixed
exposures. Two studies in Finland found no increase in the frequency of spontaneous abortion or congenital
malformation among the wives of men exposed occupationally to styrene (Taskinen et al., 1989; Lindbohm et al.,
1991).
The conclusion thus remains that no clear association has been established between occupational exposure of either
mothers or fathers to styrene and the frequency of spontaneous abortions or congenital malformations.
Summary of long-term exposure effect levels (noncancer)
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
64
Study
17
MLE
7.2
BC05
2.6
ADJ
CNS; neuropsychological tests: reaction time, s/l
term logic memory, s/l term verbal memory,
digit-symbol association, block design and figure
identification.
Occupational, 8.6y Mutti et al. 1984b
OEHHA (1999);
REL: 0.9
94
95%lCL
34
ADJ
>94
95%lCL
same as above Occupational, 8.6y;
Mutti et al. 1984b
EPA-IRIS (1992)
; RfC: 1
107
Study
25
ADJ
same as above Occupational 8.6y; Mutti et al. 1984b ATSDR (1992);
MRL: 0.25;
WHO (2001);
GV: 0.26;
RIVM (2000)
260
EXP
65
ADJ
46
HEC
Neurological developmental; body weight,
biochemical parameters in the brain and
behaviour
Rat offspring Kishi et al.,1992a,b Health Canada
(1993);
TC: 0.092
Final statement (UNIMI) : Human LOAEL: 25 mg/m³
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration ;
95%lCL
lower limit of the 95% confidence interval
The INDEX project Final report
204
OEHHA (1999)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Derivation of the Chronic Reference Exposure Level (BMC Approach) :
Study Mutti et al. (1984b)
Study populations Human (occupational)
Exposure method Inhalation
Critical effects Central nervous system
LOAEL 15 ppm
NOAEL Not established
BMC
05
1.7 ppm
Exposure continuity 8 hr/d (10 m
3
per 20 m
3
day), 5 d/wk
Exposure duration 8.6 years (average years at work)
Average occupational exposure 0.61 ppm (1.7 x 10/20 x 5/7)
Human equivalent concentration 0.61 ppm
LOAEL uncertainty factor Not needed in the BMC approach
Subchronic uncertainty factor 1 (average exposure 12.3% of lifetime)
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 3
Cumulative uncertainty factor 3
Inhalation reference exposure level 0.2 ppm (200 ppb; 0.9 mg/m
3; 900 µg/m
3
)
The most relevant chronic noncancer effect due to styrene exposure is neurotoxicity. The Mutti et al. (1984b)
occupational study presented convincing dose-response information and was well designed and executed in terms of
experimental protocol and statistical evaluation, which included tests for false positive and false negative responses.
While not all confounders could be ruled out (e.g., compensatory mechanisms, biorhythms, workers who leave because
of styrene related illness), careful attention was paid to include eligibility criteria for the control group that correct for
confounders unique for this population (e.g., limited ethanol intake, a control work-force not exposed to neurotoxic
substances, and a test to allow a match for general intelligence). The use of urinary metabolites to measure exposure
dose is based on the observation that the next-morning urinary mandelic acid (MA) and phenylglyoxylic acid (PGA) is
directly related to the air level of styrene. The Guillemin et al. (1982) study provides the basis for the conversion of
urinary MA+PGA levels to styrene exposure levels used by Mutti et al. (1984b). The quantal dose-response data by
Mutti et al. (1984b) is applicable for use in a benchmark concentration (BMC) approach. The quantal grouping of the
number of subjects that performed abnormally in >3 tests based on their urinary metabolite concentrations was chosen
for a BMC analysis. Basing the BMC on abnormal responses to >3 tests reduces the complexity of multiple test
comparisons and the potential for inappropriate comparison of different neuropsychological tests between control and
exposure groups for statistical purposes. Also, the potential for false positive responses is reduced due to the zero
background level of abnormal responses in the control group when the criteria are >3 abnormal tests. Using a log-
normal probit analysis (Tox-Risk, version 3.5; ICF-Kaiser Inc., Ruston, LA) the maximum likelihood estimate (MLE)
for a 5% response was 17 mg/m
3
(4.0 ppm). The resulting 95% lower confidence limit at the MLE provided a BMC
05
of
7.2 mg/m
3
(1.7 ppm). A BMC
05
is considered to be similar to a NOAEL in estimating a concentration associated with a
low level of risk. Following adjustment for exposure continuity (10 m
3
per 20 m
3
day for 5 d/wk) and application of an
UF of 3 to account for human intraspecies variability, a REL of 0.9 mg/m
3
(0.2 ppm) was attained. For exposure data
that utilizes healthy human subjects, the resulting BMC represents a less than 10% incidence in the general population.
When combined with an UF of 3, as carried out above, the resulting REL will be protective of the vast majority of
individuals. This analysis of the quantal data is supported by recognizing that, in a population of 50 subjects, individual
test-specific effects that occur at low doses may not have been observed. If the criterion for abnormality is expressed in
terms of CNS dysfunction, defined by all tests, the sensitivity of the testing procedure is increased and the low dose
effects are more easily observed. The quantal data of Mutti et al. (1984b), i.e., the proportion of subjects responding
abnormally to the tests, therefore provide a more sensitive approach to detecting low dose eff ects. Collasping a battery
of test data to increase sensitivity may introduce the dilemma of multiple test comparisons, as noted above. However,
OEHHA believes that a statistical method to correct for this, known as a Bonferroni correction, is unnecessary. The
REL development is based on calculating a statistic of one effect of a complex of responses (or a syndrome) that results
from CNS dysfunction, and not based on calculating a statistic for each test within the group of tests. The apparent
global nature of the neurological syndrome resulting from long-term styrene exposure, in addition to basing the BMC
on abnormal responses to >3 tests, should more than adequately address any concerns that may result from combining
neurological test data. Applying NOAEL/LOAEL methodology to the Mutti et al. (1984b) quantal data yields an
exposure value similar to that attained with the BMC approach. The LOAEL of 64 mg/m
3
(15 ppm) is adjusted to an
equivalent continuous exposure of 23 mg/m
3
(64 mg/m
3
x 10/20 m3 x 5/7 d/wk). Use of a LOAEL UF of 3 and an
intraspecies UF of 10 resulted in an estimated REL of 0.8 mg/m
3
(0.2 ppm).
The INDEX project Final report
205
The U.S. EPA (1996) calculated a reference concentration (RfC) of 1 mg/m
3
(0.3 ppm), which is slightly higher than the
OEHHA-derived chronic REL of 0.9 mg/m
3
(0.2 ppm). The RfC for styrene is also based on the findings of Mutti et al.
(1984b), but utilized the continuous data for development of the RfC and used standard NOAEL methodology for the
RfC derivation. U.S. EPA (1996) established a NOAEL for the lowest exposure group (<150 MA+PGA mmole/mole
creatinine; equivalent to < 106 mg/m
3
styrene).
However, OEHHA staff believe that the use of the continuous data to establish a NOAEL overlooks the advantages of
using the BMC approach using the quantal data. These advantages are that the BMC
05 reflects the shape of the dose-
response curve and takes into account the number of subjects involved in the study. In addition, OEHHA staff evaluated
the quantal data with the Fisher’s Exact Test and determined the probabilities of abnormal responses among the exposed
subjects based on the unexposed subjects whose responses were assumed to be normal. At the lowest exposure, the
probability that the proportion of subjects responding abnormally to >1 and >3 tests was within the expected range was
p = 0.005 and p = 0.04, respectively, indicating that neuropsychological deficits due to styrene occur in the low dose
subgroup. Thus, the quantal data indicate that a NOAEL was not established in this study.
U.S.EPA - IRIS (1992)
Integrated Risk Information System
Determination of the Reference Concentration for Chronic Inhalation Exposure (RfC).
Critical effect Exposures* UF MF RfC
CNS effects
Occupational Study
Mutti et al. (1984b)
NOAEL: 94 mg/cu.m (25 ppm = 150 mmole
urinary styrene metabolites/mole creatinine
adjusted to lower 95%
confidence limit = 22 ppm)
NOAEL(HEC): 34 mg/cu.m
LOAEL: >94 mg/cu.m (>22 ppm
derived as in NOAEL listing)
30 1 1 mg/m
3
*Conversion Factors: MW = 104.15. Assuming 25 C and 760 mmHg, NOAEL (mg/cu.m) = NOAEL (ppm) x MW/24.45 = 94 mg/cu.m. The NOAEL
exposure level is based on a back extrapolation from worker urinary concentration of styrene metabolites reported in the principal study and adjusted
to the lower 95% confidence limit listed in Guillemin et al. (1982), which was 88%, 25 ppm x 0.88 = 22 ppm. The NOAEL(HEC) is calculated using
an 8-hour TWA occupational exposure. MVho = 10 cu.m/day, MVh = 20 cu.m/day. NOAEL(HEC) = 94 mg/cu.m x MVho/MVh x 5 days/7 days =
34 mg/cu.m. The feasibility of applying the exposure model of Perbellini et al. (1988) for extrapolation of the values in the principal study is currently
being investigated. Application of this model may result in changes in the NOAEL(HEC) value and, therefore, the RfC.
The concentration-response relationship between urinary metabolite concentration (mandelic acid and phenylglyoxylic
acid levels normalized to creatinine in "morning-after" urine) and test results indicated a significant effect level in the
subgroup whose urine contained 150-299 mmole urinary metabolites/mole creatinine. Workers with metabolite
concentrations of up to 150 mmoles/mole appeared to have no significant effects, and this level is therefore designated
as the NOAEL in this study. The authors state that this level of urinary metabolites corresponds to a mean daily 8-hour
exposure to air styrene of 107 mg/m
3
(25 ppm). Derivation of this air level is from the creatinine-normalized, combined
concentration of the styrene metabolites, MA and PGA, in urine collected from the workers on Saturday mornings.
Guillemin et al. (1982) demonstrated a logarithmic relationship (r = 0.871) between the summation of urinary
metabolites (MA + PGA, next morning) and air concentrations of styrene (ppm x hours). Guillemin calculated the mean
combined urinary metabolite concentration (next morning) for an 8-hour exposure to 426 mg/m
3
. This relationship was
used by both Mutti et al. (1984b) and Guillemin and Berode (1988) in a proportional manner to obtain styrene air levels
at lower urinary metabolite concentrations. The 95% confidence interval was also calculated for an 8-hour exposure at
426 mg/m
3
, the lower limit of the confidence calculation being 88% of the mean styrene exposure. This factor was
applied directly to the NOAEL of 107 mg/m
3
[107 mg/m
3
x 0.88 = 94 mg/m
3
]. Due to the construction of the
subgroups, designation of a LOAEL was the lower limit of the subgroup in which adverse effects were observed [i.e.,
greater than the NOAEL of 94 mg/m
3
].
Confidence in the Inhalation RfC: The study of Mutti et al. (1984b) documents concentration-response relationships of
CNS effects in a relatively small worker population. However, the results of this study are consistent with a number of
other studies showing central effects in chronically exposed worker populations, most notably that of Moller et al.
(1990). The urinary metabolites, MA and PGA, are direct biological indicators of exposure to styrene. Numerous
studies have demonstrated the relationship between urinary metabolites and air levels of styrene to be reliable and
quantitative. Physiologically based pharmacological modeling of this exposure methodology demonstrates that it
reflects and incorporates at least a portion of intrahuman variability related to pharmacokinetics. The study is therefore
assigned a medium confidence level. The data base can be considered medium to high as chronic laboratory animal
studies addressing noncancer endpoints are not yet available, but a number of human exposure studies support the
choice of critical effect. Preliminary information in mice indicate that styrene is a respiratory tract irritant in mice at
concentrations lower than 47.5 mg/cu.m. The RfC is assigned an overall confidence rating of medium.
The INDEX project Final report
206
ATSDR (1992)
Agency for Toxic Substances and Disease Registry
Derivation of the acute Minimal Risk Level (MRL):
Studies in workers exposed to styrene in workplace air suggest that neurological effects are probably the most sensitive
indicator of styrene toxicity. Available data do not.provide a clear picture of the NOAEL for neurological effects
following acute- or intermediate-duration inhalation exposure, but two chronic studies in workers identify LOAELs of
107 mg/m
3
(25 ppm) (Mutti et al. 1984a) and 132 mg/m
3
(31 ppm) (Harkonen et al. 1978). Based on the lower LOAEL
(25 ppm), a chronic inhalation MRL of 0.25 mg/m
3
(0.06 ppm) has been derived using factors of 8/24 and 5/7 to
account for exposure 8 hours/day, 5 days/week, and an uncertainty factor of 100 (10 to account for human variability,
and 10 to account for use of a LOAEL).
WHO (2001)
Air Quality Guidelines for Europe, 2000
Although genotoxic effects in humans have been observed at relatively low concentrations, they were not considered as
critical endpoints for development of a guideline, in view of the equivocal evidence for the carcinogenicity of styrene.
In occupationally exposed populations, subtle effects such as reductions in visuomotor accuracy and verbal learning
skills, and subclinical effects on colour vision have been observed at concentrations as low as 107–213 mg/m
3
(25–50
ppm). Taking the lower number of this range for precautionary reasons and adjusting this value in order to allow for
conversion from an occupational to a continuous pattern of exposure (a factor of 4.2), and incorporating a factor of 10
for interindividual variation and 10 for use of a lowest-observed-adverse-effect level (LOAEL) rather than a no-
observed-adverse-effect level (NOAEL) this results in a guideline of 0.25 mg/m
3
(weekly average). This value should
also be protective for the developmental neurological effects observed in animal species.
The air quality guideline could also be based on the odour threshold. In that case, the peak concentration of styrene in
air should be kept below the odour detection threshold level of 70 µg/m
3
as a 30-minute average.
Health Canada
Canadian Environmental Protection Act (CEPA).
Derivation of a tolerable concentration (TC):
The available data on nonneoplastic effects (principally neurological) in humans are considered to be inadequate to
serve as the basis for the development of a tolerable concentration (TC), due to such factors as the lack of precise
exposure data, simultaneous exposures to other chemicals, the short duration of available clinical studies, and the small
numbers of subjects in many studies. It should be noted, however, that a TC derived on the basis of neurotoxic effects in
cross-sectional epidemiological studies and clinical studies in human volunteers would not vary greatly from that
derived below on the basis of studies in animal species.
The lowest reported levels inducing meaningful effects in experimental animals exposed to styrene by inhalation fall
within a similar range; indeed, there is no clearly superior critical study and TCs derived on the basis of the lowest
effect levels from several studies would be similar. For this assessment, the study by Kishi et al. (1992a,b) has been
selected for the derivation of a TC since it leads to the most conservative value and since observed effects included both
body weight changes and manifestations of neurotoxicity (including biochemical and behavioral effects).
On the basis of the 260 mg/m
3
(60 ppm) LOEL of the Kishi et al. (1992a,b) studies, a TC of 0.092 mg/cu.m was derived
for inhalation exposure. A factor of 6/24 was used to convert from intermittent to continuous exposure. The ratio of
inhalation volume to body weight of rats [(0.11 m
3
/day)/0.35 kg] to humans aged 5 to 11 [(12 m
3
/day)/27 kg] was
applied. An uncertainty factor of 500 (10 for interspecies variation, and 10 for intraspecies variation and 5 for use of a
LOEL as the effects observed at this concentration were not clearly adverse).
Limited available data indicate that humans form less of the putative toxic metabolite, styrene-7,8-oxide, and hydrolyze
it more quickly than experimental animals; however, available data were insufficient to take these differences into
account in the development of the uncertainty factor, and the relevance of this metabolite to developmental and
neurotoxic effects is not clear.
This tolerable concentration is within the range of that which would be derived on the basis of neurological effects in
occupationally exposed populations.
The INDEX project Final report
207
Macromolecular adducts and genetic toxicity
The genetic toxicology of human styrene exposure and styrene macromolecular binding have been reviewed by several
authors (IARC, 1994; Phillips and Farmer, 1994; Barale 1991; Norppa and Sorsa, 1993) and have mostly been studied
in the reinforced plastics industry where styrene is the main chemical exposure. Only a few studies are available on
other branches of industry, such as styrene production, where exposure to styrene is low. Exposure levels in the studies,
evaluated on the basis of urinary styrene metabolites or workplace air samples, have ranged widely.
The levels of the N-terminal valine adduct of haemoglobin, N-(1-hydroxy-2-phenylethyl)valine, have been found to be
four times higher in styrene-exposed workers than in controls. In a Swedish-Finnish study (Christakopoulos et al.,
1993), increased levels of adducts to N-terminal valine in haemoglobin were seen in reinforced plastics workers (mean
28 pmol × g
-1
globin) in relation to control persons (mean <13 pmol × g
-1
globin). This level was low, while a
background adduct level was also seen among the controls. A significant correlation of individual adduct levels and free
styrene-7,8-oxide and styrene glycol in blood, as well as mandelic acid in urine, was seen in a regression analysis.
Extrapolation from the metabolite monitoring data gave an estimate of about 320 mg/m
3
(75 ppm) for the workplace air
styrene concentration. In another study, using similar techniques, but at lower exposure levels (30 mg/m
3
,
7 ppm), no
styrene-7,8-oxide adducts in N-terminal valine of haemoglobin were seen (Severi et al., 1994).
The first DNA adducts studies on humans exposed to styrene were published by Liu et al. (1988) who detected styrene-
7,8-oxide modified guanine adducts in a single exposed person but none in a single unexposed individual.
Extensive studies on biomonitoring of styrene exposure using DNA adducts have been performed (Canadian
Environmental Protection Act, 1993; Vodicka and Hemminki, 1993). Lamination workers from work sites representing
high exposures (mean air styrene level 370 mg/m
3
,
87 ppm) and moderate exposures (mean air styrene level 210 mg/m
3
,
49 ppm) were studied. Using synthetic standards and the
32
P-postlabelling technique, O
6
-(2-hydroxy-1-phenylethyl)-2'-
deoxy-guanosine-3'-monophosphate (O
6
-dGMP) adducts were detected among the exposed workers, at 7–16 times
higher level than in the controls.
In a further study, where DNA adducts were investigated before and after a 2-week vacation, no decline in adduct levels
was seen, indicating a slow removal of the specific O
6
-guanine adducts of styrene from DNA (Vodicka et al., 1994).
Horvath et al. (1994) studied DNA adducts by postlabelling in mononuclear blood cells of lamination workers and
found two types of adducts, one identified as guanine N
2
adduct, to be increased with a significant linear relationship to
individual measurements of airborne styrene, but not to SCE frequencies among the same workers. DNA adduct levels
were highly elevated among the styrene exposed workers, with a correlation to DNA strand breakage measured by the
comet assay, but not to mutant frequencies (hypoxanthine-guanine phosphoribosyl transferase (HPRT) locus) (Vodicka
et al., 1995). These results suggest that no simple quantitative relationships exist between the adducts and other
parameters of DNA damage in human lymphocytes. In this study, control subjects from the administration of the same
plastics factory also showed elevated adduct levels in comparison with unexposed control persons.
In studies on cytogenetic parameters – chromosomal aberrations, SCEs, and micronuclei – in peripheral lymphocytes of
workers employed in the reinforced plastics industry (Sorsa et al., 1992; Mäki-Paakkanen et al., 1991; Brenner at al.,
1991; Meretoja et al., 1977; Meretoja et al., 1978; Fleig and Thiess, 1978; Thiess and Fleig, 1978; Högstedt et al., 1979;
Thiess et al., 1980; Andersson et al., 1980; Watanabe et al., 1981; Watanabe et al., 1983; Högstedt et al., 1983; Camurri
et al., 1983; Camurri et al., 1984; Hansteen et al., 1983; Nordenson and Beckman, 1984; Pohlova and Sram, 1985;
Mäki-Paakkanen, 1987; Forni et al., 1988; Jablonicka et al., 1988; de Jong et al., 1988; Hagmar et al., 1989; Kelsey et
al., 1990; Yager et al., 1990; Yager et al., 1993; Perera et al., 1992; Tomanin et al., 1992; Hallier et al., 1994; Van
Hummelen et al., 1994; Artuso et al., 1995; Anwar and Shamy, 1995; Norppa et al., 1981), positive findings have
primarily concerned chromosomal aberrations, which have been observed to be increased in the majority of the studies.
SCEs and micronuclei were also found to be increased in some studies.
On the basis of rough exposure estimates and observed frequencies of chromosomal aberrations, the NOAEL for long-
term styrene exposure has been proposed to be 85–128 mg/m
3
(20–30 ppm) (Knudsen and Sorsa, 1993) or 213 mg/m
3
(50 ppm) (Barale, 1991).
A recent meta-analysis on cytogenetic data from 25 reports on occupational styrene exposure (Bonassi et al., 1996)
showed a significant increase in weighed frequency ratio for chromosomal aberrations from studies with median styrene
exposure above the chosen dichotomization point, 125 mg/m
3
(30 ppm); results for SCEs and micronuclei were
inconclusive. Fleig and Thiess (1978) observed an increase in chromosomal aberrations in a worker group with high
exposure to styrene (mean 486 mg/m
3
, 114 ppm) but not in two other groups with lower exposure levels (2 and 8
mg/m
3
). Anderson et al. (1980) observed an increase in the frequency of chromosomal aberrations with increasing total
styrene exposure (expressed as the average concentration in mg/m
3
multiplied by the number of years of employment)
The INDEX project Final report
208
in a low-dose group (mean total styrene exposure 137 mg/m
3
) but not in a high-dose group (mean 1204 mg/m
3
). Forni et
al. (1988) found an increase of chromosomal aberrations in workers from a factory where current exposure to styrene
was high (41–198 mg/m
3
) but not in workers from another factory with low current exposure (1.7–17 mg/m
3
); the latter
group of workers – who had experienced a high exposure to styrene in the past and had, therefore, a high cumulative
exposure – showed an increase in chromosomal aberrations. Mäki-Paakkanen et al. (1991) obtained a significant
correlation between the number of cells with chromosomal aberrations and years of exposure. Tomanin et al. (1992)
could demonstrate an elevation of chromosomal aberrations in a high-exposure group (115–443 mg/m
3
) but not in a
low-exposure group (21–102 mg/m
3
). Artuso et al. (1995) obtained a significant styrene-exposure dependent trend for
both chromosomal aberrations and SCEs among a high-dose group (gel coaters, laminators, rollers and assemblers;
exposure range 85–1389 mg/m
3
), a low-dose group (other tasks; exposure range 2–119 mg/m
3
), and controls.
Camurri et al. (1984) observed a significant increase in SCEs in workers in reinforced plastics plants with mean
airborne styrene levels of more than 200 mg/m
3
but not in plants with mean levels of 30–200 mg/m
3
. Yager et al. (1993)
obtained a clear relationship among a group of boat manufacturers between SCE frequency and styrene exposure
measured either as the concentration of styrene in workroom air (0.9– 234 mg/m
3
; mean 64 mg/m
3
) or in exhaled air.
Hallier et al. (1994) observed an increase in SCEs among laminators exposed to high concentrations of styrene
(approximately 170 mg/m
3
) but not in formers exposed to much lower levels (43 mg/m
3
). When hygienic and technical
improvements at the workplace reduced the styrene exposure of the laminators to approximately 85 mg/m
3
, a significant
reduction in the SCE levels of the laminators was noted, although their values were still higher than in unexposed
controls.
The few studies available on DNA strand breakage and N-acetylaminofluorene-induced unscheduled DNA synthesis in
peripheral leukocytes have given positive results. DNA strand breaks were shown to disappear quickly from the
peripheral leukocytes following the exposure, so that blood samples collected before the work shift could be used as
control samples (Walles et al., 1993). Reinforced plastics workers showed elevated frequencies of GPA variant
erythrocytes – a measure of mutations in the glycophorin A locus – with a significant effect especially among workers
belonging to a high-exposure group (breathing zone styrene concentration 85 mg/m
3
, 20ppm) (Bigbee et al., 1996).
Another study also suggested an increase in GPA variant frequencies in a high-exposure group (average exposure
concentration 136 mg/m
3
, 32 ppm) as compared with a low exposure group (average exposure to 5.1 mg/m
3
, 1.2 ppm
styrene), but final conclusions were complicated by inadequate matching of the groups (Compton-Quintana et al.,1993).
Vodicka et al. (1995) observed a weak elevation in the frequency of HPRT mutations in T-lymphocytes, while another
study (Tates et al., 1994) was considered inconclusive.
In summary, genotoxic effects have been observed in the blood cells of reinforced plastics workers for various
endpoints at styrene exposure levels of around 85–128 mg/m
3
(20-30 ppm) and above. DNA breakage has been
observed at exposure levels below 43 mg/m
3
. The role of styrene in generating the genotoxic effects seen in reinforced
plastics workers is supported by the elevated levels of styrene-7,8-oxide DNA adducts observed in their peripheral
leukocytes. Such adducts appear to be distinguishable following occupational exposures to only a low concentration of
styrene.
Genotoxicity assays on styrene in experimental animals have given conflicting results. The findings suggest a weak
genotoxic activity for styrene in vivo, as revealed by the most sensitive techniques (SCEs and DNA strand breakage),
especially in the mouse – the rodent species expected to be more susceptible than the rat or Chinese hamster to styrene.
Carcinogenic potential
IARC has classified styrene as a Group 2B, possibly carcinogenic to humans. EPA does not have a carcinogen
classification for styrene; the chemical currently is undergoing an EPA Integrated Risk Information System (IRIS)
review to establish such a classification. ATSDR, Health Canada, and RIVM have evaluated the carcinogenicity data
for styrene. Health Canada classified styrene as "possibly carcinogenic to humans" (Group III) and also a "possible
human germ cell mutagen"(Group III). RIVM concluded that styrene is not a genotoxic compound, and that the
carcinogenic potential of styrene is related to the metabolite styrene oxide, but concentration of this metabolite in
humans is very low due to rapid biotransformation to styrene glycol. Consequently, RIVM derived risk values based on
noncancer endpoints. ATSDR has published a Toxicological Profile for Styrene. Although ATSDR discusses the
carcinogenicity data in its Toxicological Profiles, it does not currently assess cancer potency or perform cancer risk
assessments.
Cases of leukaemia and lymphoma were identified among workers exposed in the manufacture of styrene and
polystyrene, and in the production or manufacture of styrene–butadiene (IARC, 1994).
The INDEX project Final report
209
Epidemiological studies of styrene have been conducted in three types of industry: production of glass-reinforced
plastics products, production of styrene monomers and styrene polymerization, and production of styrene–butadiene
rubber. The malignancies observed in excess most frequently are of the lymphatic and haematopoietic system.
Reinforced plastics industry
In a European multinational study of more than 40 000 workers in the glass-reinforced plastics industry (660 plants), no
overall excess of deaths from lymphatic and haematopoietic cancers was observed in comparison with national controls
(Kogevinas et al., 1994). An increased risk for lymphatic and haematopoietic tumours was observed in Poisson
regression models for years since first exposure (P = 0.012) and for average exposure (P = 0.19) but not for cumulative
exposure. Within the models, there was an increasing trend in risk for lymphatic and haematopoietic cancer with
average intensity of exposure, culminating in a relative risk (RR) of 3.6 (95% confidence interval (CI), 1.0–13) for the
highest category, >852 mg/m
3
; for more than 20 years since first employment, the RR was 4.0 (95% CI, 1.3–12).
Nonsignificant increases in risk with time since first exposure or cumulative exposure were noted for cancers of the
pancreas, kidney, and oesophagus.
A study of cancer incidence in the reinforced plastics industry in Denmark involved 12 800 male workers who had been
included within the above study and a further 24 000 workers with a lower probability of exposure to styrene. The mean
annual air concentrations of styrene calculated for 128 of the companies studied ranged from 767 mg/m
3
in 1964–1970
to 183 mg/m
3
in 1976–1988. Within this cohort, there were 112 malignancies of the lymphatic and haematopoietic
system, with 93.7 expected (standardized incidence ratio (SIR), 1.2; 95% CI, 0.98–1.4). In workers with more than 10
years since first employment, the SIR for leukaemia was 157 (107–222). The excess was concentrated mainly in those
workers not previously included in the European study, in short-term workers with at least 10 years since first
employment and in those employed before 1970 (Kolstad et al., 1994). The same authors also reported the occurrence
of deaths from solid cancers among the 36.610 reinforced plastics workers (Kolstad et al., 1995). An increased
incidence rate ratio (IRR) for pancreatic cancer was found (IRR 2.2, 95% CI, 1.1– 4.5).
In a large study on 5826 employees who had worked for at least 6 months between 1948 and 1977 in 30 reinforced
plastics plants in the United States, no overall increase in risk for lymphatic and haematopoietic malignancies was
observed (Wong, 1990; Wong et al., 1994). The follow-up continued until the year 1989. The overall mortality rate was
108 (95% CI, 103–113), and the mortality rate from all cancers was 116 (95% CI, 105–127). Total lympho-
haematopoietic cancers showed no excess. For workers involved in open-mould processing with high exposure to
styrene, the standardized mortality ratio (SMR) for lymphatic and haematopoietic cancers was 141 (based on four
cases). For the highest cumulative exposure (>426 mg/m
3
× years) and more than 20 years of latency, the SMR was 134
(5–373). Mortality for cancers at a number of sites was increased significantly. These included oesophagus (mortality
rate 198; 95% CI, 105–322), bronchus, trachea, and lung (141; 120–164), cervix uteri (284; 136–521), and other female
genital organs (202; 107–345). No positive dose–response relationship was found for the lympho-haematopoietic
cancers or any other cancer in excess; IARC (1994), however, noted that the possibility that the two exposure variables
included in the regression model were correlated may have reduced the likelihood of accurate assessment
A smaller study of 5021 employees in two reinforced plastics boat-building facilities in the United States showed no
deaths from leukaemia or lymphomas (1.7 and 2.1 expected, respectively) (Okun et al., 1985). 2060 individuals were
considered to have had high exposure to styrene (mean levels of styrene in the air in two facilities, 181 and 305 mg/m
3
);
48% of them had worked only for one month to one year and only 5% for more than 5 years. In this group, no
lymphatic or haematopoietic cancers were seen (about 1 expected).
A deficit of deaths from lymphatic and haematopoietic malignancies (6 observed, 14.9 expected) was reported in a
cohort of 7949 men and women employed during 1947–1984 in eight companies in the United Kingdom manufacturing
glass-reinforced plastics involving high exposure to styrene (Coggon et al., 1987). A nonsignificant excess was seen in
deaths from cancers of the lung, pleura and mediastinum (SMR, 126; 95% CI, 94–166); this finding particularly related
to workers who had had 1–9 years of exposure to styrene, but the risk did not increase with time from the first exposure.
Follow-up of this cohort was later extended to 1990 as part of an international collaborative study (Kogevinas et al.,
1994). By that time the previous deficit of lymphatic and haematopoietic cancer had largely disappeared (13 deaths;
SMR, 88; 95% CI, 47–151) and the excess of lung cancer was less marked (77 deaths; SMR, 106; 95% CI, 84–132).
Styrene manufacture and polymerization
A study of chemical workers in the production of styrene and styrene derivatives in the United States found seven
deaths from lymphatic and haematopoietic malignancies (except leukaemias) (SMR, 132; 95% CI, 58–272) and six
from leukaemias (SMR 176; 95% CI, 64–383) (Ott et al., 1980). In an update, a total of 28 deaths from lymphatic and
haematopoietic cancer were recorded (SMR 144; 95% CI, 95–208) (Bond et al., 1992).
The INDEX project Final report
210
A smaller United Kingdom study of 622 men exposed for at least a year in the production, polymerization and
processing of styrene found an excess of deaths from lymphoma (3 observed, 0.56 expected) (Hogdson and Jones,
1985).
Styrene–butadiene rubber production
The large cohorts of styrene–butadiene rubber workers showed increased risks for lymphatic and haematopoietic
malignancies, but nested case–control analyses found little evidence for relationship to styrene (Matanoski et al., 1990;
Santos-Burgoa et al., 1992; Matanoski et al., 1993). The styrene exposures in these industries are at least one order of
magnitude lower than in the reinforced plastics industry.
In summary, several epidemiological studies have suggested that workers exposed to styrene in the reinforced plastics
industry have increased risk of lymphatic and haematopoietic tumours. The studies are not, however, fully conclusive
because the observed associations are often based on small numbers, and in larger studies, dose relations are somewhat
obscure.
Interactions with other chemicals
Styrene metabolism is known to be inhibited by the presence of other chemicals such as toluene, trichloromethylene,
and ethyl benzene. The biotransformation of styrene in rats to PGA, MA, and hippuric acid was suppressed by
coadministration of toluene (Ikeda et al. 1972). This may be due to competitive inhibition of oxidative mechanisms.
Similar results were reported by Ikeda and Hirayama (1978) in rats when styrene metabolism was inhibited by the
administration of trichloroethylene. Urinary metabolites of styrene may be markedly reduced when humans or animals
are concurrently exposed to organic solvents that inhibit styrene metabolism.
Odour perception
Source: WHO (2001)
The odour threshold for styrene is only 70 µg/m
3
(0.016 ppm). Its characteristic pungent odour is recognized at
concentrations 3–4 times higher than this threshold value. Some individuals can perceive the odour at levels lower than
70 µg/m
3
, but, in general, odour problems are not likely to occur if peak concentrations in the ambient air are kept
below this threshold value. When styrene is emitted into the air, its half-time is estimated to be 2 hours. In ambient air it
is chemically transformed into benzaldehyde and formaldehyde, both of which are odorous air pollutants (WHO, 1987).
Source: AIHA (1989)
Detection: 72 – 8100 µg/m
3
(0.017-1.9 ppm)
Recognition: 640 µg/m
3
(0.15 ppm)
Source: New Jersey department of health and senior services - Hazardous Substance Fact Sheet
Odor threshhold = 85 – 2000 µg/m
3
(0.02 - 0.47 ppm)
Source: (Hoshika et al., 1993)
Barely perceptible concentration level of styrene: 0.068/0.140 mg/m
3
(in The Nederlands and Japan, respectively)
International comparison of odor threshold values of several odorants in Japan and in The Netherlands, given as the
barely perceptible concentration level revealed striking similarities for hydrogen sulfide (in Japan 0.0005 ppm/in The
Netherlands 0.0003 ppm), phenol (0.012/0.010), styrene (0.033/0.016), toluene (0.92/0.99), and tetrachloroethylene
(1.8/1.2) but not for m-xylene (0.012/0.12). Such a similarity was not found with any other literature sources.
The INDEX project Final report
211
Summary of Styrene Dose Response Assessment
________________________________________________________________________________________________
Exposure other than inhalation: Dermal absorption from ambient air <5% of the dose absorbed in the respiratory tract
Toxicokinetics: 60-70% pulmonary retention; widely distributed throughout the body; styrene-oxide in blood linearly
correlated with ambient styrene; no evidence of accumulation at occupational levels (160 mg/m
3
) due to fast enzymatic
oxidation/hydrolysis to MA and PGA and biphasic elimination; MA+PGA used for biological monitoring.
Health effect levels of short- and long-term exposure (noncancer)
NOAEL
mg/m³
LOAEL
mg/m³
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
Short-term exposure
217
EXP
422
EXP
Eye and throat irritation
Volunteers,
1h (NOAEL),
20m (LOAEL)
Stewart et al., 1968 OEHHA (1999);
REL: 21
Long-term exposure
64
Study
17
MLE
7.2
BC05
2.6
ADJ
CNS; neuropsychological tests: reaction time, s/l
term logic memory, s/l term verbal memory,
digit-symbol association, block design and figure
identification.
Occupational, 8.6y Mutti et al. 1984b
OEHHA (1999);
REL: 0.9
94
95%lCL
34
ADJ
>94
95%lCL
see above Occupational, 8.6y;
Mutti et al. 1984b
EPA-IRIS (1992)
; RfC: 1
107
Study
25
ADJ
see above Occupational 8.6y; Mutti et al. 1984b ATSDR (1992);
MRL: 0.25;
WHO (2001);
GV: 0.26;
RIVM (2000)
260
EXP
65
ADJ
46
HEC
Neurological developmental; body weight,
biochemical parameters in the brain and
behaviour
Rat offspring Kishi et al.,1992a,b Health Canada
(1993);
TC: 0.092
Carcinogenicity: IARC: 2B ; ACGIH: A4 ; Little evidence of carcinogenicity in humans (WHO)
Genotoxicity: Genotoxic in vitro and in vivo; increased chromosomal aberrations evidenced in numerous occupational
studies; proposed NOAELs based on this endpoint were >85 mg/m
3
. Although genotoxic effects in humans have been
observed at relatively low concentrations, they were not considered as critical endpoints for development of a guideline,
in view of the equivocal evidence for the carcinogenicity of styrene (WHO).
Odour threshold: Range: 0.072 - 8.1 mg/m
3
, recognition: 0.64 mg/m
3
(AIHA); Threshold: 0.07 mg/m
3
(WHO); 0.23
(Devos); Barely perceptible concentration levels: 0.140/0.068 mg/m
3
in Japan/The Nederlands, respectively (Hoshika et
al., 1993).
Susceptible population: Asthmatics may be more sensitive to adverse pulmonary effects; immediate
bronchoconstriction observed in occupational (styrene) asthmatics at 64 mg/m
3
.
Remarks: RD
50
: 3.3 g/m
3
; styrene toxicity is enhanced (metabolism slowed down, with increase of styrene oxide in
blood) by exposure to other solvents (incl.ethanol)
________________________________________________________________________________________________
The INDEX project Final report
212
6. Risk Characterization
Health hazard and cancer risk evaluation of short- and long-term exposure
(Adapted from WHO, 2001 - Air Quality Guidelines for Europe)
Potentially critical effects for the derivation of an exposure limit (EL) for styrene are considered to be
carcinogenicity/genotoxicity and neurological effects, including effects on development. The value of the available
evidence for an association between exposure to styrene and small increases in lymphatic and haemotopoietic cancers
observed in workers in some studies is limited by concurrent exposure to other substances, lack of specificity and
absence of dose–response. In limited studies in animals, there is little evidence that styrene is carcinogenic. IARC has
classified styrene in group 2B.
Styrene was genotoxic in vivo and in vitro following metabolic activation. In cytogenetic studies on peripheral
lymphocytes of reinforced plastics workers, there were increased rates of chromosomal aberrations at mean levels of
styrene of more than 120 mg/m
3
. Elevated levels of single-strand breaks and styrene-7,8-oxide adducts in DNA and
haemoglobin have also been observed. Although these genotoxic effects have been observed at relatively low
concentrations, they were not considered as critical endpoints for the development of a WHO Air Quality Guideline, in
view of the equivocal evidence of carcinogenicity for styrene.
The available data, although limited, indicate that neurotoxicity in the form of neurological developmental impairments,
is among the most sensitive of endpoints. In the offspring of rats exposed to styrene at a concentration of 260 mg/m
3
,
there were effects on biochemical parameters in the brain and behaviour.
Studies in workers exposed to styrene in workplace air suggest that neurological effects are probably the most sensitive
indicator of styrene toxicity. Available data do not.provide a clear picture of the NOAEL for neurological effects
following acute- or intermediate-duration inhalation exposure. A chronic study in workers (Mutti et al. 1984a) identified
subtle effects such as reductions in visuomotor accuracy and verbal learning skills, and subclinical effects on colour
vision have been observed at concentrations as low as 107–213 mg/m
3
. Taking the lower number of this range as a
LOAEL (for precautionary reasons) and adjusting this value in order to allow for conversion from an occupational to a
continuous pattern of exposure (dividing by 4.2) a concentration of 25 mg/m
3
is obtained. Incorporating a factor of 10
for interindividual variation and 10 for use of a LOAEL rather than a NOAEL this results in an EL of 0.25 mg/m
3
(to be
applied as a weekly average; see also Table 6.1). This value should also be protective for the developmental
neurological effects observed in animal species.
Since available data do not.provide a clear picture of the NOAEL for neurological effects following acute exposure, a
short-term EL was here derived based on irritation of the eyes and mucous membranes. Stewart et al. (1968) found eye
and throat irritation in 3 out of 6 volunteers exposed to 422 mg/m
3
styrene for 20 minutes. No symptoms were reported
in 3 subjects after exposure to 217 mg/m
3
for 1 hour. A short-term EL is here derived applying an assessment factor of
100 for interindividual variation (10) and the possibility (limited evidence) that asthmatics may be more sensitive to
adverse pulmonary effects (10) from styrene exposure (Moscato et al., 1987).
The odour threshold for styrene is only 70 µg/m
3
. Its characteristic pungent odour is recognized at concentrations 3–4
times higher than this threshold value. Some individuals can perceive the odour at levels lower than 70 µg/m
3
, but, in
general, odour problems are not likely to occur if peak concentrations in the ambient air are kept below this threshold
value.
Table 6.1: Derivation of short- and long-term exposure limits
Effect level - mg/m
3
Assessment
factor
EL
mg/m
3
Toxicological endpoint
Short-term
Exposure
Limit
Human NOAEL
Volunteers
217 100
bc
2 Eye and throat irritation
Long-term
Exposure
Limit
Human LOAEL
Occupational
25 100
ab
0.25
CNS; neuropsychological tests: reaction
time, s/l term logic memory, s/l term verbal
memory, digit-symbol association, block
design and figure identification.
a
not considering a NOAEL (10);
b
interindividual variation (10) ;
c
susceptible population (10)
The INDEX project Final report
213
Relevance of EU-population exposure to styrene
Table 6.2: Percentage of population exposed beyond the derived EL and margins of safety
Available exposure data
EL
Margins of
Safety (MOS)
Description (Study, Year) N 0.25* mg/m
3
50
th
(90
th
)
Athens 30-h TWA (Expolis, 96-98) 42 < 181 (71)
Basel 30-h TWA (Expolis, 96-98) 47 < 470 (182)
Helsinki 30-h TWA (Expolis, 96-98) 188 < 184 (103)
Milan 30-h TWA (Expolis, 96-98) 38 < 77 (20)
Oxford 30-h TWA (Expolis, 98-00) 40 < 225 (64)
Prague 30-h TWA (Expolis, 96-98) 46 < 164 (64)
German Survey 48-h PEM (GerEs II,
1990-92)
113 < 125 (36)
French National Survey 7d-TWA (IAQ
observatory; 2003-04)
109 < 250 (96)
Avg. MOS: 235 (79)
< out of the evaluation range (i.e. <5% of the environments investigated);
* corresponds with WHO guideline value (weekly average)
Result
A long-term exposure limit of 250 µg/m
3
has been derived based on the assumption that neurological effects are
probably the most sensitive indicator of styrene toxicity. When examining the results of eight monitoring surveys (Table
6.2) it can be concluded that background styrene concentrations in European residences are of no concern to human
health since median levels are, on average, two orders of magnitude below the established EL. Although no acute
exposure data were available, it is unlikely that styrene emissions associated with human indoor activities would
generate levels up to the proposed short-term EL of 2000 µg/m
3
, considered protective for irritative effects in
asthmatics. Although genotoxic effects in humans have been observed at relatively low concentrations, they were not
considered as critical endpoints for the derivation of the exposure limit, in view of the equivocal evidence for the
carcinogenicity of styrene in humans (WHO).
The INDEX project Final report
214
Ammonia
Synonyms: -
CAS Registry Numbers: 7664-41-7
Molecular Formula: NH
3
1. Compound identification
Ammonia is a colourless gas with a sharp, irritating odour. Ammonia has both natural and anthropogenic sources. It is a
key compound in the global nitrogen cycle. It is formed in the body during decomposition of organic materials. The
most common natural sources of ammonia include the natural breakdown of manure and dead plants and animals.
Majority of all manufactured ammonia is used as fertilizers. It is also released to the atmosphere by leaks during
commercial synthesis, production, and transportation, or sewage, burning of coal, wood, and other natural products, and
volcanic activity. Ammonia is used to manufacture synthetic fibres, plastics, explosives, and many cleaning products. It
is also used as a refrigerant. (WHO 1997, ATSDR, 2002)
Since ammonia occurs naturally in the environment, the general population is exposed to ammonia in air, soil, and
water. People are frequently exposed to ammonia while using household products that contain ammonia, such as
cleaning solutions, window cleaners, floor waxes, and smelling salts. Household ammonia solutions usually contain 5-
10% ammonia in water. Also those who live near farms or cattle feedlots, poultry confinement buildings, or in the
vicinity of other areas with high animal populations may be exposed to elevated ammonia levels (ATSDR 2002,
EPA/Cal 2003).
Typical environmental ammonia concentrations have not been reported to cause adverse health effects in general
population. However, low levels of ammonia may harm some asthmatics and other sensitive individuals (ATSDR
2002).
2. Physical and Chemical properties
Molecular weight (g/mol) 17.03
Melting point (°C) -77.7
Boiling point (°C) 33.35
Density (g/l at 0 oC, 1 atm) 0.77
Relative density (air =1) 0.59
Solubility: Soluble in water and oxygenated solvents
Conversion factors at 20 °C and 760 mm Hg:
1 ppb = 0.707 µg/m3
1 µg/m3 = 1.414 ppb
Source: Verschueren 2001, HSDB 2003.
3. Indoor Air Exposure assessment
Indoor air and exposure concentrations
There is a lack of knowledge and publications about indoor concentrations and exposures of ammonia. Ammonia is
studied mostly as an ambient pollutant. Typical ambient background concentrations of ammonia are between 0.71
The INDEX project Final report
215
µg/m
3
and 3.55 µg/m
3
(1 and 5 ppb) (ATSDR 2002). Fisher et al (2003) determined indoor and outdoor concentrations
of ammonia in an unoccupied residence in Clovis California from October 2000 to January 2001. Monthly mean indoor
concentrations ranged from 10.9 µg/m
3
to 15.1 µg/m
3
, while mean outdoor concentrations were always lower ranging
from 8.80 µg/m
3
to 9.66 µg/m
3
.
Considerably high ammonia concentrations have been measured in new buildings built in 1994–1997 in Finland.
Average concentrations exceeded 50 µg/m
3
. Construction materials such as glues, fillers used in floors, walls and
ceilings, have been identified as ammonia sources in indoor environments. Ammonia is emitted from these materials by
chemical breakdown processes, especially in the presence of moisture.
Puhakka et al (2000) studied reparation methods to reduce ammonia concentrations in ‘ammonia problem’ buildings.
They found the highest indoor air concentrations during preparation and very high levels inside the floor structures up
to 15370 µg/m
3
. The reparation of floor did not reduce ammonia concentrations in two months after the reparation, but
in other studies indoor concentrations started to decline in 6–8 months after the reparation.
Residential indoor concentrations of ammonia in homes with and without known indoor air quality (IAQ) problems
have been monitored in Finland (VTT 2003). Average indoor concentrations of ammonia in homes with and without
IAQ problems were 30 µg/m
3
and 24 µg/m
3
, respectively. Especially, if the building materials became wet due to water
damage or poor ventilation of the building structures, elevated ammonia concentrations were usually detected in indoor
air. Casein-containing materials were identified a source of ammonia in these cases (Saarela, 2003). Cumulative
distributions of ammonia in homes with and without IAQ problems in Finland are presented in Figure 3.1. Short time
mean concentrations of ammonia are summarised in Table 3.1.
0
20
40
60
80
100
0 10203040506070
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
normal IAQ
IAQ problems
Figure 3.1. Cumulative distributions of residential indoor concentrations of ammonia in Finland in homes without and
with indoor air quality (IAQ) problems (VTT 2003).
The INDEX project Final report
216
Table 3.1. Short time ammonia concentrations related to specific microenvironments or emission sources.
Averaging time Reference
A
Mrange
Problem buildings before floor repairs 27 - 79 Puhakka et al 2000
during repairs 46 - 237
2 months after repairs 29 - 99
inside the floor structure 3810 - 15370
Dwellings empty dwellings 100-min 21 5 - 49 Saarinen et al 2002
inhabited dwellings 100-min 9 - 90
New apartments 2-hour 21 - 42 Tuomainen and Pirinen 2002
Toilets with septic tank 10-minute 1244 Hyung-Suk and Byung-Kee 1993
traditional vault system 7395
Homes kerosene heaters 8 -12-hour 11 Leaderer et al 1993
gas range 8 -12-hour 8.5
no gas appliances 8 -12-hour 7.1
Archives of the university library Helsinki downtown 4-12 Raiala et al 1993
Mikkeli < 5
AM = arithmetic mean
Concentration (µg/m
3
)
Environment or
emission source
4. Toxicokinetics
Absorption
At low concentrations, inhaled ammonia dissolves in the mucous fluid lining of the upper respiratory tract and little
reaches the lower airways. At ammonia levels associated with ambient air (i.e., 1 - 200 µg/m
3
), very little, if any, is
absorbed through the lungs.
Experiments with volunteers show that ammonia, regardless of its tested concentration in air (range = 40–350 mg/m
3
),
is almost completely retained in the nasal mucosa (83–92%) during short-term exposure, i.e., up to 120 seconds
(Landahl and Herrmann 1950). However, longer-term exposure (10–27 minutes) to a concentration of 350 mg/m
3
resulted in lower retention (4–30%), with 244-279 mg/m
3
eliminated in expired air by the end of the exposure period
(Silverman et al. 1949), suggesting an adaptive capability or saturation of the absorptive process. Nasal and pharyngeal
irritation, but not tracheal irritation, suggests that ammonia is retained in the upper respiratory tract. Unchanged levels
of blood-urea-nitrogen (BUN), non-protein nitrogen, urinary-urea, and urinary-ammonia are evidence of low absorption
into the blood.
Distribution
Absorption data from human inhalation exposure suggest that only small amounts of ammonia are absorbed into the
systemic circulation (Silverman et al. 1949; WHO 1986). Toxic effects reported from inhalation exposure suggest local
damage, or changes resulting from necrotic tissue degradation, rather than the presence of elevated levels of NH
4
+
, per
se, in tissues other than the respiratory/pharyngeal tissues.
Information on the distribution of endogenously-produced ammonia suggests that any NH
4
+
absorbed through
inhalation would be distributed to all body compartments via the blood, where it would be used in protein synthesis or
as a buffer, and that excess levels would be reduced to normal by urinary excretion, or converted by the liver to
glutamine and urea. If present in quantities that overtax these organs, NH
4
+
is distributed to other tissues and is known
to be detoxified in the brain (Takagaki et al. 1961; Warren and Schenker 1964).
Metabolism and elimination
Quantitative data on human metabolism of exogenously introduced ammonia were not located in the available
literature.
The INDEX project Final report
217
Ammonia and ammonium ion are metabolized to urea and glutamine mainly in the liver (Fürst et al., 1969; Pitts,1971).
However, it can be rapidly converted to glutamine in the brain and other tissues as well (Takagaki et al. 1961; Warren
and Schenker 1964).
Studies using low levels of ammonia show that inhaled ammonia is temporarily dissolved in the mucus of the upper
respiratory tract, and then a high percentage of it is released back into the expired air.
The quantitative difference between inspired and expired ammonia suggests that small amounts are absorbed across the
nasopharyngeal membranes into the systemic circulation. Absorbed ammonia is excreted by the kidneys as urea and
urinary ammonium compounds (Gay et al. 1969; Pitts 1971; Richards et al. 1975; Summerskill and Wolpert 1970), as
urea in feces (Richards et al. 1975), and as components of sweat (Guyton 1981; Wands 1981), but quantitative data are
lacking. Toxic levels do not develop as a result of chronic inhalation exposure because the body has multiple effective
mechanisms for detoxifying and excreting it.
5. Health effects
Effects of short-term exposure
The available human and animal data provide strong evidence that acute-duration exposure to ammonia can result in
site-of-contact lesions primarily of the eyes and the respiratory tract. Even fairly low airborne concentrations (35
mg/m
3
) of ammonia produce rapid onset of eye, nose, and throat irritation, coughing, and narrowing of the bronchi.
More severe clinical signs include immediate narrowing of the throat and swelling, causing upper airway obstruction
and accumulation of fluid in the lungs. This may result in low blood oxygen levels and an altered mental status.
Mucosal burns to the tracheobronchial tree can also occur. Children may be more vulnerable to corrosive agents than
adults because of the smaller diameter of their airways.
The eye is especially sensitive to alkali burns. Ammonia combines with moisture in the eyes and mucous membranes to
form ammonium hydroxide. Ammonium hydroxide causes saponification and liquefaction of the exposed, moist
epithelial surfaces of the eye and can easily penetrate the cornea and damage the iris and the lens (CCOHS, 1988; Way
et al., 1992). Damage to the iris may eventually lead to cataracts (CCOHS, 1988).
Silverman and coworkers (1949) exposed 7 volunteers to 348 mg/m³ (500 ppm) ammonia for 30 minutes using an oral-
nasal mask. Symptoms due to ammonia inhalation varied widely among the 7 subjects. All seven subjects experienced
upper respiratory irritation, which was graded as severe in 2 subjects. Only 2 subjects were able to continue nasal
breathing throughout the 30 minute exposure. Reactions included irritation of the nose and throat, hypoesthesia of the
exposed skin, and lacrimation. In two subjects, the nasopharyngeal irritation persisted for 24 hours after the exposure.
One of the 7 subjects was only exposed to ammonia for 15 minutes rather than the full 30 minutes. The reason for this
deviation in the exposure regimen was not given. In a previous experiment, brief exposure to 696 mg/m³ (1000 ppm)
reportedly resulted in immediate coughing in human subjects.
Ferguson and coworkers (1977) used six human subjects to demonstrate that a tolerance to ammonia exposure of 70
mg/m³ (100 ppm) can be developed with a two-to-three week inurement period during which volunteers were exposed
to lesser concentrations. The results tended to support the belief that persons with no recent history of ammonia
exposure are more sensitive to the irritating effects than those who are acclimated to the noxious gas.
Verberk (1977) exposed sixteen volunteers (8 science faculty with knowledge of the effects of ammonia and 8 non-
science university students not familiar with ammonia health effects) were exposed, 4 at a time, to 35, 56, 77, 98 mg/m³
(50, 80, 110, and 140 ppm) ammonia for 2 hours. Each group was exposed to each exposure level with 1 week in
between exposures. Immediately before and after exposure, respiratory function tests (vital capacity [VC], forced
expiratory volume in the first second [FEV
1], and forced inspiratory volume in the first second [FIV1]) were done.
During exposure, each participant recorded subjective effect levels for smell, taste, irritation of eyes, irritation of nose,
irritation of throat, irritation of breast, urge to cough, headache, and general discomfort. The scale used was: 0=no
sensation, 1=just perceptible, 2=distinctly perceptible, 3=nuisance, 4=offensive, and 5=unbearable. A (+) or (–) could
be used to interpolate between the levels. A few weeks after the experiments, the histamine threshold was determined
for 13 of the 16 volunteers as a measure of pre-existing non-specific reactivity of the airways to exogenous stimuli.
None of the participants was hypersusceptible to nonspecific irritants. No participant had a decrease of more than 10%
of pre-exposure values for VC, FEV
1, or FIV1. There was a difference between the science faculty group (experts) and
the students for the subjective scoring. Students consistently scored higher for smell and there was little increase in
score with concentration. Score for irritation of the eyes increased with concentration and there was no difference
The INDEX project Final report
218
between groups. Irritation of the throat had a sharp increase in score with concentration and scores were higher for
students. All students left the exposure chamber between 0.5 and 1.25 hours in the 98 mg/m³ exposure because of
severe irritation. Scores for urge to cough and general discomfort were low in the expert group, but increased with
concentration in the student group. All students left the chamber before 2 hours of exposure to 98 mg/m³.
At the end of the initial 30 minutes of the 2-hour exposure period, nuisance level smell, eyes, nose, or throat irritation,
or cough urge were reported by 7 of 16 (44%), 9 of 16 (56%), 12 of 16 (75%), or 15 of 16 (94%) individuals at
concentrations of 35, 56, 77, 98 mg/m³, respectively.
MacEwen et al. (1970) exposed groups of 5 and 6 human subjects to respective ammonia concentrations of 21 and 35
mg/m³ (30 and 50 ppm). The volunteers subjectively rated irritation for the 10-minute exposures. No moderate or higher
irritation was discerned by the group at the lower exposure level; however, 4 of the 6 subjects rated the 10 minute
exposure at 35 mg/m
3
as causing moderate irritation.
The Industrial Bio-Test Laboratories (1973) evaluated ten human subjects for the irritation threshold of ammonia from
exposures to ammonia gas at four different concentrations: 22, 35, 50, and 93 mg/m³. Irritation was taken to be any
annoyance to the eyes, nose, mouth, throat, or chest which persisted throughout the 5-minute exposure period. At 50
mg/m³ three subjects experienced eye irritation, two had nasal irritation, and three had throat irritation. At 93 mg/m³,
five of the ten subjects experienced lacrimation and eye irritation, seven complained of nasal irritation, eight had throat
irritation, and one experienced chest irritation. The authors only used 5-minute exposure durations; and it is possible
that irritation symptoms could have developed with longer exposure durations at the lower exposures. The authors
discounted the significance of nasal dryness reported at the two lowest levels.
Douglas and Coe (1987) determined a lachrymatory threshold of 38 mg/m³ for ammonia following approximately 15
second exposures of volunteers via tight-fitting goggles. The threshold for bronchoconstriction, determined as a 20%
increase in airway resistance, was slightly higher at 59 mg/m³ following 10 breaths of ammonia via mouthpiece.
Tolerance appears to develop with repeated exposure (Sekizawa and Tsubone 1994; Verberk 1977). Thus, subjects
exposed to 70, but not 35 mg/m
3
ammonia 6 hours/day, 5 days/week for 6 weeks experienced nose and throat irritation
only during the first week (Ferguson et al. 1977).
Estimates of odor thresholds for ammonia vary from 0.03-72 mg/m³ (Ferguson et al., 1977; Henderson and Haggard,
1943; Ruth, 1986). Near the odor threshold, persons exposed to ammonia can experience annoyance and believe the
odor to be a nuisance. Exposure to ammonia may result in an exacerbation of preexisting asthma. Shim and Williams
(1986) surveyed 60 patients with a history of asthma worsened by certain odors. Nearly 80% of these patients claimed
to have an exacerbation of asthma following exposure to household cleaners containing ammonia.
Summary of short-term exposure effect levels
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
14.0
1h-ADJ-MLE
9.5
BC05
eye and respiratory irritation volunteers,
1h-adjusted
Benchmark approach
Industrial Biotest
Laboratories, 1973;
MacEwen et al., 1970;
Silverman et al., 1949;
Verberk, 1977
OEHHA 1999;
REL: 3.2
35
EXP
irritation to nose and throat; urge to cough volunteers, 2h Verberk, 1977 ATSDR 2002;
MRL: 1.2
No study with asthmatics!
Final statement (UNIMI): Human LOAEL: 35 mg/m³
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
The INDEX project Final report
219
OEHHA (1999)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Derivation of Acute Reference Exposure Level protective against mild adverse effects (for a 1-hour exposure)
Study Industrial Biotest Laboratories, 1973; MacEwen et al., 1970;
Silverman et al., 1949; Verberk, 1977
Study population humans
Exposure method inhalation
Critical effects eye and respiratory irritation
LOAEL varied (see the text)
NOAEL varied (see the text)
Exposure duration varied (see the text)
Extrapolated 1 hour concentration 9.5 mg/m
3
(BC05)
LOAEL uncertainty factor not needed in BC approach
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 3
Cumulative uncertainty factor 3
Reference Exposure Level 4.5 ppm (3.2 mg/m³; 3200 µg/m³)
The exposure concentrations from the 4 studies were adjusted to 1-hour durations using the formula C
n
x T = K (Table
5.2). The value for the exponent n was empirically derived from the preceding data sets. The value of n (in the formula
C
n
x T = K) was sequentially varied for the log-normal probit relationship analysis. Using a chi-square analysis, a value
of n = 4.6 was found to be the best fit.
The REL was calculated by a benchmark concentration (BC) approach using a log-normal probit analysis (Crump and
Howe, 1983; Crump. 1984). The 95% lower confidence limit of the concentration expected to produce a response rate
of 5% is defined as the BC
05. The maximum likelihood estimate for a 5% response was 14.0 mg/m
3
(20.1 ppm) and the
95% LCL on this value (BC
05) for ammonia from this analysis was 9.5 mg/m
3
(13.6 ppm).
An uncertainty factor (UF) of 3 was used to account for intraspecies variation in the human population.
Table 5.2: Ammonia, Human Irritation, 60 Minute Exposures (adjusted), mg/m
3
Study Study
concentration
Exposure time 60 min
adjusted
concentration
Response
Industrial Biotest Laboratories, 1973 22 5 13 0/10
MacEwen et al., 1970 21 10 14 0/5
Industrial Biotest Laboratories, 1973 35 5 20 0/10
MacEwen et al., 1970 35 10 24 4/6
Industrial Biotest Laboratories, 1973 50 5 29 3/10
Verberk, 1977 35 120 30 7/16
Verberk, 1977 56 120 48 9/16
Industrial Biotest Laboratories, 1973 93 5 54 8/10
Verberk, 1977 77 60 66 12/16
Verberk, 1977 98 60 84 15/16
Silverman et al., 1949 348 30 299 7/7
Table adapted from: Verberk, 1977; Industrial Biotest Laboratories, 1973; MacEwen et al., 1970; and Silverman et al., 1949. The two
lowest concentrations were combined for the log-probit analysis since this improved the fit of the data.
Response rate MLE (mg/m
3
) 95% LCL
(mg/m
3
)
1% 9.3 5.4
5% 14.0 9.5 (BC
05
)
The INDEX project Final report
220
Graphic Representation of Benchmark Concentration Determination
ATSDR (1999)
Agency for Toxic Substances and Disease Registry
Derivation of a Minimal Risk Level (MRL):
An acute inhalation MRL of 1.2 mg/m³ (1.7 ppm) was derived from the Verberk (1977) study. Dose and end point used
for MRL derivation are 35 mg/m³ (50 ppm) for mild irritation to the eyes, nose, and throat in humans exposed to
ammonia gas for 2 hours. An Uncertainty Factors of 30 was used in MRL derivation: 3 for use of a minimal LOAEL
and 10 for human variability
The MRL is supported by other observations of respiratory effects associated with acute- and intermediate-duration
exposure including transient irritation of the nose and throat of humans exposed to 70 mg/m
3
(Ferguson et al. 1977);
nasal discharge in rats at 262 mg/m
3
(Coon et al. 1970); nasal lesions in rats at 104 mg/m
3
(Broderson et al. 1976); and
nasal inflammation and lesions in rats at 348 mg/m
3
(Richard et al. 1978). A study of piggerie workers exposed to a
mean level of 5.5 mg/m
3
ammonia measured lung function change over a workshift; a small but borderline significant
decrease in lung function was noted (Heederik et al. 1990). This was not used as a basis for MRL derivation because the
workers were also exposed to other potential respiratory toxicants (dust and endotoxins).
Effects of long-term exposure
Several studies have examined the relationship between chronic exposure to ammonia and respiratory effects. Studies of
farmers working in enclosed livestock facilities provide evidence that ammonia may contribute to transient respiratory
distress (Choudat et al. 1994; Cormier et al. 2000; Donham et al. 1995, 2000; Heederik et al. 1990, 1991; Melbostad
and Eduard 2001; Reynolds et al. 1996; Vogelzang et al. 1997, 2000); however, co-exposure to total dust, respirable
dust, carbon dioxide, total endotoxins, respirable endotoxins, fungi, bacteria, and/or molds complicates the
interpretation of these studies.
The INDEX project Final report
221
Comparisons were made between 52 workers and 31 control subjects in a soda ash plant for pulmonary function and
eye, skin and respiratory symptomatology (Holness et al., 1989). The pulmonary function tests included FVC (forced
vital capacity – the total amount of air the subject can expel during a forced expiration), FEV
1 (forced expiratory
volume in one second), FEF50 (forced expiratory flow rate at 50% of the FVC) and FEF75 (forced expiratory flow rate
at 75% of the FVC). Age, height, and pack-years smoked were treated as covariates for the comparisons. The workers
were exposed on average for 12.2 years to mean (time-weighted average) ammonia concentrations of 6.4 ± 1.0 mg/m
3
(9.2 ± 1.4 ppm), while controls were exposed to 0.21 ± 0.7 mg/m
3
(0.3 ± 0.1 ppm). No differences in any endpoints
(respiratory or cutaneous symptoms, sense of smell, baseline lung function, or change in lung function over a work shift
at the beginning and end of a workweek) were reported between the exposed and control groups.
Groups of human volunteers were exposed to 0, 17, 35, or 70 mg/m
3
(25, 50, or 100 ppm) ammonia 5 days/week for 2,
4, or 6 hours/day, respectively, for 6 weeks (Ferguson et al., 1977). Another group of 2 volunteers was exposed to 35
mg/m
3
ammonia for 6 hours/day for 6 weeks. Pulmonary function tests (respiration rate, FVC and FEVl) were measured
in addition to subjective complaints of irritation of the eyes and respiratory tract. The difficulty experienced in
performing simple cognitive tasks was also measured, as was pulse rate. There were reports of transient irritation of the
nose and throat at 35 or 70 mg/m
3
. Acclimation to eye, nose, and throat irritation was seen after two to three weeks (in
addition to the short-term subjective adaption). No significant differences between subjects or controls on common
biological indicators, in physical exams, or in performance of normal job duties were found. After acclimation,
continuous exposure to 70 mg/m
3
, with occasional excursions to 139 mg/m
3
, was easily tolerated and had no observed
effect on general health.
Broderson et al. (1976) exposed groups of F344 rats (6/sex/dose) continuously to 17, 35, 104 or 174 mg/m
3
ammonia
(HEC = 1.9, 3.7, 11.2 or 18.6 mg/m
3
, respectively) for 7 days prior to inoculation with Mycoplasma pulmonis and from
28-42 days following M. pulmonis exposure. Each treatment group had a corresponding control group exposed only to
background ammonia and inoculated with M. pulmonis in order to produce murine respiratory mycoplasmosis (MRM).
The following parameters were used to assess toxicity: clinical observations and histopathological examination of nasal
passages, middle ear, trachea, lungs, liver and kidneys. All levels of ammonia, whether produced naturally or derived
from a purified source, significantly increased the severity of rhinitis, otitis media, tracheitis and pneumonia
characteristic of M. pulmonis. Furthermore, there was a significant concentration response between observed
respiratory lesions and increasing environmental ammonia concentration for gross and microscopic lesions. All lesions
observed were characteristic of MRM. Gross bronchiectasis and/or pulmonary abscesses and the extent of gross
atelectasis and consolidation was consistently more prevalent in exposed animals at all concentrations than in their
corresponding controls. The severity of the microscopic lesions in the nasal passages, middle ears, tracheas and lungs
was significantly greater in all exposed groups compared with controls. Increasing ammonia concentration was not
associated with an increasing frequency of M. pulmonis isolations. Additionally, rats not exposed to M. pulmonis and
exposed to ammonia at 174 mg/m
3
developed nasal lesions (epithelial thickening and epithelial hyperplasia) unlike
those observed in inoculated rats.
The growth of bacteria in the lungs and nasal passages, and the concentration of serum immunoglobulin were
significantly increased in rats exposed to 70 mg/m
3
(100 ppm) ammonia over that seen in control rats (Schoeb et al.,
1982).
Guinea pigs (10/group) and mice (20/group) were continuously exposed to 14 mg/m
3
(20 ppm) ammonia for up to 6
weeks (Anderson et al., 1964). Separate groups of 6 guinea pigs and 21 chickens were exposed to 35 and 14 mg/m
3
(50
and 20 ppm) ammonia for up to 6 and 12 weeks, respectively. All species displayed pulmonary edema, congestion, and
hemorrhage after 6 weeks exposure, whereas no effects were seen after only 2 weeks. Guinea pigs exposed to 35 mg/m
3
ammonia for 6 weeks exhibited enlarged and congested spleens, congested livers and lungs, and pulmonary edema.
Chickens exposed to 140 mg/m
3
(200 ppm) for 17-21 days showed liver congestion and slight clouding of the cornea.
Anderson and associates also showed that a 72-hour exposure to 14 mg/m
3
ammonia significantly increased the
infection rate of chickens exposed to Newcastle disease virus, while the same effect was observed in chickens exposed
to 35 mg/m
3
for just 48 hours.
Coon et al. (1970) exposed groups of rats (as well as guinea pigs, rabbits, dogs, and monkeys) continuously to ammonia
concentrations ranging from 40 to 470 mg/m
3
. There were no signs of toxicity in 15 rats exposed continuously to 40
mg/m
3
for 114 days or in 48 rats exposed continuously to 127 mg/m
3
for 90 days. Among 49 rats exposed continuously
to 262 mg/m
3
for 90 days, 25% had mild nasal discharge. At 455 mg/m
3
50 of 51 rats died. Thus 127 mg/m3 (179 ppm)
is a subchronic NOAEL for upper respiratory effects in rats. Coon et al. (1970) also found no lung effects in 15 guinea
pigs exposed continuously to 40 mg/m
3
(28 ppm) ammonia for 114 days.
No chronic-duration oral or dermal data were located. Studies by these routes of exposure would provide useful
information on the identification of target organs especially after low-dose exposure.
The INDEX project Final report
222
Summary of long-term exposure effect levels
NOAEL
mg/
LOAEL
mg/
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
6.4
Study
2.3
ADJ
Pulmonary function, eye, skin, and respiratory
symptoms of irritation
occupational, 12.2y
Holness et al., 1989
OEHHA 1999;
REL: 0.2
EPA-IRIS 1991;
RfC: 0.1
17
EXP
1.9
HEC
Increased severity of
rhinitis and pneumonia
with respiratory
lesions
rats, subchronic Broderson et al., 1976 OEHHA 1999
EPA-IRIS 1991
8.7
Study
2.2
ADJ
Pulmonary function, eye, skin, and respiratory
symptoms of irritation, sense of smell
occupational, 12.2y Holness et al., 1989 ATSDR 2002;
MRL: 0.2
Final statement (UNIMI) : Human NOAEL: 2 mg/m³
Study
average concentration ;
EXP
experimental concentration ;
ADJ
concentration adjusted from an intermittent to a continuous exposure ;
1h-ADJ
concentration adjusted to 1-hour exposure duration ;
HEC
human equivalent concentration ;
MLE
maximum likelihood estimate for 5%
response
;
BC05
95% lower confidence limit of the concentration expected to produce a response rate of 5% (Benchmark concentration
approach) ;
STAT
lowest statistically significant effect concentration
OEHHA (1999)
Office of Environmental Health Hazard Assessment of the Californian Environmental Protection Agency
Derivation of Chronic Reference Exposure Level
Study Holness et al., 1989 (supported by Broderson et al., 1976)
Study population 52 workers; 31 controls
Exposure method Occupational inhalation
Critical effects Pulmonary function, eye, skin, and respiratory symptoms of irritation
LOAEL 25 ppm (Broderson et al., 1976) (rats)
NOAEL 9.2 ppm (Holness et al., 1989)
Exposure continuity 8 hours/day (10 m
3/day occupational inhalation rate), 5 days/week
Exposure duration 12.2 years
Average occupational exposure 3 ppm for NOAEL group (9.2 x 10/20 x 5/7)
Human equivalent concentration 3 ppm for NOAEL group
LOAEL uncertainty factor 1
Subchronic uncertainty factor 1
Interspecies uncertainty factor 1
Intraspecies uncertainty factor 10
Cumulative uncertainty factor 10
Inhalation reference exposure level 0.3 ppm ( 300 ppb; 0.2 mg/m
3
; 200 µg/m
3
)
The Holness et al. (1989) study was selected because it was a chronic human study and was published in a respected,
peer-reviewed journal. It is also the only chronic study available. The U.S.EPA (1995) based its RfC of 100 µg/m
3
on
the same study but included a Modifying Factor (MF) of 3 for database deficiencies. The criteria for use of modifying
factors are not well specified by U.S. EPA. Such modifying factors were not used by OEHHA.
For comparison with the proposed REL of 200 µg/m
3
based on human data, OEHHA estimated RELs from 2 animal
studies. (1) Anderson et al. (1964) exposed guinea pigs continuously to 35 mg/m
3
(50 ppm) ammonia for 6 weeks and
observed pulmonary edema. Use of an RGDR of 0.86 and a cumulative uncertainty factor of 3000 (10 for use of a
LOAEL, 10 for subchronic, 3 for interspecies, and 10 for intraspecies) resulted in a REL of 10 µg/m
3
. Staff note that the
nearly maximal total uncertainty factor of 3000 was used in this estimation. (2) Coon et al. (1970) exposed rats
continuously to 127 mg/m
3
ammonia for 90 days and saw no signs of toxicity. Use of an RGDR(ET) of 0.16 for nasal
effects (observed in rats exposed to higher levels of ammonia in Broderson et al. (1976) and a cumulative uncertainty
factor of 100 (3 for subchronic, 3 for interspecies, and 10 for intraspecies) resulted in a REL of 200 µg/m
3
.
Data Strengths and Limitations for Development of the REL: Significant strengths in the ammonia REL include (1) the
availability of long-term human inhalation exposure data (Holness et al., 1989), (2) the demonstration of consistent
effects in experimentally exposed human volunteers following short-term exposures (Ferguson et al., 1977), and (3)
reasonable consistency with animal data (Coon et al., 1970).
The INDEX project Final report
223
Major areas of uncertainty are: (1) the lack of a NOAEL and LOAEL in a single study, (2) a lack of animal data with
chronic exposure and histopathological analyses, and (3) difficulties in estimated human occupational exposures. The
overall database for this common chemical is limited.
ATSDR (2002)
Agency for Toxic Substances and Disease Registry
Derivation of the Minimal Risk Level (MRL):
In the Holness et al. (1989) study, the cohort was also divided into groups that were exposed to low (<4.35 mg/m
3
),
medium (4.35-8.7 mg/m
3
), and high (>8.7 mg/m
3
) ammonia levels and analyzed for change in lung function. Analysis
was performed using each worker’s personal exposure and his change in lung function over the workweek. Differences
due to number of years of ammonia exposure was also assessed. No statistically significant differences were seen
between the level of personal exposure and change in lung function or in lung function between low, medium, and high
exposed groups. No association was evident between years of exposure and lung function changes. The dose and end
point used for MRL derivation are: 8.7 mg/m
3
(NOAEL) for sense of smell, prevalence of respiratory symptoms
(cough, bronchitis, wheeze, dyspnea, and others), eye and throat irritation, and lung function parameters (FVC, FEV1,
FEV1/FVC, FEF50, and FEF75).
The NOAEL was adjusted for continuous exposure as follows: 8.7 mg/m
3
x 8.4/24 hours x 5/7 days = 2.2 mg/m
3
. An
Uncertainty Factors of 10 was used in MRL derivation for human variability: MRL = 0.2 mg/m
3
(0.3 ppm).
The MRL is supported by other observations of respiratory effects associated with chronic-duration exposure including
an association between exposure to pollutants, including ammonia, in livestock confinement buildings and an increase
in respiratory symptoms (such as bronchial reactivity/hyperresponsiveness, inflammation, cough, wheezing, or
shortness of breath) and/or a decrease in lung function (such as forced expiratory volume in the first second [FEV1.0],
maximum expiratory flow rates [MEF50 and MEF75], and maximal mid-expiratory flow rate [MMEF]) in farmers
exposed to ammonia levels of 1.6-14.4 mg/m
3
(Choudat et al. 1994; Cormier et al. 2000; Donham et al. 1995, 2000;
Heederik et al. 1990; Reynolds et al. 1996; Vogelzang et al. 1997, 2000). The farmers were also exposed to other
possible respiratory toxins, such as dust and endotoxins. A cross-sectional study of male workers at two fertilizer
factories in Saudi Arabia showed a significant association between exposure to ammonia gas and respiratory symptoms
and bronchial asthma (Ballal et al. 1998). No continuous exposure levels could be calculated for these workers because
the number of days worked per week was not provided.
USEPA-IRIS (1991)
Integrated Risk Information System
Determination of the Reference Concentration for Chronic Inhalation Exposure (RfC)
Critical effect Exposures* UF MF RfC
Lack of evidence of
decreased pulmonary
function or changes
in subjective
syptomatology
Occupational Study
Holness et al., 1989
NOAEL: 6.4 mg/cu.m (9.2 ppm)
NOAEL(ADJ): 2.3 mg/cu.m
NOAEL(HEC): 2.3 mg/cu.m
LOAEL: None
30 1 0.1 mg/m
3
Increased severity of
rhinitis and pneumonia
with respiratory
lesions
Rat Subchronic
Inhalation Study
Broderson et al., 1976
NOAEL: None
LOAEL: 17.4 mg/cu.m (25 ppm)
LOAEL(ADJ): 17.4 mg/cu.m
LOAEL(HEC): 1.9 mg/cu.m
*Conversion Factors: MW = 17.03 Holness et al., 1989: Assuming 25C and 760 mm Hg, NOAEL (mg/cu.m) = 9.2 ppm x 17.03/24.45 =
6.4 mg/cu.m. The NOAEL is based on an 8-hour TWA occupational exposure. MVho = 10 cu.m/day, MVh = 20 cu.m/day. NOAEL(ADJ)
= 6.4 mg/cu.m x (MVho/MVh) x 5 days/7 days = 2.3 mg/cu.m.
Broderson et al., 1976: Assuming 25C and 760 mm Hg, the LOAEL (mg/cu.m) = 25 ppm x 17.03/24.45 = 17.4 mg/cu.m. The
LOAEL(HEC) was calculated for a gas:respiratory effect in the ExtraThoracic region. MVa = 0.14 cu.m/day, MVh = 20 cu.m/day, Sa(ET)
= 11.6 sq. cm., Sh(ET) = 177 sq. cm. RGDR(ET) = (MVa/Sa) / (MVh/Sh) = 0.1068. NOAEL(HEC) = 17.4 x RGDR = 1.9 mg/cu.m.
The INDEX project Final report
224
The use of Holness et al. (1989) as the principal study can only be supported in the context of the data array. It is not
surprising that no effects were seen on screening spirometry since the exposure levels were low. Comparing the 9.2
TWA of Holness et al. (1989) with other data on the respiratory effects of ammonia, a trend is observed that at lower
concentrations the extrathoracic region of the respiratory system is affected due to the chemical's solubility and
reactivity; while at higher concentrations, the lower part of the respiratory system is involved in both experimental
animals (Dahlman, 1956; Gamble and Clough, 1976) and humans (Flury et al., 1983). Thus, no effects were observed in
the lower respiratory system as reflected by pulmonary function. Pulmonary function may not be a particularly sensitive
test because exposure to this type of agent at low concentrations is not expected to result in significant exposure of the
lower respiratory region. No objective investigation of the workers' nasal epithelium was performed and the complaint
of exacerbated upper respiratory symptoms suggests sensory irritation and supports the extrathoracic region as the
critical region for an effect. The possibility of selection bias against atopic predispositions in the population is suggested
by the significantly lower prevalence of hay fever in the exposed versus control cohort. Thus, there is a concentration-
response in the extrathoracic region in experimental animals beginning at a LOAEL at essentially the same HEC as the
NOAEL in Holness et al. (1989) and the NOAEL may be based on a less sensitive endpoint. Also the apparent
discrepancy of a lower LOAEL(HEC) from Broderson et al. (1976) and the identified NOAEL(HEC) of the Holness et
al. (1989) study may be the result of differences in air flow patterns since rats are obligate nose- breathers and humans
breathe oronasally. The use of the NOAEL from Holness et al. (1989) can be supported as marginal in this context due
to the symptomatology complaints and because human data engenders less uncertainty than extrapolation from the
experimental animal data.
Confidence in the Inhalation RfC: Confidence in the principal study is medium. Although a relatively small sample size
(males only) was studied and a free standing NOAEL was determined, mild extrathoracic effects were observed in rats
near the same HEC as reported in the Holness study. Additional human subchronic and acute studies support the
NOAEL. Confidence in the data base is medium to high. Although developmental, reproductive or chronic toxicity
following ammonia exposure has not been adequately tested, pharmacokinetic data suggests systemic distribution at the
HEC level is unlikely. Reflecting medium confidence in the principal studies and medium to high confidence in the data
base, confidence in the RfD is medium.
Reproductive, mutagenic and carcinogenic effects
Toxicological effects, such as mutagenic, reproductive, or carcinogenic effects, would not be expected from exposures
insufficient to cause local effects. There is no evidence that ammonia is carcinogenic, though it can produce
inflammatory lesions of the colon and cellular proliferation, which could increase susceptibility to malignant change.
There was no evidence that ammonia was responsible for the increased incidence of tumours with increased dietary
protein intake. Ammonia did not either cause tumours or increase the spontaneous incidence of tumours in life-time
studies on mice.
Interactions with other chemicals
Exposure to substances that would increase the pH of exposed tissues could be expected to enhance the alkalotic effects
of ammonia, and vice versa.
Data regarding exposure to mixtures of atmospheric contaminants indicate that, contrary to what might be expected,
increased carbon dioxide concentration (up to 5% in air) does not alter the hyperventilatory rate induced by
hyperammonemia in dogs (Herrera and Kazemi 1980). Ammonia in expired air may neutralize inhaled acid aerosols
(EPA 1979; Larson et al. 1980; Utell et al. 1989).
Odour perception
Odour is characterized as sharp, pungent and intensely irritating.
Source: Devos et al., (1990)
Odour threshold: 1 mg/m
3
Source: Amoore and Hautala (1983)
Odor threshold: 17 mg/m
3
(25 ppm)
The INDEX project Final report
225
Source: Leonardos et al. (1969)
Recognition threshold: 33 mg/m
3
(47 ppm)
The considerable variability in threshold data prompted work by Leonardos et al. (1969), who used a standardized
procedure to determine recognition thresholds rather than detection thresholds for 53 chemicals. The odour threshold
was defined as the first concentration at which all four panel members (trained odour analysts) were able to recognize
the characteristic odour of the chemical. The panel tested only one chemical per day. Concentrations examined were
multiples by 10 of 0.7, 1.47, and 3.3 mg/m
3
(1, 2.1, and 4.6 ppm). The recognition threshold for the odour of ammonia
was 32.6 mg/m
3
(46.8 ppm) (WHO, 1986).
Source: AIHA (1989)
Reported odour threshold values range from 0.03 to 37.5 mg/m
3
(0.041 to 53 ppm) with a geometric mean of 11.8
mg/m
3
(17 ppm)
Source: WHO (1986)
Estimates of odor thresholds for ammonia vary from 0.04-103 ppm (0.03-73 mg/m³) (Ferguson et al., 1977; Henderson
and Haggard, 1943; Ruth, 1986). Near the odor threshold, persons exposed to ammonia can experience annoyance and
believe the odor to be a nuisance.
Summary of Ammonia Dose Response Assessment
________________________________________________________________________________________________
Exposure other than inhalation: Endogenous ammonia (dietary proteins) used for protein synthesis and as a buffer,
rapid conversion of excesses to glutamine/urea (liver) and urinary excretion.
Toxicokinetics: At ambient air levels (<350 mg/m
3
), almost completely (83-92%) dissolved (NH
4
OH) in the upper
respiratory tract (nasal and pharyngeal, but not tracheal irritation), reduced absorption at prolonged exposure (>10min,
>350 mg/m
3
) due to saturation. Low absorption into the blood (systemic circulation), elimination mainly through
expired air at the end of exposure. Only small proportion absorbed across the nasopharyngeal membranes.
Health effect levels of short- and long-term exposure
NOAEL
mg/m³
LOAEL
mg/m³
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
Short-term exposure
14.0
1h-ADJ-MLE
9.5
BC05
eye and respiratory irritation volunteers,
1h-adjusted
Benchmark approach
Industrial Biotest
Laboratories, 1973;
MacEwen et al., 1970;
Silverman et al., 1949;
Verberk, 1977
OEHHA 1999;
REL: 3.2
35
EXP
irritation to nose and throat; urge to cough volunteers, 2h Verberk, 1977 ATSDR 2002;
MRL: 1.2
General remark: No study with asthmatics!
Long-term exposure
6.4
Study
2.3
ADJ
Pulmonary function, eye, skin, and respiratory
symptoms of irritation
occupational, 12.2y
Holness et al., 1989
OEHHA 1999;
REL: 0.2
EPA-IRIS 1991;
RfC: 0.1
17
EXP
1.9
HEC
Increased severity of rhinitis and pneumonia
with respiratory lesions
rats, subchronic Broderson et al., 1976 OEHHA 1999
EPA-IRIS 1991
8.7
Study
2.2
ADJ
Pulmonary function, eye, skin, and respiratory
symptoms of irritation, sense of smell
occupational, 12.2y Holness et al., 1989 ATSDR 2002;
MRL: 0.2
Carcinogenicity: No evidence in life-time studies on mice. Mutagenic or carcinogenic effects would not be expected
from exposures insufficient to cause local effects (inflammatory lesions and cell proliferation).
Genotoxicity: No evidence
Odour threshold: Range: 0.03-38 mg/m
3
, with geometric mean: 12 mg/m
3
(AIHA) and recognition: 33 mg/m
3
; 1
mg/m
3
(Devos); Coughing reflex and lachrymation starting from 35 mg/m
3
, immediate coughing at 700 mg/m
3
Susceptible population: Exposure to ammonia result in an exacerbation of preexisting asthma (48/60 asthmatics
claimed following exposure to household cleaners containing ammonia). Wide variability of symptoms depending on
recent history of ammonia exposure; tolerances up to 70 mg/m
3
after weekly inurement period.
Remarks: RD
50
: 211 mg/m
3
The INDEX project Final report
226
________________________________________________________________________________________________
6. Risk Characterization
Health hazard evaluation of short-term exposure
Toxicological endpoint: Irritation to nose and throat; urge to cough
Derivation of a limit of exposure (EL)
LOAEL
a
LOAEL
10
b
LOAEL
100
c
LOAEL
300
d
35 mg/m
3
3.5 mg/m
3
0.35 mg/m
3
0.1 mg/m
3
a
Human LOAEL ;
b
not considering a NOAEL (10);
c
intraspecies variability
(10) ;
d
susceptible population (3)
Odour threshold: Range: 0.03-38 mg/m
3
, with geometric mean: 12 mg/m
3
(AIHA) and recognition: 33 mg/m
3
; 1 mg/m
3
(Devos); Coughing reflex and lachrymation starting from 35 mg/m
3
, immediate coughing at 700 mg/m
3
Health hazard evaluation of long-term exposure
Toxicological endpoint: Pulmonary function, eye, skin, and respiratory symptoms of irritation
Relevance of EU-population exposure to Ammonia
Non population-based study
NOAEL
a
NOAEL
10
b
NOAEL
30
c
Margin of
safety (MOS)
Description (Study, Year) N 2 mg/m
3
0.2 mg/m
3
0.07 mg/m
3
50
th
(90
th
)
Helsinki (VTT 2003)
Homes without known IAQ problems
< < < 3.0 (1.8)
Helsinki (VTT 2003)
Homes with known IAQ problems
< < < 2.6 (1.3)
a
Occupational LOAEL, adjusted for continuous exposure;
b
intraspecies variability (10);
c
susceptible population;
< out of the evaluation range (i.e. <5% of the environments investigated)
Result
There is a lack of knowledge concerning indoor concentrations and exposures of ammonia. Exposure data are limited
on only one non population-based study describing concentrations of ammonia in Finnish homes with and without
known indoor air quality (IAQ) problems. In both cases measured concentrations were within the same order of
magnitude with both exposure limits here esteblished for short- and long-term effects (70 and 100 µg/m
3
, respectively)
relating on irritative effects and pulmonary funtions and taking into account the particular susceptibility of asthmatic
subjects. It is assumed that exposure concentrations in the order of the short-term EL could easily be attained during
domestic activities making use of ammonia containing household products.
The INDEX project Final report
227
Limonene
Synonyms: 1-methyl-4-(1-methylethenyl)cyclohexene
CAS Registry Numbers: 138-86-3
Molecular Formula: C
10
H
16
1. Compound identification
Limonene exists as two optical isomers, d-limonene (pleasing orange scent) and l-limonene (smells piney, turpentine
smell; mixture of both isomers: dipentene), and is used as a flavour and fragrance additive in food, household cleaning
products and perfumes or could be contained in solvents (e.g. turpentine).
Limonene is both a naturally occurring and a synthetic colourless mobile liquid, which is used in many food products,
for its characteristic lemon-like flavour and odour. It is also used as a solvent, wetting agent, in resins, and as a
monomer and copolymer. It may be emitted to indoor air also from cats and dogs repellent spray and shampoo, pet
sleeping quarters, household dwellings, indoor premises, and human clothing. Limonene emissions have been detected
in the industries such as extraction of pine gum, paper and pulp mills, plastics materials-synthetic resins, perfumes,
cosmetics and other toilet preparations, organic solvents and lubricating oils and greases (HSDB 2003).
Limonene may be emitted to household environments from furniture polishes and room fresheners. Occupational
exposure to limonene may occur by inhalation or dermal contact during its production, formulation, transport or use.
The main routes of human exposure to limonene in the general population is assessed to be inhalation of limonene and
ingestion of food in which it occurs naturally or to which it has been added as a flavour or fragrance (EPA 1994, HSDB
2003).
2. Physical and Chemical properties
Molecular weight (g/mol) 136.23
Melting point (°C) -74
Boiling point (°C) 176
Density (at 20 °C, 1 atm) 0.8402
Relative density (air =1) 4.7
Solubility: Miscible with alcohol and ether
Conversion factors at 20 °C and 760 mm Hg:
1 ppb = 5.654 µg/m
3
1 µg/m
3
= 0.177 ppb
Sources: Verschueren 2001, HSDB 2003
3. Indoor Air Exposure assessment
Contribution of inhalation exposure to total exposure
Food is the principal source of exposure to limonene (96%), the contribution from ambient air being approximately 4%
(WHO, 1998). d-limonene is generally recognized as safe in food by the Food and Drug Administration. Based on daily
US consumption of d-limonene per capita, the intake of d-limonene from food for the general population was estimated
to be 0.27 mg/kg body weight per day (Flavor and Extract Manufacturers Association, 1991). The intake of limonene
from indoor and outdoor air for the general population is estimated to be 10 and 0.1 µg/kg body weight per day,
The INDEX project Final report
228
respectively. This is based on the daily inhalation volume for adults of 22 m
3
, a mean body weight for males and
females of 64 kg, the assumption that 4 of 24 hours are spent outdoors (IPCS, 1994), and arithmetic mean limonene
levels in indoor and outdoor air of 0.04 and 0.002 mg/m
3
, respectively, in a study from Los Angeles (Wallace et al.,
1991).
Terpene/ozone reaction products
It has been proposed that reactions between unsaturated volatile compounds (e.g. limonene, α-pinene, styrene) and
ozone or hydroxyl (OH) radicals produce chemically reactive products more likely to be responsible for eye and airway
irritation than the chemically non-reactive VOCs usually measured indoors. Many of the known reaction products are
oxygenates (e.g. formaldehyde and other aldehydes, carboxylic acids, peroxides) which may cause irritation at low
concentrations or odor annoyance due to their usually low odour thresholds (Weschler and Shields, 1997; Wolkoff et
al., 1997; 2000). Ozone enters the indoor environment (<0.3 mg/m3) by infiltration of the outdoor air (<0.6 mg/m3) or
could be produced by photocopiers and certain air cleaners.
Indoor air and exposure concentrations
Residential indoor concentrations of limonene were clearly higher than respectively outdoor concentrations in all
regions of Europe based on EXPOLIS results (Table 4.0.1, Jantunen et al 1999). Average indoor concentrations were
lowest in Central Europe and highest in Athens (Figure 3.1, Annex 1). Personal exposures to limonene, ranging from 19
µg/m
3
to 56 µg/m
3,
were lower than residential indoor concentrations (Jantunen et al 1999, Hoffman et al 2000).
Kostiainen et al (1995) reported clearly lower mean and median indoor concentrations of limonene in Helsinki, Finland,
14.2 µg/m
3
and 8.8 µg/m
3
respectively. Maroni et al (1995) reported typical limonene concentrations in residential
indoor environments in Europe and in the United States, being on average 30 µg/m
3
. Indoor concentrations measured in
offices in Milan, Italy were about half of the residential levels, being on average 17 µg/m
3
. Brown (2002) studied
limonene concentrations in new and established buildings. Limonene concentrations in a new building increased after
construction, being 12 µg/m
3
, 15 µg/m
3
, and 34 µg/m
3
2, 19, and 72 days after construction respectively. This
decreasing trend suggested the dominant source of limonene being human activities, possibly consumer products, rather
than building materials.
TEAM study carried out in Los Angeles, the USA showed a maximum 12-hour day time exposure of 990 µg/m
3
to
limonene (Wallace et al 1991). Respective residential indoor and outdoor concentrations showed concentrations of 230
µg/m
3
and 10 µg/m
3
. Maximum exposure concentration in California showed higher value than in European studies.
Maximum residential indoor concentration was in the range with European maximum levels.
WHO (1989) has assessed indoor air concentrations of VOCs in “typical homes” giving median and 90
th
percentile
concentrations of 15 µg/m
3
and 70 µg/m
3
for limonene. Brown et al (1994) calculated weighted geometric mean
(WAGM) indoor concentrations for several building types using mean values collected from the studies carried out
worldwide. Also they reported lower indoor values, WAGM of 21 µg/m
3
and 90
th
percentile of 85 µg/m
3
for limonene
than found in European population based studies.
Residential indoor air concentrations of limonene in European urban populations measured in the EXPOLIS study and
the National Survey in England are presented in Figure 3.1 and Figure 3.2. Respective exposure concentrations are
presented in Figure 3.3.
The INDEX project Final report
229
0
20
40
60
80
100
0 50 100 150 200 250
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
Hel
Mil
Oxf
Pra
Figure 3.1. Cumulative frequency distributions of indoor air concentrations of limonene in Athens (Ath, n= 42), Basel
(Bas, n= 47), Helsinki (Hel, n= 188), Milan (Mil, n= 41) Oxford (Oxf, n= 40) and Prague (Pra, n= 46) (EXPOLIS
2002).
0
20
40
60
80
100
0 1020304050
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Limonene 28-day
Figure 3.2. Cumulative frequency distribution of 28-day indoor air concentrations of limonene in UK (GM= 6.2 µg/m
3
,
max 308 µg/m
3
, n=796, Brown et al 2002).
The INDEX project Final report
230
0
20
40
60
80
100
0 50 100 150 200 250
Exposure (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
GeS
Hel
Oxf
Pra
Figure 3.3. Cumulative frequency distributions of 48-hour personal exposure concentrations of limonene in Athens
(Ath), Basel (Bas), Helsinki (Hel), Oxford (Oxf) and Prague (Pra) (EXPOLIS 2002), and 1-week mean exposures of the
German Survey GerES II (GeS) (Hoffman et al 2000).
4. Toxicokinetics
Absorption
Limonene has a high partition coefficient between blood and air (λ
blood/air
= 42) and is easily taken up in the blood at the
alveolus (Falk et al., 1990). The net uptake of limonene in volunteers exposed to the chemical at concentrations of 450,
225, and 10 mg/m
3
for 2 hours during light physical exercise averaged 65% (Falk Filipsson et al., 1993). Orally
administered limonene is rapidly and almost completely taken up from the gastrointestinal tract in humans as well as in
animals (Igimi et al., 1974; Kodama et al., 1976).
Distribution
Limonene is rapidly distributed to different tissues in the body and is readily metabolized. Clearance from the blood
was 1.1 litre/kg body weight per hour in males exposed for 2 hours to d-limonene at 450 mg/m
3
(Falk Filipsson et al.,
1993). A high oil/blood partition coefficient and a long half-life during the slow elimination phase suggest high affinity
to adipose tissues (Falk et al., 1990; Falk Filipsson et al., 1993).
In rats, the tissue distribution of radioactivity was initially high in the liver, kidneys, and blood after the oral
administration of [
14
C] d-limonene (Igimi et al., 1974); however, negligible amounts of radioactivity were found after
48 hours. Differences between species regarding the renal disposition and protein binding of d-limonene have been
observed.
Metabolism and elimination
The biotransformation of d-limonene has been studied in many species, with several possible pathways of metabolism
(see Figure 4.1). Metabolic differences between species have been observed with respect to the metabolites present in
both plasma and urine. About 25-30% of an oral dose of d-limonene in humans was found in urine as d-limonene-8,9-
diol and its glucuronide; about 7-11% was eliminated as perillic acid (4-(1-methylethenyl)-1-cyclohexene-1-carboxylic
acid) and its metabolites (Smith et al., 1969; Kodama et al., 1976). d-Limonene-8,9-diol is probably formed via d-
limonene-8,9-epoxide (Kodama et al., 1976; Watabe et al., 1981). In another study, perillic acid was reported to be the
principal metabolite in plasma in both rats and humans (Crowell et al., 1992). Other reported pathways of limonene
metabolism involve ring hydroxylation and oxidation of the methyl group (Kodama et al., 1976).
The INDEX project Final report
231
Following the inhalation exposure of volunteers to d-limonene at 450 mg/m
3
for 2 hours, three phases of elimination
were observed in the blood, with half-lives of about 3, 33, and 750 minutes, respectively (Falk Filipsson et al., 1993).
About 1% of the amount taken up was eliminated unchanged in exhaled air, whereas about 0.003% was eliminated
unchanged in the urine. When male volunteers were administered (per os) 1.6 g [
14
C] d-limonene, 50-80% of the
radioactivity was eliminated in the urine within 2 days (Kodama et al., 1976). Limonene has been detected, but not
quantified, in breast milk of non-occupationally exposed mothers (Pellizzari et al., 1982).
Figure 4.1: Major pathways for d-limonene metabolism (Source: WHO 1998, from Kodama et al., 1976)
5. Health effects
Effects of short-term exposure
In general, limonene could be considered, with the exception of its irritative (skin, eyes) and sensitizing properties, to be
a chemical with fairly low acute toxicity (WHO, 1998). Potential hazard to the general population are skin irritancy
and sensitisation from use of consumer products, varying with the concentration of limonene in the product and, for
sensitisation, with its oxidation status (addition of oxidised limonene to the list of substances used in allergy testing has
been recommended)..
None of eight volunteers reported any discomfort, irritation, or symptoms related to central nervous system effects
during a 2-hour inhalation exposure in an exposure chamber (work load 50 W) to d-limonene at 10, 225, or 450 mg/m
3
;
however, a slight decline in vital capacity was observed following exposure to the highest concentration (Falk Filipsson
et al., 1993).
Eye irritation of l-limonene was measured using goggles instrumentation and a relatively short exposure time of 2
minutes. The threshold level for irritation in 12 volunteers was 1700-3400 mg/m
3
(Mølhave et al., 2000).
d-Limonene infused directly into the bile system of human volunteers to dissolve gallstones caused pain in the upper
abdomen, nausea, vomiting, and diarrhoea, as well as increases in serum aminotransferases and alkaline phosphatase
(Igimi et al., 1976, 1991). The oral administration of 20 g d-limonene to volunteers resulted in diarrhoea, painful
constrictions, and proteinuria, but no biochemical changes (total protein, bilirubin, cholesterol, aspartate
aminotransferase, alanine aminotransferase, alkaline phosphatase) in the liver (Igimi et al., 1976).
The INDEX project Final report
232
It has recently been proposed that reactions between limonene (or other unsaturated volatile compounds) and ozone or
reactive radicals produce chemically reactive products more likely to be responsible for eye and airway irritation than
the chemically non-reactive VOCs usually measured indoors (Weschler and Shields, 1997; Wolkoff et al., 1997; 2000).
Reaction mixtures of excess alpha-pinene, limonene and isoprene with ozone considerably below their NOEL
concentrations resulted in significant upper airway irritation (Wolkoff et al, 2000). The reduction of the respiratory rate
was from 30% to about 50%. Chemical analysis of reaction mixtures by conventional methods showed that readily
identified stable products and residual reactants at the concentrations found could not account for the observed
reductions of the respiratory rate, assuming additivity of the reaction products. The results suggest that, in addition to
known irritants (formaldehyde, acrolein, methacrolein, methyl vinylketone), one or more strong airway irritant(s) of
unknown structure(s) were formed (Wolkoff et al, 2000). Effects of variation of reaction time, relative humidity and
initial ozone concentration on irritant formation were described by Wilkins et al. (2003).
Kleno and Wolkoff (2004) exposed 8 male subjects for 20 min to limonene oxidation products (LOPs) and measured
relative changes in blink frequencies. The subjects were exposed locally in the non-dominant eye and single blind in
random order. Blinking was video-recorded and evaluated for full sessions of 36 min while the subjects viewed an
educational film. The mean blink frequency increased significantly at lower ppb levels of LOPs, 42% ( P<0.0001),
compared with that at baseline. Neither the residual reactants nor clean air changed the blink frequency significantly.
The findings coincided with qualitative reporting of weak eye irritation symptoms (Kleno and Wolkoff, 2004).
Irritative effects in experimental animals exposed to oxidation products of ozone and unsaturated hydrocarbons,
including limonene, were recently described in Wolkoff et al. (2000), Clausen et al. (2001), Rohr et al. (2002), Wilkins
et al. (2003).
Oxidised Histo-clear solvent (used in some pathology laboratories, containing d-limonene as a replacement for xylene)
produced respiratory irritation requiring medical treatment in a hystology technician (NICNAS, 2002). It was
considered that the autoxidation products might be the cause of the irritation. An increase in nasal or throat irritation
was also reported by 4/12 workers in a facility were a limonene-based cleaner was stored in an uncovered tank
(NICNAS, 2002).
The acute toxicity of d-limonene in rodents is fairly low after oral, intraperitoneal, subcutaneous, and intravenous
administration, based on the magnitude of the LD
50
values. LD
50
values were approximately 5 g/kg body weight for the
oral administration of d-limonene or d/l-limonene to rats and for dermal application of d/l-limonene to rabbits and 6
g/kg body weight for oral administration to mice (Tsuji et al., 1974, 1975b; Opdyke, 1978).
Effects observed following the acute exposure of rodents to limonene include increased bile flow at 85 mg/kg body
weight (Kodama et al., 1976), inhibition of S-3-hydroxy-3-methylglutaryl-CoA reductase activity at 409 mg/kg body
weight (Clegg et al., 1980), enzyme induction at 600 and 1200 mg/kg body weight (Ariyoshi et al., 1975), and
decreased motor activity, hypothermia, and potentiation of hexobarbital-induced sleep at 3 ml/kg body weight (Tsuji et
al., 1974).
Studies in guinea-pigs have revealed that air-oxidized d-limonene, but not d-limonene itself, induced contact allergy
(Karlberg et al., 1992).
Summary of short-term exposure effect levels
NOAEL
mg/m
3
LOAEL
mg/m
3
Target system;
Critical effect studied
Remarks Study Source
(Organization)
225 450 CNS related symptoms, irritation,
Endpoint: decline in vital capacity
Volunteers, 2h - work load 50W Falk Filipsson et al., 1993
Effects of long-term exposure
No information is available on the chronic health effects of inhalation exposure to d-limonene in humans, and no long-
term inhalation studies have been conducted in laboratory animals. Limonene/ozone reaction products have not yet been
submitted to chronic toxicity studies.
The INDEX project Final report
233
In numerous experimental studies on animals, oral exposure (gavage) to limonene has been shown to affect the liver.
Exposure affects the amount and activity of liver enzymes, liver weight, cholesterol levels and bile flow. However, no
toxic effects on the liver have been reported.The liver effects in animals are thought to be due to physiological
adaptation. Owing to a lack of data on d-limonene exposure in humans, this organ cannot with certainty be stated as the
critical organ in humans. From the data available, it is not possible to identify a NOAEL for these effects.
The only microscopic evidence of compound-related toxicity noted in rats was nephropathy in the males. d-Limonene is
one of a diverse group of hydrocarbons that has been shown to induce a unique syndrome of nephropathy in male rats
following subchronic or chronic exposure. Based on a review of the literature concerning this effect (U.S. EPA, 1991),
EPA's Risk Assessment Forum concluded that nephropathy in male rats that is associated with alpha- 2u-globulin
accumulation in hyaline droplets is not an appropriate endpoint to determine noncancer effects potentially occurring in
humans (U.S. EPA, 1993).
In two subchronic oral exposure studies in rats, the following effect levels were identified. The NOEL, based upon
histopatological examination of the kidneys, was considered to be 5 mg/kg bw/d. The LOEL for increased liver and
kidney weight was 75 mg/kg bw/d. For effects in the liver the NOEL was 10 mg/kg bw/d and the LOAEL was 30
mg/kg bw/d (Ariyoshi et al., 1975; Webb et al., 1989).
Based on available data, food is believed to be the principal source of exposure (96%) to limonene; the contribution
from ambient air is approximately 4%. To calculate a tolerable intake for humans, the animal study was chosen in
which effects on the liver were observed at the lowest exposure level (Webb et al., 1989). In this study, gavage
administration of d-limonene (5 days/week for 13 weeks) to rats caused increased relative liver weight at 30 and 75
mg/kg body weight per day. The NOEL for the liver was considered to be 10 mg/kg body weight per day. Using
uncertainty factors of 10 for intraspecies differences and 10 for interspecies differences, a tolerable intake for ingestion
of d-limonene by humans of 0.1 mg/kg body weight per day may be calculated from the NOEL. This value is of a
similar magnitude as the estimated daily US consumption of d-limonene of 0.27 mg/kg body weight per day (Flavor
and Extract Manufacturers Association, 1991). U.S. FDA approves d-limonene for use as a food additive.
Matthys et al. (2000) observed that “Myrtol standardized” (Gelomyrtol forte), a phytotherapeutic extract (distillate)
consisting mainly of three monoterpenes: d-limonene, (+)alpha-pinene, and 1,8-cineole, is a well-evidenced alternative
to antibiotics for acute bronchitis treatment and was considered safe and tolerable in 170 patients at daily doses of 4 x
300 mg for 2 weeks.
Occupational exposure standards for limonene are summarised in Table 5.1. Supporting documentation for the exposure
standard adopted by the American Industrial Hygiene Association (AIHA) states that the 167 mg/m
3
(30 ppm) 8-hr
TWA was set to protect against liver effects seen in male mice and reduced survival in female rats in a 2-yr NTP study
(AIHA, 1993). In Germany no MAK (maximum workplace concentration) has been set for limonene as it was
considered that insufficient information was available.
Table 5.1: National occupational exposure standards for limonene
Country 8-h TWA
mg/m
3
STEL
mg/m
3
Year adopted Reference
Norway 140 - 1999 RTECS, 2001
Sweden 140 280 1990 Karlberg and Lindell, 1993
Finland 140 280 not known Finnish Institute of Occupational Health, 2001
AIHA 167 - 1993 AIHA, 1993
TWA: time-weighted average
STEL: short-term (15 min) exposure limit
U.S. EPA deemed the database at the time of review (1993) to be insufficient to derive an inhalation RfC according to
the Interim Methods for Development of Inhalation Reference Concentrations (U.S. EPA, 1990).
Carcinogenic and genotoxic effects
There is inadequate evidence in humans for the carcinogenicity of d-limonene.
There is sufficient evidence in experimental animals for the carcinogenicity of d-limonene. Overall evaluation: In
making its overall evaluation of the carcinogenicity to humans of d-limonene, the IARC Working Group concluded that
d-limonene produces renal tubular tumors in male rats by a non-DNA reactive alpha-2-globulin associated response.
Therefore, the mechanism by which d-limonene increases the incidence of renal tubular tumors in male rats is not
The INDEX project Final report
234
relevant to humans. d-Limonene is not classifiable as to its carcinogenicity to humans (Group 3) (IARC, 1999).
The few available data indicate that d-limonene and its 1,2-epoxide metabolite are not genotoxic. Limonene
administered in the diet was not mutagenic in the liver or kidney of male Big Blue rats Turner et. al. (2001).
Interactions with other chemicals
The formation of unidentified strong upper airway irritants in reaction mixtures of terpenes and ozone has been recently
reported. Identified products included formaldehyde and other aldehydes, carboxylic acids, peroxides, which may cause
irritation at low concentrations or odor annoyance due to their usually low odour thresholds (Weschler and Shields,
1997; Wolkoff et al., 1997; 2000).
Maximum irritation of reaction mixtures of limonene and ozone, measured in mice, was observed at low humidity (<2%
RH) and short time (16-30 s) reaction mixtures. Moderate humidity (approximately 32% RH) and longer reaction times
(60-90 s) resulted in significantly less irritation, suggesting that some unidentified intermediates react with water vapor
to give less irritating products (Wilkins et al., 2003). Irritation measured at four ozone concentrations (1, 2, 4 and 7
mg/m
3
) using low humidity/short time reaction conditions for limonene (280 mg/m
3
) and isoprene (1400 mg/m
3
)
revealed that at 1 mg/m
3
, irritation was at the same level as that for the pure terpenes, indicating that at 1 mg/m
3
ozone
the combined irritant effect was near the no effect level for the product mixture (Wilkins et al., 2003).
Odour perception
Source: Leffingwell & Associates
d-limonene - fresh citrus, orange-like - odour perception : 1.1 mg/m
3
(200 ppb)
l-limonene - harsh, turpentine-like, lemon note - odour perception : 2.8 mg/m
3
(500 ppb)
Summary of Limonene Dose Response Assessment
________________________________________________________________________________________________
Exposure other than inhalation: Oral: About 96% of limonene absorbed by humans originate from food. Generally
recognized as safe in food by U.S.FDA. Dermal: Potential hazard to the general population may derive from skin
irritancy and sensitisation from use of consumer products For contact allergy, air-oxidized limonene seems to play a
major role than limonene itself.
Toxicokinetics: ~65% net uptake of the inhaled amount, rapid distribution and high affinity for adipose tissue
Health effect levels of short- and long-term exposure
NOAEL
mg/m³
LOAEL
mg/m³
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
Short-term exposure
225 450 CNS related symptoms, irritation, Endpoint:
decline in vital capacity
Volunteers, 2h - work
load 50W
Falk Filipsson et al.,
1993
Long-term exposure (no studies have been found in literature)
Carcinogenicity: Inadequate evidence in humans; sufficient evidence in animals, with mechanism not relevant to
humans.
Genotoxicity: The few available data indicate that d-limonene and its 1,2-epoxide metabolite are not genotoxic.
Odour threshold: 2.15 mg/m
3
(Devos); d-limonene (orange-like): 1.1 mg/m
3
; l-limonene (turpentine-like) : 2.8 mg/m
3
(Leffingwell & Associates)
Susceptible population:
Remarks: RD
50
: 6 g/m
3
; In addition to known irritant reaction products (formaldehyde, acrolein, methacrolein, methyl
vinylketone), one or more strong airway irritant(s) of unknown structure(s) could be formed following the reaction
between limonene and ozone or reactive radicals.
The INDEX project Final report
235
6. Risk Characterization
Health hazard evaluation of long-term exposure
Effect level - mg/m
3
Assessment
factor
EL
mg/m
3
Toxicological endpoint
Long-term
Exposure Limit
Human
LOAEL
Volunteers
450 1000
abc
0.45
CNS related symptoms, irritation, Endpoint:
decline in vital capacity
a
extrapolation from subacute to chronic exposure (10);
b
not considering a NOAEL (10)
c
intraspecies variability (10);
Relevance of EU-population exposure to limonene
Population based studies
EL
derived
Margin of
safety
(MOS)
Description (Study, Year) N 0.45 mg/m
3
50
th
(90
th
)
Athens (Expolis, 96-98) 42 < 11 (3)
Basel (Expolis, 96-98) 47 < 49 (14)
Helsinki (Expolis, 96-98) 188 < 33 (6)
Milan (Expolis, 96-98) 41 < 14 (3)
Oxford (Expolis, 98-00) 40 < 43 (11)
Prague (Expolis, 96-98) 46 < 23 (8)
England 28d-TWA (BRE, 97-99) 796 < 56 (11)
Avg MOS: 33 (8)
< out of the evaluation range (i.e. <5% of the environments investigated)
Result
An attempt has been done in deriving an exposure limit (EL) for long-term effects associated with limonene exposure
by refering to a study on volunteers exposed at sub-acute (2 hours) inhalation doses. When comparing this EL (450
µg/m
3
) with the results from seven indoor surveys it is concluded that no neurologic effects would be expected at
background limonene levels encountered in European homes, with median (90
th
percentile) levels at least 10 (3) times
lower than the proposed EL. It is assumed that at 10-fold the level set as the EL, irritation could be expected following
acute exposure. Due to its widespread use as a flavouring agent in numerous consumer products, short-term exposures
at levels in the order of some mg/m
3
could not be excluded, although significative exposure data are lacking.
The INDEX project Final report
236
alpha-Pinene
Synonyms: 2,6,6-Trimethylbicyclo(3.1.1)-2-hept-2-ene
2,6,6-Trimethylbicyclo(3.1.1)hept-2-ene
2-Pinene
CAS Registry Numbers: 80-56-8
Molecular Formula: C
10
H
16
1. Compound identification
Alfa-pinene is a colourless liquid with a characteristic odour of pine. It is a naturally occurring terpene, which is emitted
by trees, fruits, grasses, bushes, fungi, herbs, and flowers and it is a constituent of many common oils. Alfa-pinene will
exist solely as a vapour in the atmosphere. It is used in aerosol paint concentrates, cleaning and sanitation products,
paints and varnish removers, waterproofing compounds solvent, and flavouring. It is also emitted to indoor air from
commonly used wooden furniture and waxes (HSDB 2003, Maroni et al 1995).
Alpha and beta pinene (C
10
H
16
) are principal constituents of turpentine. Turpentine is a thin volatile essential oil
(C
10
H
16
) obtained by steam distillation from the wood or the exudate of pine trees. Main uses of turpentine include as a
solvent thinner for paint, varnishes, and lacquer, and as a vehicle for paints (professional painters, private individuals).
Turpentine is also used in perfumery, sprays, and deodorizers. The composition of wood turpentine varies with species,
location, and season. Steam-distilled (wood) turpentine produced in the United States is made up primarily of
alphapinene (75 to 85%) with varying amounts of beta-pinene (up to 3%), camphene (4 to 15%), limonene (dipentene, 5
to 15%), 3-carene, and terpinolene (percentages not provided) (Chinn, 1989). In a Swedish study (Falk Filipsson, 1996)
volunteers were exposed to turpentine vapour with a composition measured in air: 54% alpha-pinene, 11% beta-pinene
and 35% 3-carene.
Exposure to alpha-pinene in the general population may occur by inhalation, by dermal contact of consumer products
and by ingestion of foods.
2. Physical and Chemical properties
Molecular weight (g/mol) 136.23
Melting point (°C) -55
Boiling point (°C) 156
Density (at 20 oC, 1 atm) 0.859
Relative density (air =1) 4.7
Solubility: Insoluble in water, soluble in alcohol, chloroform,
ether, and glacial acetic acid
Conversion factors at 20 °C and 760 mm Hg:
1 ppb = 5.654 µg/m
3
1 µg/m
3
= 0.177 ppb
Sources: Verschueren 2001, HSDB 2003
The INDEX project Final report
237
3. Indoor Air Exposure assessment
Terpene/ozone reaction products
It has been proposed that reactions between unsaturated volatile compounds (e.g. limonene, a-pinene, styrene) and
ozone or hydroxyl (OH) radicals produce chemically reactive products more likely to be responsible for eye and airway
irritation than the chemically non-reactive VOCs usually measured indoors. Many of the known reaction products are
oxygenates (e.g. formaldehyde and other aldehydes, carboxylic acids, peroxides) which may cause irritation at low
concentrations or odour annoyance due to their usually low odour thresholds (Weschler and Shields, 1997; Wolkoff et
al., 1997; 2000).
Indoor air and exposure concentrations
Exposures to alpha-Pinene in indoor environments did not follow the same increasing trend from north to south as many
other VOC compounds do (Figure 3.1). Average indoor concentrations were at about the same level in Northern and
Central Europe, being typically 15 – 17 µg/m
3
(EXPOLIS 2002, Edwards et al 2001)). Slightly lower levels were
measured in Southern Europe, being there about 10-17 µg/m
3
. Maroni et al (1995) concluded, as a summary of several
studies, a typical mean concentration being 10 µg/m
3
for alpha-Pinene in indoor air. The results presented by Kostiainen
et al (1995) in Helsinki, Finland showed similar mean and median indoor concentrations, 9.3 µg/m
3
and 7.7 µg/m
3
,
respectively. Wolkoff et al (2000) reviewed alpha-Pinene concentrations in major indoor air studies. Mean
concentrations ranged from 10 µg/m
3
to 40 µg/m
3
and maximum concentrations from 8 µg/m
3
to 250 µg/m
3
. Rehwagen
et al reported 4-week mean concentration of 25 µg/m
3
and a maximum value of 393 µg/m
3
in Leipzig, Germany.
Interesting detail in this study was the increasing trend of terpenes between 1996 and 1999 on the contrary to other
VOCs, which showed a decreasing trend. Explanations to this trend for alpha-Pinene could not be identified.
Average personal exposures to alpha-Pinene ranged from 7 µg/m
3
to 18 µg/m
3
(Figure 3.2, Jantunen et al 1999,
Hoffman et al 2000). All over Europe average population exposures tend to be lower than respective indoor
concentrations, suggesting the presence of remarkable indoor sources of alpha-Pinene. Exposures to alpha-Pinene in
Europe were considerably higher than those measured in TEAM studies in the USA, median daytime exposures ranging
from 1.0 µg/m
3
to 2.8 µg/m
3
(Wallace et al 1996).
0
20
40
60
80
100
0 1020304050607080
Indoor concentration (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
Hel
Mil
Oxf
Pra
Figure 3.1. Cumulative frequency distributions of indoor air concentrations of alpha-Pinene in Athens (Ath, n=42),
Basel (Bas, n=47), Helsinki (Hel, n=188), Milan (Mil, n=41) Oxford (Oxf, n=40) and Prague (Pra, n=46) (EXPOLIS
2002).
The INDEX project Final report
238
0
20
40
60
80
100
0 1020304050607080
Exposure (µg/m
3
)
Cumulative frequency (%)
Ath
Bas
GeS
Hel
Oxf
Pra
Figure 3.2. Cumulative frequency distributions of 48-hour personal exposure concentrations of alpha-Pinene in Athens
(Ath), Basel (Bas), Helsinki (Hel), Oxford (Oxf) and Prague (Pra) (EXPOLIS 2002), and 1-week mean exposures of the
German Survey GerES II (GeS) (Hoffman et al 2000).
4. Toxicokinetics
Absorption
The toxicokinetics of alpha-pinene was studied in volunteers experimentally exposed to the vapour of the pure
compound and of turpentine with composition in air: 54% alpha-pinene, 11% beta-pinene and 35% 3-carene (Falk et
al., 1990a; Falk Filipsson, 1996). In the first study eight healthy males were exposed to 0, 10, 225, or 450 mg/m
3
(+)-
alpha-pinene or 450 mg/m
3
(-)-alpha-pinene for 2 hr in an inhalation chamber. During exposure they exercised on a
cycle ergometer at the rate 50 watts. Average pulmonary uptake of (+)-alpha-pinene and (-)-alpha-pinene amounted to
59% (62%, 66%, and 68% respectively for turpentine vapour constituents in Falk Filipsson, 1996) of the exposure
concentration. The blood:air partition coefficient for alpha-pinene is 15. Absolute uptake increased linearly with
concentration. Blood alpha-pinene concentration increased rapidly at first then tapered off. Mean blood concentration at
the end of exposure were linearly related to inhaled concentration.
Distribution
The solubility of monoterpenes in olive oil is high; the oil:air partition coefficient for alpha-pinene is 2900. This should
imply accumulation in adipose tissue (Falk et al., 1990b). In rats, terpenes accumulate in peripheral fat, kidneys, and the
brain (Savolainen and Pfaffli, 1978; Sperling et al. 1967).
Metabolism and elimination
Slow metabolism and renal elimination of monoterpenes were suggested after a patient acutely poisoned by pine oil was
studied (Koppel et al., 1981). Up to 4% of the total uptake of alpha-pinene after human exposure by inhalation was
eliminated in the urine as cis- and transverbenol (Levin et al. 1992). Both the respiratory elimination of alpha-pinene
and the urinary excretion of verbenols after inhalation of alpha-pinene enantiomers were studied (same exposure
conditions as in Falk et al., 1990a). Respiratory elimination of both pinene enantiomers was similar; at a concentration
of 450 mg/m
3
, 7.7% of the total uptake of (+)alpha-pinene and 7.5% of the total uptake of (-)alpha-pinene was
eliminated. Urinary excretion of verbenol 4 hr after exposure to (+)alpha-pinene ranged from 1.7% at 450 mg/m
3
to
3.8% at a dose of 10 mg/m
3
. Urinary excretion of (-)alpha-pinene was similar. A semilogarithmic plot of the excretion
data suggested the existence of more than one rate constant for the elimination of (+)alpha-pinene and (-)alpha-pinene.
Most of the verbenols were eliminated within 20 hr after a 2 hr exposure. The renal excretion of unchanged alpha-
pinene was less than 0.001%. The determination of urinary verbenols may be useful as a biological exposure index for
exposure to terpenes.
The INDEX project Final report
239
Elimination of alpha-pinene from the blood was triphasic (Falk et al., 1990a). Half times for elimination of inhaled (+)-
alpha-pinene from the blood during the three phases were 4.8, 39, and 695 minutes. Elimination half times for (-)-alpha-
pinene were 5.6, 40, and 555 minutes.
5. Health effects
Effects of short-term exposure
Most of the studies identified relative to α-pinene were associated with human responses to one or several
monoterpenes. Acute toxic effects of α-pinene are stated as similar to those resulting from turpentine exposures
(Budavari, 1996; NIEHS, 2002).
Pulmonary function and subjective ratings of discomfort were described in two studies (Falk et al., 1990a; Falk
Filipsson, 1996), whereby eight healthy male volunteers were exposed for 2 hr in an inhalation chamber (working load
50W with a cycle ergometer) to 0, 10, 225, or 450 mg/m
3
(+)-alpha-pinene or 450 mg/m
3
(-)-alpha-pinene and to 450
mg/m
3
turpentine vapour (composition in air: 54% alpha-pinene, 11% beta-pinene and 35% 3-carene) , respectively.
Five among eight subjects complained of eye, nose and throat irritation, after being exposed to 450 mg/m
3
alpha-pinene.
No exposure related changes in lung function were seen. At the concentration tested the capacity of the liver to
metabolize alpha-pinene was not exceeded (Falk et al., 1990a). Subjects experienced discomfort in the throat and
airways during exposure to turpentine vapour (450 mg/m
3
) and airway resistance was increased after the end of
exposure (Falk Filipsson, 1996).
In male and female volunteers (mean age: 35 years) exposed to 0 or 450 mg/m
3
of a 10:1:5 mixture of alpha- and beta-
pinene and 3-carene (a synthetic turpentine) for 12 hr, 4 times during a 2 week period, an acute alveolar cellular
inflamatory reaction was observed. Exposure did not significantly alter bronchial hyperreactivity to methacholine
(Bingham et al., 2001).
Mølhave et al (2000) concluded that similarly to other 3 terpenes ((+)3-carene, (-)limonene, (rac)alpha-terpineol) α-
pinene can probably be ruled out as cause of acute eye irritation indoors. After applying eye goggles, too few subjects
reported eye-irritation for α-pinene to allow estimates of a threshold of this compound, having much less irritative
potency than n-butanol, 3-carene, and limonene.
No acute effects on forced vital capacity or forced expiratory volume (1s) were detected after personal exposure to 10-
214 mg/m
3
monoterpenes (alpha-pinene, beta-pinene and delta 3-carene) of 38 joinery workers (Eriksson et al., 1997)
and to 11-158 mg/m
3
terpenes of 48 sawmill workers (Eriksson et al., 1996). A decrease in carbon monoxide lung
diffusing capacity after a workshift was detected. Workers with 5 years of sawmill employment showed a higher
reactivity to methacholine than those with < 5 years. Eye irritation increased during a workday (Eriksson et al., 1996).
For single monoterpenes, mixtures of monoterpenes and for turpentine the Swedish occupational exposure limit (OEL)
value is 150 mg/m
3
. In U.S. no occupational exposure limits have been established for alpha-Pinene. For turpentine,
workplace exposure limits for 8-h workshift are 557 mg/m
3
(OSHA), 557 mg/m
3
(NIOSH), and 111 mg/m
3
(ACGIH).
As in humans, turpentine acts as a CNS depressant, with symptoms progressing from lethargy, prostration, and
convulsions to death in animals (Bystrom, 2000). Acute toxicity in animals included irritation of the skin, eyes, nose,
and mucous membranes. Signs and symptoms of acute toxicity are CNS depression and increased respiration rate with a
decrease in tidal volume. Major systemic effects include kidney and bladder injury, and are thought to be attributable to
induction of an acute inflammatory response (Key et al., 1977; cited by Baxter, 2001).
In mice exposed by inhalation to turpentine, the RD
50
(the concentration that causes a 50% decrease in respiratory
frequency) of turpentine (6500 mg/m
3
) was the same order of magnitude as those of α-pinene (5900 mg/m
3
) and β-
pinene (7100 mg/m
3
) (Kasanen et al., 1999; Kasanen et al., 1998). The effect on breathing was most likely due to
sedation or anesthesia since no histological observations were reported in the exposed animals.
The INDEX project Final report
240
Summary of short-term exposure effect levels
NOAEL
mg/m
3
LOAEL
mg/m
3
Target system;
Critical effect studied
Remarks Study Source
(Organization)
225 450 eyes, nose and throat irritation Volunteers, 2h - work load 50W Falk et al., 1990b
UBA, 2003
GV II: 2;
GV I: 0.2
UBA, 2003
Effects of long-term exposure
No chronic exposure studies were identified for both α-pinene and turpentine.
Forty-eight subjects exposed to terpenes (mean air concentration 258 mg/m
3
) and 47 unexposed subjects, all employed
at sawmills, were studied with regard to symptoms and pulmonary function. Dyspnoea and chest oppression were
significantly increased in the exposed subjects compared to the unexposed controls. A reduced FEV1, on spirometry
and an increased CV% and slope of the alveolar plateau (phase III) on single breath nitrogen washout were seen on
Monday morning before exposure to terpenes. There was no correlation between exposure time (duration of
employment) and lung function impairment. A day of industrial exposure to terpenes caused no further change in any
lung function variable. The unexposed controls showed normal spirometry and nitrogen washouts. The findings indicate
a slight stable lung function impairment of an obstructive nature which does not necessarily undergo further
deterioration with increased duration of exposure (Hedenstierna et al., 1983).
Prolonged exposure to terpenes may result in allergic contact dermatitis (Brun, 1975; Dooms-Goossens et al., 1977) or
chronic impairment of lung function (Hedenstierna et al., 1983).
Chronic effects associated with occupational exposures to turpentine include cerebral atrophy, behavioral changes,
anemia and bone marrow damage, glomerulonephritis, and dermatitis. Urinary disturbances, albuminuria, and urinary
casts were observed in workers exposed to paints and varnishes. However, renal damage associated with occupational
exposures to turpentine was transient and reversible (NIEHS, 2002).
Inhalation of turpentine (4000 mg/m
3
), four hours per day, 45 or 58 days failed to result in any hematological changes
in guinea pigs, though slight changes in the liver and moderate scattered tubular degeneration in the kidneys were
observed. Subchronic inhalation of turpentine (1670 mg/m
3
) in rats resulted in the accumulation of α-pinene in brain
and perinephric fat. At higher concentrations (estimated at 5000 to 10,000 mg/m
3
) for up to 293 hours over a period of
time ranging to 14 months provided no histological evidence of nephritis or chronic Bright’s disease. There were foci of
pneumonitis and extensive lung abscesses in many of these animals (NIEHS, 2002).
Carcinogenic and genotoxic potential
The International Agency for Research on Cancer has not evaluated the carcinogenicity of this chemical. The US
National Toxicology Program has not listed this chemical in its report on carcinogens. The American Conference of
Governmental Industrial Hygienists has designated α-pinene as not classifiable as a human carcinogen (A4). No human
information is available and the limited amount of animal information available is inconclusive.
Toxicity associated with turpentine oleoresin [CAS RN 9005-90-7] includes the development of benign tumors after
chronic exposures (Budavari, 1996).
A case-control study of workers in particle-board, plywood, sawmill, and formaldehyde glue factories demonstrated a
statistically significant association between chronic exposure (longer than 5 years) to terpenes (the principal component
of turpentine) and the development of respiratory tract cancers (HSDB, 1989).
α-Pinene was not found to be mutagenic, assayed on TA98 or TA100 in the presence of S9 (Rockwell et.al., 1979), in a
screening assay using the Ames test (Florin et. al., 1980) or using a battery of bacterial test strains (Connor et. al.
1985).
The INDEX project Final report
241
Interactions with other chemicals
It has recently been proposed that reactions between α-pinene (or other unsaturated volatile compounds) and ozone or
reactive radicals produce chemically reactive products more likely to be responsible for eye and airway irritation than
the chemically non-reactive VOCs usually measured indoors (Weschler and Shields, 1997; Wolkoff et al., 1997; 2000).
Irritative effects in experimental animals exposed to these oxidation products, were also described in Clausen et al.
(2001), Rohr et al. (2002) and Wilkins et al. (2003).
Reaction mixtures of excess alpha-pinene, limonene and isoprene with ozone considerably below their NOEL
concentrations resulted in significant upper airway irritation (Wolkoff et al, 2000). The reduction of the respiratory rate
was from 30% to about 50%. Chemical analysis of reaction mixtures by conventional methods showed that readily
identified stable products and residual reactants at the concentrations found could not account for the observed
reductions of the respiratory rate, assuming additivity of the reaction products. The results suggest that, in addition to
known irritants (formaldehyde, acrolein, methacrolein, methyl vinylketone), one or more strong airway irritant(s) of
unknown structure(s) were formed (Wolkoff et al, 2000). Effects of variation of reaction time, relative humidity and
initial ozone concentration on irritant formation were described by Wilkins et al. (2003).
Odour perception
Source: Leffingwell & Associates
(1R,5R)-(+)-alpha-pinene : harsh, terpene-like, minty
Odour threshold: 12 mg/m
3
(2.1 ppm)
(1S,5S)-(-)-alpha-pinene : harsh, terpene-like, coniferous
Odour threshold: 18 mg/m
3
(3.3 ppm)
Summary of alpha-Pinene Dose Response Assessment
________________________________________________________________________________________________
Exposure other than inhalation: Dermal: Potential hazard to the general population may derive from skin irritancy
and sensitisation from use of consumer products.
Toxicokinetics: ~62% net uptake of the inhaled amount, high affinity for adipose tissue
Health effect levels of short- and long-term exposure
NOAEL
mg/m³
LOAEL
mg/m³
Target system; critical effects Remarks Study Source; Ref.
Value mg/m³
Short-term exposure
225
450 eye, nose and throat irritation Volunteers, 2h (50W) Falk et al., 1990 UBA, 2003
GV II: 2;
GV I: 0.2
Long-term exposure (no studies have been found in literature)
Carcinogenicity: No information available for humans, the limited information for animals is inconclusive. Not
classifiable as a human carcinogen (ACGIH).
Genotoxicity: Not found to be mutagenic
Odour threshold: 3.9 mg/m
3
(Devos); (+)-alpha-pinene (harsh, terpene-like, minty): 12 mg/m
3
; (-)-alpha-pinene
(harsh, terpene-like, coniferous): 18 mg/m
3
(Leffingwell & Associates)
Remarks: RD
50
: 6 g/m
3
; In addition to known irritant reaction products (formaldehyde, acrolein, methacrolein, methyl
vinylketone), one or more strong airway irritant(s) of unknown structure(s) could be formed following the reaction
between a-pinene and ozone or reactive radicals.
________________________________________________________________________________________________
The INDEX project Final report
242
6. Risk Characterization
Health hazard evaluation of long-term exposure
Effect level - mg/m
3
Assessment
factor
EL
mg/m
3
Toxicological endpoint
Long-term
Exposure Limit
Human
LOAEL
Volunteers
450 1000
abc
0.45 Eyes, nose and throat irritation
a
extrapolation from subacute to chronic exposure (10);
b
not considering a NOAEL (10)
c
intraspecies variability (10);
Relevance of EU-population exposure to α-Pinene
Population based studies
LOAEL
1000
c
Margin of
safety
(MOS)
Description (Study, Year) N 0.45 mg/m
3
50
th
(90
th
)
Athens (Expolis, 96-98) 42 < 59 (17)
Basel (Expolis, 96-98) 47 < 191 (43)
Helsinki (Expolis, 96-98) 188 < 46 (11)
Milan (Expolis, 96-98) 41 < 69 (10)
Oxford (Expolis, 98-00) 40 < 42 (11)
Prague (Expolis, 96-98) 46 < 73 (17)
Avg MOS: 80 (18)
< out of the evaluation range (i.e. <5% of the environments investigated)
Result
An attempt has been done in deriving an exposure limit (EL) for long-term effects associated with α-pinene exposure by
refering to a study on volunteers exposed at sub-acute (2 hours) inhalation doses. When comparing this EL (450 µg/m
3
)
with the results from six indoor surveys it is concluded that no irritative effects to the eyes, nose and throat would be
expected at background α-pinene levels encountered in European homes, with median (90
th
percentile) levels at least 40
(10) times lower than the proposed EL. It is assumed that at 10-fold the level set as the EL, effects could be expected
following acute exposure. Due to its widespread use as a flavouring agent in numerous consumer products, short-term
exposures at levels in the order of some mg/m
3
could not be excluded, although significative exposure data are lacking.
An exacerbation of effects (not better defined) could be expected following the concomitant presence in residences of
ozone emitting sources, due to the formation of irritant reaction products.
The INDEX project Final report
243
5. Risk management tools
The general boundary conditions of and options for indoor air risk management are discussed in the beginning of this
report, in Chapter 3.2. Risk Management. The present chapter focuses on the characteristics of alternative indoor air
risk management tools, which are then applied for the indoor air pollutants in the first priority list (Chpt. 6.1). These
tools are:
IAQ Standards and guidelines
Building codes and ventilation standards
Equipment standards and permits
Mandatory maintenance and inspections
Limits, labelling and reporting of the contents of or releases from building products, furnishing materials,
equipment and consumer products
Public awareness raising and information
5.1 IAQ Standards and guidelines
Legally binding standards for indoor air pollution exist in most countries for a large selection of (industrial) workplace
air contaminants. The maximum allowable contaminant levels (occupational exposure limits) specified in those
standards apply for healthy adult individuals, controlled exposures and normal workweek exposure duration (40h/168h),
with provisions for simultaneous exposures to more than one regulated contaminant. Consequently the maximum
allowed concentration values (MAK-Werte, TLV, HTP etc.) are usually 1 - 3 orders of magnitude higher than those
measured in residences, offices, schools, shops and public buildings. Clearly, neither the approach nor the levels are
applicable for general indoor air risk management, although occupational exposure limit values divided by 10 (to allow
for much longer exposure time and heterogeneous target population) have been sometimes used - in the absence of
anything better - as indicative levels also in non-industrial indoor spaces.
Ambient air quality standards provide much more relevant reference values than occupational exposure limits for indoor
air quality management. Crucial differences in indoor and ambient exposures, however, exist. Occupancy is the first.
While it is virtually impossible to avoid exposure to ambient air pollution, outdoors or indoors, some indoor spaces
(residences) expose the most vulnerable occupants (infants & the old and sick) almost all the time, other indoor spaces
(work, school) are occupied only a fraction of time, yet others by only select groups for a small fraction of time (gyms,
warehouses etc). Weather is the second difference. The daily variation of weather, particularly wind, ensures that at
any location the annual average ambient air pollution concentrations are far lower than the 24 h or 1 h peak levels, and
the different ambient air pollution concentration standards for the different averaging times reflect this fact. Indoor
ventilation rates and temperatures vary much less. The levels of pollutants indoors, if they come from frequently used
indoor sources (e.g. gas appliance) or are off-gassed from building materials or furnishings (e.g. formaldehyde), can
exert high peak values virtually every day, or high and constant levels through the year. On the other hand, elevated
indoor air pollution level coming from, e.g. a fresh paint on the wall, may return to normal within days and not be
repeated in the same room for a decade. Therefore, if ambient air quality standard values are applied to assess the
quality of indoor air, it is important to know the sources of the measured pollutants, assess the long term exposures of
the occupants, and apply short and/or long term ambient air quality standards accordingly.
Specific air quality guideline values for general indoor settings have been established for only a few air contaminants
and only in some countries. The obvious reasons for these limited guideline values are on one hand the privacy of most
indoor environments, and on the other the huge numbers of buildings/rooms in any country. Consequently, in contrast
to ambient air, any indoor air monitoring data represent for short periods of time, and tiny, usually non-representative
fractions of indoor spaces. We know of only two European national surveys to monitor indoor air pollution levels in a
representative sample of residences, the three times repeated German Environmental Survey (since 1986), and the
French Indoor Air Observatory (started in 2004). More limited, but locally representative, indoor air pollution surveys
include the 7-city EXPOLIS study (1996-8) and the English Avon Study. Indoor air concentration guidelines exist in
some countries for formaldehyde (to force building material development and selection), radon (to promote radon safe
building practices and renovations in high radon regions), and CO
2
(used as ventilation sufficiency indicator). These
three exhibit typically much higher indoor than outdoor concentrations. For pollutants with significant sources and
concentrations both indoors and outdoors, indoor concentration limits cannot be managed without acknowledging the
outdoor concentrations as an unavoidable and variable background.
The INDEX project Final report
244
5.2 Building codes and ventilation standards
Building codes and ventilation standards include both legally binding governmental regulations and industry standards
or recommended professional practices (with much the same effect) and are in general more practical for indoor air risk
management than air quality standards. On one hand they are often back calculated from guideline or other reference
values to limit material and equipment/appliance emissions below certain levels (building codes) and to ensure that the
unavoidable emissions or effluents (at minimum the bio-effluents and CO
2
from the occupants) are sufficiently diluted
(ventilation standards). Compliance of all new and renovated buildings (and, as needs arise, non-residential old ones)
with building codes and ventilation standards is routinely (if not always thoroughly) controlled, while actual indoor air
quality monitoring to ensure compliance with guidelines will ever take place in only a small fraction of buildings.
5.3 Equipment/appliance standards and permits
Many indoor air risks can be managed by (i) setting standards for the design and performance of equipment (and their
installations), which might contaminate indoor air (e.g. combustion devices for heating and cooking) or fail to exhaust
the pollutants generated (e.g. kitchen extract vents/fans), (ii) implementing a mandatory test/inspection and permit
procedure to ensure that the equipment and their installations meet the standards, and (iii) requiring special training and
certification of the personnel authorised to install the equipment (e.g. gas and oil fired heaters). Such standards and
permits can be quite effective, but it should be kept in mind that they may also hinder innovation, form de facto market
barriers, unnecessarily complicate the equipment, reduce competition and thus increase the costs to end users.
5.4 Mandatory maintenance and inspections
All (combustion)heating, ventilation and air conditioning equipment need some maintenance to function properly over
time, may malfunction without obvious signal to the occupants, and their malfunction may cause or amplify indoor
hazards. Maintenance is mostly left to the judgement of the building owner or user, who may or may not know,
understand or care until something goes notably wrong. An alternative is to set legal requirements for timed inspections
and maintenance of such building equipment, which may pose health or safety risks if not functioning properly. This
can be strengthened by setting special training and certification requirements for the personnel which inspects,
maintains, repairs or in some cases even operates (e.g. public building HVAC systems) the equipment.
5.5 Limiting, labelling and reporting of the contents of or releases from building
products, furnishing materials, equipment and consumer products
Legally mandated enforced bans on the use of defined toxic compounds, concentration or emission limit values for
products which are used for constructing and furnishing buildings (e.g. asbestos, formaldehyde) and for consumer
products used in buildings (e.g. benzene) can ensure that non-necessary exposures are avoided, and that maximum
releases of these compounds to indoor air can be predicted e.g. for the calculation of ventilation needs. Legal processes,
however, are heavy instruments, which often respond only slowly to changing knowledge and needs.
Voluntary industrial standards or recommended professional practices combined with consumer awareness and
accessible information may bring the same benefits as legally mandated restrictions with better efficiency and more
flexibility. Keeping the industrial, professional and consumer dimensions of product quality information relatively
independent of each other would sacrifice some of the consistency, but improve the credibility and timeliness of the
product information.
Clearly, a combination of both legal and voluntary actions are needed. Significant health hazards call for legal
requirements, comfort and quality considerations are better managed by voluntary actions. Legal or voluntary, the
labelling and product information schemes should be coordinated at European level. Otherwise they have the risk of (i)
developing into de facto trade barriers reducing competition and increasing costs, or (ii) becoming too many and diverse
to win common approval and have the wanted impact on product safety and public health.
The INDEX project Final report
245
5.6 Awareness-raising and public information
Most indoor air risk situations must - by the necessity of their sheer numbers - be detected and managed by the
occupants themselves. Such management actions can only be based on 1) underlying general understanding of the
potential and nature of a risk associated to certain action, equipment or indoor microenvironment, and 2) ability to find
comprehensible and sufficiently detailed information for taking correct action. Therefore public awareness-raising and
information should be essential components in all indoor air risk management programmes, in addition to being
independent risk management tools.
Effective distribution of indoor air quality and risk information to the general public, which consists of individuals with
highly variable levels of background knowledge, interest and needs, is a challenge. The first priority is to prepare and
broadly distribute general information about indoor air quality and risks, aimed at awareness raising for everyone.
Awareness raising should also point out specific topics such as tobacco smoke, combustion devices/appliances,
consumer products, building materials, dampness & moisture damages, ventilation needs, etc.
This general information material should also provide links for finding more focused and detailed public information
prepared for the needs of individuals living in specific environments (e.g. high radon areas), using specific
equipment/appliances (e.g. fireplaces or gas stoves), having specific health needs (e.g. asthmatics), preferences (e.g.
natural materials), or responsibilities (e.g. building managers). Reports, information packages and websites for finding
more detailed, technical and focused information in domestic languages should be provided, so that all levels of
information needs and sophistication could be met. Also professional personal guidance should be available to be found
and consulted, when necessary.
While avoiding undue concern, public information should also be the prime method for helping people avoid or control
those lesser indoor air quality nuisances and problems, which do not warrant strict regulatory action, such as minor
odours and irritants (low TVOC levels and bioeffluents), or exposures of concern but no verified risks at common
indoor air levels (e.g. glycol ethers). Such information empowers people to improve their own indoor air quality
towards fresher and more pleasant even when other health benefits may be in doubt.
The INDEX project Final report
246
6. Recommendations and management options
The recommendations and management options proposed would - according to present knowledge - protect the general
population and most individuals most of the time, but they will not prevent every cancer from indoor exposures nor
protect the most susceptible individuals in all conditions, such as highly reactive asthmatics, individuals with other
respiratory or cardiovascular disease, genetic predisposition to haemolytic anaemia, etc. Due to the principle of diluting
the indoor air pollution by outdoor air, the outdoor air quality is a most significant factor for indoor air quality.
In addition to the specific recommendations reported below for each individual compound, the following general
recommendations and management options apply to most or many indoor air contaminants in the high and low priority
lists:
Ban tobacco smoking in all indoor spaces under public jurisdiction, and working places. Raise public
awareness on the hazards of tobacco smoke, and discourage smoking in the homes, particularly in the presence
of children.
Develop building codes to restrict the construction of attached garages, and to isolate the garages from living
and working spaces (closing the doorways, sealing the structures and ensuring proper air pressure difference
between garage and other indoor spaces).
The design of ventilation systems should ensure the dilution of all known and predictable indoor emissions of
the 1
st
priority list compounds below the recommended guideline levels, with margin of safety. Proper design
should be complemented with maintenance and use instructions to ensure that the design objectives are met
over time.
Raise public awareness about the various acute and long term risks of exposure to indoor air pollution from
indoor sources by campaigns focused on specific and concrete issues and on relevant target populations.
6.1 High priority chemicals
The high priority compound list consists of 5 gas/vapour phase chemicals, which may occur indoors in high
concentrations due to indoor sources, which have uncontested individual and public health impacts, and for which
effective risk management options are known.
Formaldehyde
The no-effect level (acute and chronic) is estimated to be at 30 µg/m
3
as 30-minute average. Pending the outcome of the
current IARC revision of the carcinogenecity of formaldehyde, a guideline value should be as low as reasonably
achievable.
Additional recommended formaldehyde risk management options are:
Minimise the emissions of formaldehyde from building materials, products, furnishings and household/office
chemicals.
Require product labelling to inform about Formaldehyde content and potential formaldehyde release from
household and building products
Discourage the use of any formaldehyde containing products.
Raise public awareness and provide information to the public about the sources, nature and levels of risks of
formaldehyde in indoor air.
Nitrogen Dioxide
A long term guideline value of 40 µg/m
3
(1-week average) and a short term guideline value of 200 µg/m
3
are proposed.
Additional recommended nitrogen dioxide risk management options are:
Apply the indoor air concentration guideline in the building and ventilation design process.
Develop building codes, ventilation standards and equipment/appliance standards (design, maintenance and
use) so that all indoor combustion equipment will exhaust into chimneys/hoods/vents leading outdoors.
Require standardised NO2 emission information - for normal use and extreme release - about all combustion
devices which do not exhaust directly into a chimney.
The INDEX project Final report
247
Provide public information about the sources, risks and means of controlling NO2 indoors.
Carbon Monoxide
The 1-hour average guideline value of 30 mg/m
3
and the 8-hour average guideline value of 10 mg/m
3
are recommended.
Additional recommended indoor air carbon monoxide risk management options are:
Apply the indoor air concentration guideline in the building and ventilation design process, considering the
possibility of excessive releases from the sources to be installed.
Develop building codes, ventilation standards and equipment/appliance standards so that they require all
indoor combustion equipment to exhaust into chimneys/hoods/vents leading outdoors.
Require standardised information CO emission under intended use for all combustion devices which do not
exhaust directly into a chimney.
Require regular mandatory inspections for indoor combustion equipment.
Recommend alarm systems responding to abnormally high concentrations.
Raise public awareness about the risks of indoor air CO, and provide public information about its sources, risks
and reasons for suspecting high CO levels.
Benzene
As benzene is a human carcinogen, its concentration in indoor air should be kept as low as reasonably achievable.
Indoor concentrations of benzene should not exceed outdoor concentrations.
Additional recommended indoor air benzene risk management options are:
Sources emitting benzene should not be allowed in the indoor environment.
Lower the permissible benzene content in any building material and consumer product, and report about
known benzene levels also when below permissible levels.
Raise public awareness and provide information to the public about the sources, nature and levels of risks of
benzene in indoor air.
Naphtalene
A long term guideline value of 10 µg/m
3
is recommended based on irritation/inflammation/hyperplasia. This level is at
the lower extreme of the olfactory perception range.
Additional recommended indoor air naphtalene exposure management options are:
Restrict the use of naphthalene containing household products, particularly mothballs.
Raise public awareness about the sources, risks, means of detecting and avoiding naphtalene in indoor air
6.2 Second priority chemicals
The steering committee did not feel that there exist sufficient evidence to propose indoor air concentration guidelines
for the second priority chemicals in indoor air. These chemicals are commonly found in indoor air and they do exhibit
toxic characteristics. Their indoor air concentrations, however, are almost always orders of magnitude below their
lowest observed effect levels, and therefore are not presently judged to justify regulatory actions.
Because they do, however, contribute to annoyance and perception of poor indoor air quality, and may contribute to the
sc. cocktail effects (presumably observable adverse effect of a complex chemical mixture in which each individual
component exists clearly below its LOEL concentration), it is justified to make people aware of such potentials of these
chemicals, and provide public information, which allows them to identify sources and means to reduce their own
exposures, when it is possible, e.g. at their own home and via their personal choices.
The INDEX project Final report
248
Acetaldehyde
Not found to be a priority compound at present, because of the large interval between inhalation exposure levels and
health effect levels. Should new information about sources, concentrations or health effects emerge, this could change
the situation.
o-, p- and m-Xylene, and Toluene
Not found to be a priority compound at present, because of the large interval between inhalation exposure levels and
health effect levels. Should new information about sources, concentrations or health effects emerge, this could change
the situation.
Styrene
A long term guideline value of 200 µg/m
3
is recommended based on neurobehavioral effects. Styrene has also been
discussed for a possible mutagenic and/or carcinogenic, but the evidence is so far inconclusive. Interaction with ozone
causing biologically active products is suspected.
6.3 Chemicals requiring further research with regard to human exposue or dose
response
The steering committee did not feel that the possible health impacts of these chemicals have been sufficiently studied to
propose indoor air concentration guidelines for these chemicals in indoor air. These chemicals are commonly found in
indoor air and they do exhibit toxic characteristics. Their indoor air concentrations, however, are usually orders of
magnitude below their lowest observed adverse effect levels, and therefore are not presently judged to justify regulatory
actions.
Because they do, however, contribute to annoyance, irritation, and ammonia also to perception of poor indoor air
quality, and they may contribute to the sc. cocktail effects (presumably observable adverse effect of a complex chemical
mixture in which each individual component exists clearly below its LOEL concentration), it is justified to make people
aware of such potentials of these chemicals, and provide public information, which allows them to identify sources and
means to reduce their own exposures, when it is possible, e.g. at their own home and via their personal choices.
Ammonia
A long term guideline value of 70 µg/m
3
and a short term guideline value of 100 µg/m
3
are recommended based on
respiratory effects.
d-Limonene
There are insufficient toxicological data available to recommend a guideline value. Considering a widespread use such
data should be made available. The odour threshold is 1-2 mg/m
3
. It appears that at present there is a reasonable safety
margin between the existing exposure levels and known odour and irritation thresholds. Interaction with ozone causing
biologically active products is suspected.
a-Pinene
There are insufficient toxicological data available to recommend a guideline value. Considering a widespread use such
data should be made available. The odour threshold is 10-20 mg/m
3
. It appears that at present there is a considerable
safety margin between the existing exposure levels and known odour and irritation thresholds. Interaction with ozone
causing biologically active products is suspected.
The INDEX project Final report
249
Annex 1 Phase 1: Exposure and dose-response data for indoor air pollutants collected in the literature review.
Scientific literature was reviewed to provide information about candidate pollutants for the later stages of the project. A summary of the concentrations in
residential indoor air, in workplaces, in residential outdoor air and in personal exposures was created for these compounds:
Compound Health
relevance
Exposure Assessment
Population Ref. No. Exposure/
Dose/Response Ass.
or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
Alkanes
Decane I/W/O/P48h 13.8/14.0/4.1/17.4 µg/m3 adults, Athens, EXPOLIS 1
I/W/O/P48h 8.6/62.9/0.6/48.8 µg/m3 adults, Basel 1
I/W/O/P48h 5.2/12.5/1.0/16.3 µg/m3 adults, Helsinki 1
b
uilding materials,
furnishings, consumer
products
I/W/O/P48h 7.5/5.2/5.8/- µg/m3 adults, Milan 1
I/W/O/P48h 4.9/3.8/1.0/7.4 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/9.7 µg/m3 adults, Germany, GerESII 8
Nonane I/W/O/P48h 5.6/6.9/2.7/7.0 µg/m3 adults, Athens, EXPOLIS 1
I/W/O/P48h 3.4/24.4/0.4/13.6 µg/m3 adults, Basel 1
I/W/O/P48h 2.1/7.0/1.0/7.9 µg/m3 adults, Helsinki 1
b
uilding materials,
furnishings, consumer
products
I/W/O/P48h 6.9/3.1/3.2/- µg/m3 adults, Milan 1
I/W/O/P48h 5.4/4.1/1.6/6.4 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/6.0 µg/m3 adults, Germany, GerESII 8
Undecane I/W/O/P48h 6.8/8.5/2.5/8.7 µg/m3 adults, Athens, EXPOLIS 1
I/W/O/P48h 8.8/38.8/0.5/15.0 µg/m3 adults, Basel 1
I/W/O/P48h 5.1/11.9/-/14.2 µg/m3 adults, Helsinki 1
b
uilding materials,
furnishings, consumer
products
I/W/O/P48h 5.6/3.6/6.8/- µg/m3 adults, Milan 1
I/W/O/P48h 6.5/4.8/1.2/8.1 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/9.9 µg/m3 adults, Germany, GerESII 8
Aromatics
Benzene C 1, H I/W/O/P48h 11.1/13.7/10.5/17.8 µg/m3 adults, Athens, EXPOLIS 1 leukaemia, 4-
6#/Million when life
2; p65 WHO 5 µg/m3 1-yr avg 3 smoking
The INDEX project Final report
250
Compound Health
relevance
Exposure Assessment
Population Ref. No. Exposure/
Dose/Response Ass.
or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
long exposure to 1
µg/m3
I/W/O/P48h 3.0/7.8/1.5/5.6 µg/m3 adults, Basel 1 5 µg/m3 1-yr avg 4
I/W/O/P48h 2.2/3.8/1.6/3.4 µg/m3 adults, Helsinki 1
I/W/O/P48h 13.2/14.0/10.4/- µg/m3 adults, Milan 1 NOAEL 0.53 ppm
(1.7 mg/m3)
10 EPA/Cal, cREL 60 µg/m3 (20
ppb) (7.4 year exposure)
10
I/W/O/P48h 12.0/9.4/5.2/11.6 µg/m3 adults, Prague 1
I/W/O/P-1week 9.4/-/4.4/12.2 µg/m3 adults, Antwerp, MACBETH 7
I/W/O/P-1week 10.1/-/20.7/18.8 µg/m3 adults, Athens 7
I/W/O/P-1week 4.5/-/3.1/6.6 µg/m3 adults, Copenhagen 7
I/W/O/P-1week 12.3/-/11.7/23.1 µg/m3 adults, Murcia 7
I/W/O/P-1week 7.0/-/8.0/10.6 µg/m3 adults, Padua 7
I/W/O/P-1week 9.5/-/4.7/13.4 µg/m3 adults, Rouen 7
I/W/O/P-1week -/-/-/13.5 µg/m3 adults, Germany, GerESII 8
I/W/O/P24h 7.2/-/3.6/7.5 µg/m3 adults, 6 States in the US,
NHEXAS
5
Diisocyanate irritant, "isocyanate
asthma"
22 glues
Ethylbenzene H I/W/O/P48h 7.7/54.1/5.9/18.6 µg/m3 adults, Athens, EXPOLIS 1 NOEL 2150 mg/m3,
increase of organ
weight
11 WHO GV= 22 mg/m3 - 1-
year
11
I/W/O/P48h 2.7/9.8/1.1/13.6 µg/m4 adults, Basel 1
I/W/O/P48h 2.8/15.0/-/7.7 µg/m3 adults, Helsinki 1
I/W/O/P48h 9.1/9.6/3.3/10.2 µg/m3 adults, Milan 1 Irritation: eyes, skin,
mucous membrane;
headache; narcosis,
coma
9 NIOSH REL: TWA 100 ppm (435 mg/m3) ST 125 ppm (545
mg/m3)
I/W/O/P48h 10.7/8.8/6.7/- µg/m3 adults, Prague 1 OSHA PEL†: TWA 100 ppm (435 mg/m3)
The INDEX project Final report
251
Compound Health
relevance
Exposure Assessment
Population Ref. No. Exposure/
Dose/Response Ass.
or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
LOAEL 250 ppm
(1250 mg/m3)
10
NOAEL 75 ppm (375
mg/m3)
10
EPA CA, cREL 2 mg/m3 (0.4
ppm) (103-week exposure)
10
I/W/O/P-1week -/-/-/24.0 µg/m3 adults, Germany, GerESII 8
m&p-Xylene H I/W/O/P48h 24.0/121/20.1/54.6 µg/m3 adults, Athens, EXPOLIS 1 11 GV= 4.8 mg/m3 - 24h
GV= 0.87 mg/m3 - 1-year
11
I/W/O/P48h 7.9/34.6/3.3/39.9 µg/m3 adults, Basel
NOAEL 304 mg/m3
CNS effects, LOAEL
870 mg/m3
neurotoxicity in rats,
OT 4.35 mg/m3 odor
annouyance
EPA CA, cREL 0.7 mg/m3
(0.2 ppm) (7-year exposure)
11
I/W/O/P48h 7.8/35.2/3.0/24.9 µg/m3 adults, Helsinki LOAEL 14.2 ppm
(49.7 mg/m3)
10
I/W/O/P48h 36.5/28.8/23.4/- µg/m3 adults, Milan Nervous system;
respiratory system
10
I/W/O/P48h 21.5/25.5/8.0/25.1 µg/m3 adults, Prague
I/W/O/P-1week -/-/-/50.5 µg/m3 adults, Germany, GerESII 8
Naphtalene H I/W/O/P48h 90.1/7.5/4.4/45.5 µg/m3 adults, Athens, EXPOLIS 1 10 moth repellents
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
I/W/O/P48h -/-/-/- µg/m3 adults, Helsinki 1
I/W/O/P48h 4.1/2.2/1.5/- µg/m3 adults, Milan 1
I/W/O/P48h 2.8/2.2/1.0/3.0 µg/m3 adults, Prague 1
LOAEL 10 ppm (52.6
mg/m3), Nasal
inflammation,
olfactory epithelial
metaplasia, and
respiratory epithelial
hyperplasia
EPA/Cal, cREL 9 µg/m3 (2
ppb) (104 week exposure)
I/W/O/P-1week -/-/-/2.3 µg/m3 adults, Germany, GerESII 8
o-Xylene H I/W/O/P48h 8.3/28.7/7.2/15.0 µg/m3 adults, Athens, EXPOLIS 1 11 GV= 4.8 mg/m3 - 24h
GV= 0.87 mg/m3 - 1-year
11
I/W/O/P48h 2.7/11.3/1.2/11.5 µg/m3 adults, Basel 1
I/W/O/P48h 2.4/15.0/1.0/9.9 µg/m3 adults, Helsinki 1
I/W/O/P48h 11.5/9.3/7.8/- µg/m3 adults, Milan 1
I/W/O/P48h 7.1/7.7/3.0/7.8 µg/m3 adults, Prague 1
NOAEL 304 mg/m3
CNS effects, LOAEL
870 mg/m3
neurotoxicity in rats,
OT 4.35 mg/m3 odor
annouyance
I/W/O/P-1week -/-/-/13.6 µg/m3 adults, Germany, GerESII 8
The INDEX project Final report
252
Compound Health
relevance
Exposure Assessment
Population Ref. No. Exposure/
Dose/Response Ass.
or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
Propylbenzene I/W/O/P48h 3.1/4.1/2.2/4.1 µg/m3 adults, Athens, EXPOLIS 1
I/W/O/P-1week -/-/-/4.6 µg/m3 adults, Germany, GerESII 8
Styrene C 2B, H, Ir I/W/O/P48h 2.4/7.1/1.8/5.4 µg/m3 adults, Athens 1 LOAEL 34 mg/m3 by
German expert group
6 GV II = 0.34 mg/m3 1-week 6 material emissions
I/W/O/P48h 1.1/-/-/1.0 µg/m3 adults, Helsinki 1 GV I = 0.03 mg/m3 1-week
I/W/O/P48h 5.5/2.9/1.9/- µg/m3 adults, Milan 1 carcinogenity in
experimental animals,
IARC 2B
2 p;106 WHO 0.26 mg/m3 1-week 2 p;107
I/W/O/P48h 3.9/3.7/1.6/3.3 µg/m3 adults, Prague 1 WHO 70 µg/m3 30-minutes 2 p;107
I/W/O/P-1week -/-/-/5.1 µg/m3 adults, Germany, GerESII 8
Toluene H I/W/O/P48h 20.1/32.4/6.4/31.1 µg/m3 adults, Basel 1 LOAEL 280 mg/m3
b
y German expert
group
6 GV II = 3 mg/m3 1-2 week 6
I/W/O/P48h 20.3/24.7/5.6/25.2 µg/m3 adults, Helsinki 1 GV I = 0.3 mg/m3 1-2 week 6
I/W/O/P48h 68.0/53.5/43.3/- µg/m3 adults, Milan 1 LOAEL 332 mg/m3,
CNS effects by WHO
2 p;113 GV = 0.26 mg/m3 1-week 11, p;53
I/W/O/P48h 74.2/69.1/25.6/88.4 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/130.2 µg/m3 adults, Germany, GerESII 8
Trimethylbenzenes I/W/O/P48h 18.2/19.4/13.8/27.3 µg/m3 adults, Athens, EXPOLIS 1
1,2,4- I/W/O/P-1week -/-/-/11.8 µg/m3 adults, Germany, GerESII 8
Alcohols
2-Ethyl-1-hexanol I/W/O/P48h 3.6/3.9/1.6/6.2 µg/m3 adults, Athens, EXPOLIS 1 mucous irritant 20;p505
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
I/W/O/P48h 3.5/2.6/-/3.0 µg/m3 adults, Helsinki 1
I/W/O/P48h 2.2/4.4/2.7/- µg/m3 adults, Milan 1
I/W/O/P48h 6.8/5.7/2.3/5.4 µg/m3 adults, Prague 1
The INDEX project Final report
253
Compound Health
relevance
Exposure Assessment
Population Ref. No. Exposure/
Dose/Response Ass.
or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
I/W/O/P-1week -/-/-/4.4 µg/m3 adults, Germany, GerESII 8
Phenol H I/W/O/P48h 25.5/0.0/1.0/- µg/m3 adults, Athens, EXPOLIS 1 Systemic effects
including liver and
nervous system
10 EPA/Cal, cREL 200 µg/m3 (50 ppb)
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1 effects 10
I/W/O/P48h -/-/-/- µg/m3 adults, Helsinki 1 LOAEL 26 ppm 10
I/W/O/P48h 0.8/3.7/-/- µg/m3 adults, Milan 1 NOAEL 5 ppm 10
I/W/O/P48h 6.4/-/-/8.4 µg/m3 adults, Prague 1
Esters
2-Buthoxyethanol I/W/O/P48h 14.8/12.0/4.1/15.0 µg/m3 adults, Athens, EXPOLIS 1 skin contact allergen
III, NKB
12 19
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
I/W/O/P48h 2.0/18.8/-/6.5 µg/m3 adults, Helsinki 1
HTP (corresb. to ¨TLV" in
the USA) 98 mg/m3 (20 ppm)
8-hr
I/W/O/P48h 7.6/14.7/0.7/- µg/m3 adults, Milan 1
I/W/O/P48h 8.5/10.1/-/6.5 µg/m3 adults, Prague 1
Terpenes
alpha-pinene I/W/O/P48h 11.4/17.4/2.7/9.0 µg/m3 adults, Athens, EXPOLIS 1 23 scented deodorisers,
polishes, cigarettes
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
reactions with O
3
may
form strong airway
irritants, see ref. 23
I/W/O/P48h 15.9/4.7/1.5/9.9 µg/m3 adults, Helsinki 1
I/W/O/P48h 14.5/1.3/6.7/- µg/m3 adults, Milan 1
I/W/O/P48h 11.9/4.8/1.4/7.1 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/18.3 µg/m3 adults, Germany, GerESII 8
d-Limonene I/W/O/P48h 82.5/14.4/5.3/55.5 µg/m3 adults, Athens, EXPOLIS 1 skin contact allergen
II B
12 scented deodorisers,
polishes, cigarettes
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
I/W/O/P48h 31.5/13.8/-/18.7 µg/m3 adults, Helsinki 1 23
I/W/O/P48h 46.6/10.5/6.2/- µg/m3 adults, Milan 1
reactions with O
3
may
form strong airway
irritants, see ref. 23
I/W/O/P48h 42.2/23.1/8.6/26.3 µg/m3 adults, Prague 1
The INDEX project Final report
254
Compound Health
relevance
Exposure Assessment
Population Ref. No. Exposure/
Dose/Response Ass.
or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
I/W/O/P-1week -/-/-/53.5 µg/m3 adults, Germany, GerESII 8
3-carene I/W/O/P48h 2.8/3.1/3.9/2.1 µg/m3 adults, Athens, EXPOLIS 1 23 scented deodorisers,
polishes, cigarettes
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
reactions with O
3
may
form strong airway
irritants, see ref. 23
I/W/O/P48h 5.6/1.6/-/3.2 µg/m3 adults, Helsinki 1
I/W/O/P48h 2.2/0.5/2.0/- µg/m3 adults, Milan 1
I/W/O/P48h 8.0/2.1/-/2.9 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/9.8 µg/m3 adults, Germany, GerESII 8
Halogenated
Dichloromethane C 2B I/W/O/P48h 15/-/5/- µg/m3 USA, Canada 2; p83 carcinogenity in
experimental animals
WHO 3 mg/m3 24 -h 2; p83 paint stripping
LOAEL 690 mg/m3 -
1.5 hr
WHO 0.45 mg/m3 1-week 2; p83
GV II = 2 mg/m3 24h 6
GV I = 0.2 mg/m3 24h 6
Pentachlorophenol H 1 µg/m3 requires mitigation 6
after mitigation 0.1 µg/m3
should be attained
Tetrachloroethylene C 2A, H I/W/O/P48h 7.7/4.7/15.3/6.4 µg/m3 adults, Athens, EXPOLIS 1 effects in kidney 2:p110 dry-cleaned clothes
I/W/O/P48h 1.2/1.4/0.7/1.3 µg/m3 adults, Basel 1 LOAEL 102 mg/m3
for humans
2:p110 WHO 0.25 mg/m3 2:p111
I/W/O/P48h -/-/-/2.8 µg/m3 adults, Helsinki 1
I/W/O/P48h 12.8/7.8/12.6/- µg/m3 adults, Milan 1 sensory effects, odour 2:p36 WHO 8 mg/m3 2:p36
I/W/O/P48h 12.3/6.2/5.7/8.1 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/3.8 µg/m3 adults, Germany, GerESII 8
Trichloroethylene C 2A, H I/W/O/P48h 11.4/7.5/9.9/11.2 µg/m3 adults, Athens, EXPOLIS 1 unit risk for cancer
0.43 #/Million
2:p116
The INDEX project Final report
255
Compound Health
relevance
Exposure Assessment
Population Ref. No. Exposure/
Dose/Response Ass.
or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
I/W/O/P48h 1.0/2.2/0.6/1.4 µg/m3 adults, Basel 1
I/W/O/P48h -/-/-/- µg/m3 adults, Helsinki 1
I/W/O/P48h 89.4/10.2/3.7/- µg/m3 adults, Milan 1
I/W/O/P48h 13.6/5.1/5.4/9.1 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/2.7 µg/m3 adults, Germany, GerESII 8
Tris-(2-chloroethyl)
phosphate
indoor in homes, schools and offices 0.01-0.25
µg/m3
24;p301 neurotoxic GV II = 0.05 mg/m3 6
GV I = 0.005 mg/m3 24h 6
Aldehydes
Acetone I/W/O/P48h 31.4/7.3/1.7/23.6 µg/m3 adults, Helsinki, EXPOLIS 1 odour annouyance,
OT 240 mg/m3
11:p51
Acetaldehyde C 2B, H I/W/O/P48h 10.1/2.6/1.5/7.9 µg/m3 adults, Helsinki, EXPOLIS 1 unit risk for cancer
0.15 -0.9 #/Million
11;p56 EPA/Cal, cREL 9 µg/m3 10
Formaldehyde C 2A, H I/W/O/P48h 33.3/12.0/2.6/21.4 µg/m3 adults, Helsinki, EXPOLIS 1 irritant above 0.1
mg/m3
2;p88 WHO 0.1 mg/m3 - 30 min 2;p90 insulation, furnishing,
ETS
indoor air 30 - 60 µg/m3 2;p87 LOAEL 0.26 mg/m3 10 EPA/Cal, cREL 3 µg/m3 (2
ppb) (10 years)
10
NOAEL 0.09 mg/m3 10
living rooms 79 µg/m3 adults, East-Germany
Methyl-ethyl-ketone H I/W/O/P48h 0.71/0.58/0.32/0.89 µg/m3 adults, Helsinki, EXPOLIS 1
Propionaldehyde H I/W/O/P48h 0.91/0.54/0.14/0.74 µg/m3 adults, Helsinki, EXPOLIS 1
Classical pollutants
CO I/W/O/P48h -/-/-/1.7 mg/m3 adults non-ETS, Athens 13 40 ppm (47 mg/m3)
for 8-hours results in
5% COHb for an
adult
15;p33 2;p77
I/W/O/P48h -/-/-/0.82 mg/m3 adults, Basel 13 COHb in blood: 2.5% 14; p88-
104
WHO 10 mg/m3 (10 ppm) 8-
hour, 30 mg/m3 (25 ppm) 1-
hour, 60 mg/m3 (50 ppm)
30-minutes, 100 mg/m3 (90
ppm) 15-minutes
b
adly flued gas
cookers and heaters,
environmental
tobacco smoke,
attached garage.
The INDEX project Final report
256
Compound Health
relevance
Exposure Assessment
Population Ref. No. Exposure/
Dose/Response Ass.
or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
I/W/O/P48h 1.2/1.3/1.5/1.3 mg/m3 adults, Helsinki 21
I/W/O/P48h -/-/-/2.17 mg/m3 adults, Milan 13
I/W/O/P48h -/-/-/1.5 mg/m3 adults, Prague 13
symtoms in patients
with cardiovascular
diseases, headache
with 20-30%,
dizziness with 30-
50% and death with
>50% concentration
of COHb
peak conc. 60 mg/m3 In the UK homes 14;p19
NO
2
Ir I/W/O/P48h 27/36/36/30 µg/m3 adults, Basel 16 complex and not fully
understood, irritant
2;p 2;p179
I/W/O/P48h 18/27/24/25 µg/m3 adults, Helsinki 16 respiratory symptoms
in asthmatics and
COPD patients
I/W/O/P48h 43/30/61/43 µg/m3 adults, Prague 16 LOAEL 375-565
µg/m3
2;p178
b
adly flued gas
cookers and heaters,
environmental
tobacco smoke
home indoors 13-40 µg/m3 UK 17;p43-44 respiratory infection,
20% increase/15 ppb
(28.3 µg/m3)
6;p46
2-week average
Inorganic comp.
Cd C 1 I/W/O/P48h -/-/-/4.8 ng/m3 adults, Athens, EXPOLIS 21 renal effects 2;p138 WHO 5 ng/m3 -1 year 2;p138 tobacco smoke,
contaminated dust
I/W/O/P48h -/-//7.8 ng/m3 adults, Basel 21
I/W/O/P48h -/-/-/22.9 ng/m3 adults, Helsinki 21
I/W/O/P48h -/-/-/4.3 ng/m3 adults, Prague 21
Pb C 2B I/W/O/P48h 97.8/154/149/93 ng/m3 adults, Athens, EXPOLIS 21 hematological and
nervous system
effects
2;p150 WHO 0.5 µg/m3 -1 year 2;p152 contaminated dust,
paint
I/W/O/P48h 15.3/16.6/20.5/24.5 ng/m3 adults, Basel 21 LOAEL for children
100µg/l in blood
2;p151
I/W/O/P48h 3.4/5.6/5.0/12.3 ng/m3 adults, Helsinki 21
I/W/O/P48h 111/132/101/- ng/m3 adults, Milan 21
The INDEX project Final report
257
Compound Health
relevance
Exposure Assessment
Population Ref. No. Exposure/
Dose/Response Ass.
or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
I/W/O/P48h 46.2/-/45.4/58.7 ng/m3 adults, Prague 21
Mercury I/W/O/P48h >89% below LOD (50 ng/m3) adults, Athens, EXPOLIS 21 renal effects, LOAEL
15 - 30 µg/m3
2;p160 WHO 1.0 µg/m3 -1 year 2;p160 paints, spills of
mercury-containing
products
I/W/O/P48h >91% below LOD adults, Basel 21 GV II = 0.35 µg/m3 6
I/W/O/P48h >89% below LOD adults, Helsinki 21 GV I = 0.035 mg/m3 24h 6
I/W/O/P48h >97% below LOD adults, Milan 21
I/W/O/P48h >90% below LOD adults, Prague 21
NH3 Ir Finland 18;p137 effects on pulmonary
function
10 cleaning solutions,
meatbolic activity
home indoors (new building) 15-51 µg/m3
LOAEL 25 ppm (17.8
mg/m3)
10 10
NOAEL 9.2 ppm (6.5
mg/m3)
EPA/Cal, cREL 200 µg/m3
(300 ppb) (12.2 years)
A = allergen
C = carcinogen classified by IARC; 1 = carcinogenic to humans, 2A = propably carcinogenic, 2B= possibly carcinogenic
H = hazardous air pollutant classified in Californian Clean Air Act Amendments of 1990
Ir = irritant
I/W/O/P(time)
HTP = Finnish occupational treshold limit value corresbonding to ¨TLV" in the USA
The INDEX project Final report
258
Annex 2. Phase 2: The selection of compounds to the further analysis
In this phase the steering group excluded compounds from the previous list based on the following criteria:
- no expressed concerns for health at present levels (for example acetone, decane, ethylbenzene, phenol , propylbenzene, trimethylbenzene)
- compound already regulated by use restrictions for indoor materials (pentachlorophenol)
- incomplete or no dose-response data available at present levels (methyl-ethyl-ketone, propionaldehyde)
- the main route/media for the exposure to the compound is other than indoor air (lead, mercury).
Compound Health
relevance
Exposure Assessment Population Ref.
No.
Exposure/ Dose/Response Ass. or risk
assessment for stochastic effects
Ref.
No.
Risk Management
Regulatory standards
Ref.
No.
Sources inside the buildings
Aromatics
Benzene C 1, H I/W/O/P48h 11.1/13.7/10.5/17.8 µg/m3 adults, Athens, EXPOLIS 1 leukaemia, 4-6#/Million when life long
exposure to 1 µg/m3
2; p65 WHO 5 µg/m3 1-yr
avg
3 Smoking
I/W/O/P48h 3.0/7.8/1.5/5.6 µg/m3 adults, Basel 1 5 µg/m3 1-yr avg 4
I/W/O/P48h 2.2/3.8/1.6/3.4 µg/m3 adults, Helsinki 1
I/W/O/P48h 13.2/14.0/10.4/- µg/m3 adults, Milan 1 NOAEL 0.53 ppm (1.7 mg/m3) 10 EPA/Cal, cREL 60
µg/m3 (20 ppb) (7.4
year exposure)
10
I/W/O/P48h 12.0/9.4/5.2/11.6 µg/m3 adults, Prague 1
I/W/O/P-1week 9.4/-/4.4/12.2 µg/m3 adults, Antwerp, MACBETH 7
I/W/O/P-1week 10.1/-/20.7/18.8 µg/m3 adults, Athens 7
I/W/O/P-1week 4.5/-/3.1/6.6 µg/m3 adults, Copenhagen 7
I/W/O/P-1week 12.3/-/11.7/23.1 µg/m3 adults, Murcia 7
I/W/O/P-1week 7.0/-/8.0/10.6 µg/m3 adults, Padua 7
I/W/O/P-1week 9.5/-/4.7/13.4 µg/m3 adults, Rouen 7
I/W/O/P-1week -/-/-/13.5 µg/m3 adults, Germany, GerESII 8
I/W/O/P24h 7.2/-/3.6/7.5 µg/m3 adults, 6 States in the US,
NHEXAS
5
m&p-Xylene H I/W/O/P48h 24.0/121/20.1/54.6 µg/m3 adults, Athens, EXPOLIS 1 11 GV= 4.8 mg/m3 -
24h GV=
0.87 mg/m3 - 1-year
11 solvents, antiknock agents in gasoline
I/W/O/P48h 7.9/34.6/3.3/39.9 µg/m3 adults, Basel
NOAEL 304 mg/m3 CNS effects, LOAEL
870 mg/m3 neurotoxicity in rats, OT 4.35
mg/m3 odor annouyance
EPA CA, cREL 0.7
mg/m3 (0.2 ppm) (7-
year exposure)
11
The INDEX project Final report
259
Compound Health
relevance
Exposure Assessment Population Ref.
No.
Exposure/ Dose/Response Ass. or risk
assessment for stochastic effects
Ref.
No.
Risk Management
Regulatory standards
Ref.
No.
Sources inside the buildings
I/W/O/P48h 7.8/35.2/3.0/24.9 µg/m3 adults, Helsinki LOAEL 14.2 ppm (49.7 mg/m3) 10 19
I/W/O/P48h 36.5/28.8/23.4/- µg/m3 adults, Milan Nervous system; respiratory system 10
HTP 50 ppm or 220
mg/m3 - 8hr, 100
ppm or 440 mg/m3 -
15 min
I/W/O/P48h 21.5/25.5/8.0/25.1 µg/m3 adults, Prague
I/W/O/P-1week -/-/-/50.5 µg/m3 adults, Germany, GerESII 8
Naphtalene H I/W/O/P48h 90.1/7.5/4.4/45.5 µg/m3 adults, Athens, EXPOLIS 1 19 Moth repellents
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
HTP 10 ppm or 53
mg/m3 - 8hr, 20
ppm or 110 mg/m3 -
15 min
I/W/O/P48h -/-/-/- µg/m3 adults, Helsinki 1
I/W/O/P48h 4.1/2.2/1.5/- µg/m3 adults, Milan 1 10
I/W/O/P48h 2.8/2.2/1.0/3.0 µg/m3 adults, Prague 1
LOAEL 10 ppm (52.6 mg/m3), Nasal
inflammation, olfactory epithelial
metaplasia, and respiratory epithelial
hyperplasia
EPA/Cal, cREL 9
µg/m3 (2 ppb) (104
week exposure)
I/W/O/P-1week -/-/-/2.3 µg/m3 adults, Germany, GerESII 8
o-Xylene H I/W/O/P48h 8.3/28.7/7.2/15.0 µg/m3 adults, Athens, EXPOLIS 1 11 11 solvent
I/W/O/P48h 2.7/11.3/1.2/11.5 µg/m3 adults, Basel 1
GV= 4.8 mg/m3 -
24h
GV= 0.87 mg/m3 -
1-year
I/W/O/P48h 2.4/15.0/1.0/9.9 µg/m3 adults, Helsinki 1 19
I/W/O/P48h 11.5/9.3/7.8/- µg/m3 adults, Milan 1
HTP 10 ppm or 53
mg/m3 - 8hr, 20
ppm or 110 mg/m3 -
15 min
I/W/O/P48h 7.1/7.7/3.0/7.8 µg/m3 adults, Prague 1
NOAEL 304 mg/m3 CNS effects, LOAEL
870 mg/m3 neurotoxicity in rats, OT 4.35
mg/m3 odor annouyance
I/W/O/P-1week -/-/-/13.6 µg/m3 adults, Germany, GerESII 8
Styrene C 2B, H, Ir I/W/O/P48h 2.4/7.1/1.8/5.4 µg/m3 adults, Athens 1 LOAEL 34 mg/m3 by German expert group 26 GV II = 0.34 mg/m3
1-week
26 Material emissions
I/W/O/P48h 1.1/-/-/1.0 µg/m3 adults, Helsinki 1 GV I = 0.03 mg/m3
1-week
I/W/O/P48h 5.5/2.9/1.9/- µg/m3 adults, Milan 1 carcinogenity in experimental animals,
IARC 2B
2 p;106 WHO 0.26 mg/m3 1-
week
2
p;10
7
I/W/O/P48h 3.9/3.7/1.6/3.3 µg/m3 adults, Prague 1 WHO 70 µg/m3 30-
minutes
2
p;10
7
The INDEX project Final report
260
Compound Health
relevance
Exposure Assessment Population Ref.
No.
Exposure/ Dose/Response Ass. or risk
assessment for stochastic effects
Ref.
No.
Risk Management
Regulatory standards
Ref.
No.
Sources inside the buildings
I/W/O/P-1week -/-/-/5.1 µg/m3 adults, Germany, GerESII 8
Toluene H I/W/O/P48h 20.1/32.4/6.4/31.1 µg/m3 adults, Basel 1 LOAEL 280 mg/m3 by German expert
group
26 GV II = 3 mg/m3 1-2
week
26 solvent
I/W/O/P48h 20.3/24.7/5.6/25.2 µg/m3 adults, Helsinki 1 GV I = 0.3 mg/m3 1-
2 week
26
I/W/O/P48h 68.0/53.5/43.3/- µg/m3 adults, Milan 1 LOAEL 332 mg/m3, CNS effects by WHO 2 p;113 GV = 0.26 mg/m3 1-
week
11, p;53
I/W/O/P48h 74.2/69.1/25.6/88.4 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/130.2 µg/m3 adults, Germany, GerESII 8
Alcohols
n-butanol Ir I/W/O/P48h 12.3/5.7/-/7.7 µg/m3 adults, Helsinki 27 eye irritant (human) 50 ppm 29 19
I/W/O/P-1week -/-/-/6.9 µg/m3 adults, Germany, GerESII 25 oral NOAEL: 125 mg/kg/day 28
HTP 50 ppm or 150
mg/m3 - 8hr, 75
ppm or 230 mg/m3 -
15 min
oral LOAEL: 500 mg/kg/day 28
2-Buthoxyethanol A, Ir I/W/O/P48h 14.8/12.0/4.1/15.0 µg/m3 adults, Athens, EXPOLIS 1 skin contact allergen III, NKB 12 HTP 98 mg/m3 (20
ppm) 8-hr
19
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
I/W/O/P48h 2.0/18.8/-/6.5 µg/m3 adults, Helsinki 1
I/W/O/P48h 7.6/14.7/0.7/- µg/m3 adults, Milan 1
I/W/O/P48h 8.5/10.1/-/6.5 µg/m3 adults, Prague 1
Terpenes
Alpha-pinene I/W/O/P48h 11.4/17.4/2.7/9.0 µg/m3 adults, Athens, EXPOLIS 1 reactions with O
3
may form strong airway
irritants
23 Scented deodorisers, polishes, cigarettes
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
I/W/O/P48h 15.9/4.7/1.5/9.9 µg/m3 adults, Helsinki 1
I/W/O/P48h 14.5/1.3/6.7/- µg/m3 adults, Milan 1
I/W/O/P48h 11.9/4.8/1.4/7.1 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/18.3 µg/m3 adults, Germany, GerESII 8
d-Limonene A I/W/O/P48h 82.5/14.4/5.3/55.5 µg/m3 adults, Athens, EXPOLIS 1 skin contact allergen II B 12 HTP 25 ppm or 140 19 Scented deodorisers, polishes, cigarettes
The INDEX project Final report
261
Compound Health
relevance
Exposure Assessment Population Ref.
No.
Exposure/ Dose/Response Ass. or risk
assessment for stochastic effects
Ref.
No.
Risk Management
Regulatory standards
Ref.
No.
Sources inside the buildings
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1 mg/m3 - 8hr, 50
ppm or 280 mg/m3 -
15 min
I/W/O/P48h 31.5/13.8/-/18.7 µg/m3 adults, Helsinki 1 23
I/W/O/P48h 46.6/10.5/6.2/- µg/m3 adults, Milan 1
reactions with O
3
may form strong airway
irritants, see ref. 23
I/W/O/P48h 42.2/23.1/8.6/26.3 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/53.5 µg/m3 adults, Germany, GerESII 8
3-carene I/W/O/P48h 2.8/3.1/3.9/2.1 µg/m3 adults, Athens, EXPOLIS 1 23 Scented deodorisers, polishes, cigarettes
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
reactions with O
3
may form strong airway
irritants, see ref. 23
I/W/O/P48h 5.6/1.6/-/3.2 µg/m3 adults, Helsinki 1
I/W/O/P48h 2.2/0.5/2.0/- µg/m3 adults, Milan 1
I/W/O/P48h 8.0/2.1/-/2.9 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/9.8 µg/m3 adults, Germany, GerESII 8
Halogenated
Dichloromethane C 2B I/W/O/P48h 15/-/5/- µg/m3 USA, Canada 2;
p83
carcinogenity in experimental animals WHO 3 mg/m3 24 -h 2;
p83
Paint stripping
LOAEL 690 mg/m3 - 1.5 hr WHO 0.45 mg/m3 1-
week
2;
p83
GV II = 2 mg/m3
24h
26
GV I = 0.2 mg/m3
24h
26
Tetrachloroethylene C 2A, H I/W/O/P48h 7.7/4.7/15.3/6.4 µg/m3 adults, Athens, EXPOLIS 1 effects in kidney 2:p110 WHO 0.25 mg/m3 2:p1
11
Dry-cleaned clothes
I/W/O/P48h 1.2/1.4/0.7/1.3 µg/m3 adults, Basel 1 LOAEL 102 mg/m3 for humans 2:p110
I/W/O/P48h -/-/-/2.8 µg/m3 adults, Helsinki 1 HTP 10 ppm or 70
mg/m3 - 8h
19
I/W/O/P48h 12.8/7.8/12.6/- µg/m3 adults, Milan 1 sensory effects, odour 2:p36 WHO 8 mg/m3 2:p3
6
I/W/O/P48h 12.3/6.2/5.7/8.1 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/3.8 µg/m3 adults, Germany, GerESII 8
The INDEX project Final report
262
Compound Health
relevance
Exposure Assessment Population Ref.
No.
Exposure/ Dose/Response Ass. or risk
assessment for stochastic effects
Ref.
No.
Risk Management
Regulatory standards
Ref.
No.
Sources inside the buildings
Trichloroethylene C 2A, H I/W/O/P48h 11.4/7.5/9.9/11.2 µg/m3 adults, Athens, EXPOLIS 1 unit risk for cancer 0.43 #/Million 2:p116 19
I/W/O/P48h 1.0/2.2/0.6/1.4 µg/m3 adults, Basel 1
HTP 30 ppm or 160
mg/m3 - 8hr, 45
ppm or 250 mg/m3 -
15 min
I/W/O/P48h -/-/-/- µg/m3 adults, Helsinki 1
I/W/O/P48h 89.4/10.2/3.7/- µg/m3 adults, Milan 1
I/W/O/P48h 13.6/5.1/5.4/9.1 µg/m3 adults, Prague 1
I/W/O/P-1week -/-/-/2.7 µg/m3 adults, Germany, GerESII 8
Tris-(2-chloroethyl)
phosphate
Ir indoor in homes, schools and offices 0.01-
0.25 µg/m3
24;p
301
neurotoxic, irritant, may act as an
carsinogen
29 GV II = 0.05 mg/m3 26 plasticizer
oral-rat LD50 1230 mg kg-1 GV I = 0.005 mg/m3
24h
26
Aldehydes
Acetaldehyde C 2B, H I/W/O/P48h 10.1/2.6/1.5/7.9 µg/m3 adults, Helsinki, EXPOLIS 1 unit risk for cancer 0.15 -0.9 #/Million 11;p56 HTP 25 ppm or 46
mg/m3 - 15min
19
EPA/Cal, cREL 9
µg/m3
10
Formaldehyde C 2A, H I/W/O/P48h 33.3/12.0/2.6/21.4 µg/m3 adults, Helsinki, EXPOLIS 1 irritant above 0.1 mg/m3 2;p88 WHO 0.1 mg/m3 -
30 min
2;p9
0
insulation, furnishing, ETS
indoor air 30 - 60 µg/m3 WHO 2;p8
7
LOAEL 0.26 mg/m3 10 HTP 0.3 ppm or 0.37
mg/m3 - 8h
19
workplaces 30 - 60 µg/m3 WHO 2;p8
7
NOAEL 0.09 mg/m3 10 EPA/Cal, cREL 3
µg/m3 (2 ppb) (10
years)
10
living rooms 79 µg/m3 adults, East-Germany skin contact allergen
Hexanal Ir I/W/O/P48h 11.8/8.7/4.8/10.1 µg/m3 adults, Athens, EXPOLIS 1
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
I/W/O/P48h 11.5/3.9/-/8.1 µg/m3 adults, Helsinki 1
I/W/O/P48h 4.7/2.2/1.0/- µg/m3 adults, Milan 1
I/W/O/P48h 10.3/10.0/2.7/10.2 µg/m3 adults, Prague 1
Benzaldehyde Ir I/W/O/P48h 7.4/11.0/5.5/8.9 µg/m3 adults, Athens, EXPOLIS 1 oral rat LOEL: 400 mg/kg/day 28
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1 oral rat NOEL: 200 mg/kg/day 28
The INDEX project Final report
263
Compound Health
relevance
Exposure Assessment Population Ref.
No.
Exposure/ Dose/Response Ass. or risk
assessment for stochastic effects
Ref.
No.
Risk Management
Regulatory standards
Ref.
No.
Sources inside the buildings
I/W/O/P48h 5.0/4.9/2.6/4.7 µg/m3 adults, Helsinki 1
I/W/O/P48h 10.6/12.3/8.9/- µg/m3 adults, Milan 1
I/W/O/P48h 9.5/9.1/4.5/11.2 µg/m3 adults, Prague 1
PAHs
BaP C 2A indoor air 0.77 / 0.52 ng/m3 nighttime / daytime 30 unit risk for lung cancer 87 000 #/Million HTP 0.01 mg/m3 -
8h
19 smoking
outdoor air 1-10 ng/m3 2;p9
2
Classical pollutants
CO I/W/O/P48h -/-/-/1.7 mg/m3 adults non-ETS, Athens 13 40 ppm (47 mg/m3) for 8-hours results in
5% COHb for an adult
15;p33 2;p7
7
I/W/O/P48h -/-/-/0.82 mg/m3 adults, Basel 13 14;
p88-
104
I/W/O/P48h 1.2/1.3/1.5/1.3 mg/m3 adults, Helsinki 21
I/W/O/P48h -/-/-/2.17 mg/m3 adults, Milan 13
I/W/O/P48h -/-/-/1.5 mg/m3 adults, Prague 13
COHb in blood: 2.5% symtoms in patients
with cardiovascular diseases, headache with
20-30%, dizziness with 30-50% and death
with >50% concentration of COHb
WHO 10 mg/m3 (10
ppm) 8-hour, 30
mg/m3 (25 ppm) 1-
hour, 60 mg/m3 (50
ppm) 30-minutes,
100 mg/m3 (90 ppm)
15-minutes
b
adly flued gas cookers and heaters,
environmental tobacco smoke, attached
garage.
peak conc. 60 mg/m3 In the UK homes 14;p
19
NO2 Ir I/W/O/P48h 27/36/36/30 µg/m3 adults, Basel 16 complex and not fully understood, irritant 2;p 2;p1
79
I/W/O/P48h 18/27/24/25 µg/m3 adults, Helsinki 16 respiratory symptoms in asthmatics and
COPD patients
WHO 40 µg/m3 -
annual, 200 µg/m3 -
1hour
I/W/O/P48h 43/30/61/43 µg/m3 adults, Prague 16 LOAEL 375-565 µg/m3 2;p178
b
adly flued gas cookers and heaters,
environmental tobacco smoke
home indoors 13-40 µg/m3 UK 17;p
43-
44
respiratory infection, 20% increase/15 ppb
(28.3 µg/m3)
6;p4
6
2-week average
Isocyanates
Diisocyanate A, Ir irritant, "isocyanate asthma" 22 HTP 0.035 mg/m3 -
8h
19 plastics and rubber chemicals, glues,
polyuretan foam
Inorganic comp.
Pb C 2B I/W/O/P48h 97.8/154/149/93 ng/m3 adults, Athens, EXPOLIS 21 hematological and nervous system effects 2;p150 WHO 0.5 µg/m3 -1
year
2;p1
52
contaminated dust, paint
The INDEX project Final report
264
Compound Health
relevance
Exposure Assessment Population Ref.
No.
Exposure/ Dose/Response Ass. or risk
assessment for stochastic effects
Ref.
No.
Risk Management
Regulatory standards
Ref.
No.
Sources inside the buildings
I/W/O/P48h 15.3/16.6/20.5/24.5 ng/m3 adults, Basel 21 LOAEL for children 100µg/l in blood 2;p151
I/W/O/P48h 3.4/5.6/5.0/12.3 ng/m3 adults, Helsinki 21
I/W/O/P48h 111/132/101/- ng/m3 adults, Milan 21
I/W/O/P48h 46.2/-/45.4/58.7 ng/m3 adults, Prague 21
NH3 Ir home indoors (new building) 15-51 µg/m3 Finland 18;p
137
effects on pulmonary function 10 19 cleaning solutions, meatbolic activity
LOAEL 25 ppm (17.8 mg/m3) 10
HTP 20 ppm or 14
mg/m3 - 8h, 50
ppm or 36 mg/m3 -
15min
NOAEL 9.2 ppm (6.5 mg/m3) 10
EPA/Cal, cREL 200
µg/m3 (300 ppb)
(12.2 years)
A = allergen
C = carcinogen classified by IARC; 1 = carcinogenic to humans, 2A = propably carcinogenic, 2B= possibly carcinogenic
H = hazardous air pollutant classified in Californian Clean Air Act Amendments of 1990
Ir = irritant
I/W/O/P(time) residential indoor/workplace/residential outdoor/ambient/personal exposure concentration (mean concentration in a given time period)
HTP = Finnish occupational treshold limit value corresbonding to ¨TLV" in the USA
The INDEX project Final report
265
Annex 3. Phase 3: Compounds selected into the detailed risk assessment.
On the basis of the available information and after an extensive discussion on the chemical substances, the steering group decided to conduct detailed
assessment for the compounds listed in the following table:
Compound Health
relevance
Exposure Assessment Population Ref.
No.
Exposure/ Dose/Response Ass. or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
Aromatics
Benzene C 1, H I/W/O/P48h
11.1/13.7/10.5/17.8 µg/m3
adults, Athens, EXPOLIS 1 leukaemia, 4-6#/Million when life long exposure to 1
µg/m3
2; p65 WHO 5 µg/m3 1-yr avg 3
I/W/O/P48h 3.0/7.8/1.5/5.6
µg/m3
adults, Basel 1 5 µg/m3 1-yr avg 4
I/W/O/P48h 2.2/3.8/1.6/3.4
µg/m3
adults, Helsinki 1
I/W/O/P48h 13.2/14.0/10.4/-
µg/m3
adults, Milan 1 NOAEL 0.53 ppm (1.7 mg/m3) 10 EPA/Cal, cREL 60 µg/m3 (20
ppb) (7.4 year exposure)
10
Smoking, attached
garages, impurities in
common solvents
(consumer products)
I/W/O/P48h 12.0/9.4/5.2/11.6
µg/m3
adults, Prague 1
I/W/O/P-1week 9.4/-/4.4/12.2
µg/m3
adults, Antwerp,
MACBETH
7
I/W/O/P-1week 10.1/-
/20.7/18.8 µg/m3
adults, Athens 7
I/W/O/P-1week 4.5/-/3.1/6.6
µg/m3
adults, Copenhagen 7
I/W/O/P-1week 12.3/-
/11.7/23.1 µg/m3
adults, Murcia 7
I/W/O/P-1week 7.0/-/8.0/10.6
µg/m3
adults, Padua 7
I/W/O/P-1week 9.5/-/4.7/13.4
µg/m3
adults, Rouen 7
I/W/O/P-1week -/-/-/13.5
µg/m3
adults, Germany, GerESII 8
I/W/O/P24h 7.2/-/3.6/7.5
µg/m3
adults, 6 States in the
US, NHEXAS
5
m&p-Xylene H I/W/O/P48h
24.0/121/20.1/54.6 µg/m3
adults, Athens, EXPOLIS 1 NOAEL 304 mg/m3 CNS effects, LOAEL 870 mg/m3
neurotoxicity in rats, OT 4.35 mg/m3 odor annouyance
11 GV= 4.8 mg/m3 - 24h
GV= 0.87 mg/m3 - 1-year
11 solvents, antiknock
agents in gasoline
The INDEX project Final report
266
Compound Health
relevance
Exposure Assessment Population Ref.
No.
Exposure/ Dose/Response Ass. or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
I/W/O/P48h 7.9/34.6/3.3/39.9
µg/m3
adults, Basel EPA CA, cREL 0.7 mg/m3
(0.2 ppm) (7-year exposure)
11
I/W/O/P48h 7.8/35.2/3.0/24.9
µg/m3
adults, Helsinki LOAEL 14.2 ppm (49.7 mg/m3) 10 19
I/W/O/P48h 36.5/28.8/23.4/-
µg/m3
adults, Milan Nervous system; respiratory system 10
HTP 50 ppm or 220 mg/m3 -
8hr, 100 ppm or 440 mg/m3 -
15 min
I/W/O/P48h
21.5/25.5/8.0/25.1 µg/m3
adults, Prague
I/W/O/P-1week -/-/-/50.5
µg/m3
adults, Germany, GerESII 8
o-Xylene H I/W/O/P48h 8.3/28.7/7.2/15.0
µg/m3
adults, Athens, EXPOLIS 1 11 11 solvent
I/W/O/P48h 2.7/11.3/1.2/11.5
µg/m3
adults, Basel 1
GV= 4.8 mg/m3 - 24h
GV= 0.87 mg/m3 - 1-year
I/W/O/P48h 2.4/15.0/1.0/9.9
µg/m3
adults, Helsinki 1 19
I/W/O/P48h 11.5/9.3/7.8/-
µg/m3
adults, Milan 1
HTP 10 ppm or 53 mg/m3 -
8hr, 20 ppm or 110 mg/m3 -
15 min
I/W/O/P48h 7.1/7.7/3.0/7.8
µg/m3
adults, Prague 1
NOAEL 304 mg/m3 CNS effects, LOAEL 870 mg/m3
neurotoxicity in rats, OT 4.35 mg/m3 odor annouyance
I/W/O/P-1week -/-/-/13.6
µg/m3
adults, Germany, GerESII 8
Naphtalene H I/W/O/P48h 90.1/7.5/4.4/45.5
µg/m3
adults, Athens, EXPOLIS 1 19 Moth repellents
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
HTP 10 ppm or 53 mg/m3 -
8hr, 20 ppm or 110 mg/m3 -
15 min
I/W/O/P48h -/-/-/- µg/m3 adults, Helsinki 1
I/W/O/P48h 4.1/2.2/1.5/-
µg/m3
adults, Milan 1 10
I/W/O/P48h 2.8/2.2/1.0/3.0
µg/m3
adults, Prague 1
LOAEL 10 ppm (52.6 mg/m3), Nasal inflammation,
olfactory epithelial metaplasia, and respiratory
epithelial hyperplasia
EPA/Cal, cREL 9 µg/m3 (2
ppb) (104 week exposure)
I/W/O/P-1week -/-/-/2.3
µg/m3
adults, Germany, GerESII 8
Styrene C 2B, H, Ir I/W/O/P48h 2.4/7.1/1.8/5.4
µg/m3
adults, Athens 1 LOAEL 34 mg/m3 by German expert group 26 GV II = 0.34 mg/m3 1-week 26 Material emissions
I/W/O/P48h 1.1/-/-/1.0 µg/m3 adults, Helsinki 1 GV I = 0.03 mg/m3 1-week
I/W/O/P48h 5.5/2.9/1.9/- adults, Milan 1 carcinogenity in experimental animals, IARC 2B 2 p;106 WHO 0.26 mg/m3 1-week 2 p;107
The INDEX project Final report
267
Compound Health
relevance
Exposure Assessment Population Ref.
No.
Exposure/ Dose/Response Ass. or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
µg/m3
I/W/O/P48h 3.9/3.7/1.6/3.3
µg/m3
adults, Prague 1 WHO 70 µg/m3 30-minutes 2 p;107
I/W/O/P-1week -/-/-/5.1
µg/m3
adults, Germany, GerESII 8
Toluene H I/W/O/P48h
20.1/32.4/6.4/31.1 µg/m3
adults, Basel 1 LOAEL 280 mg/m3 by German expert group 26 GV II = 3 mg/m3 1-2 week 26 Solvent
I/W/O/P48h
20.3/24.7/5.6/25.2 µg/m3
adults, Helsinki 1 GV I = 0.3 mg/m3 1-2 week 26
I/W/O/P48h 68.0/53.5/43.3/-
µg/m3
adults, Milan 1 LOAEL 332 mg/m3, CNS effects by WHO 2 p;113 GV = 0.26 mg/m3 1-week 11, p;53
I/W/O/P48h
74.2/69.1/25.6/88.4 µg/m3
adults, Prague 1
I/W/O/P-1week -/-/-/130.2
µg/m3
adults, Germany, GerESII 8
Terpenes
alpha-pinene I/W/O/P48h 11.4/17.4/2.7/9.0
µg/m3
adults, Athens, EXPOLIS 1 reactions with O
3
may form strong airway irritants 23
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
I/W/O/P48h 15.9/4.7/1.5/9.9
µg/m3
adults, Helsinki 1
Scented deodorisers,
polishes, cigarettes
I/W/O/P48h 14.5/1.3/6.7/-
µg/m3
adults, Milan 1
I/W/O/P48h 11.9/4.8/1.4/7.1
µg/m3
adults, Prague 1
I/W/O/P-1week -/-/-/18.3
µg/m3
adults, Germany, GerESII 8
d-Limonene A I/W/O/P48h
82.5/14.4/5.3/55.5 µg/m3
adults, Athens, EXPOLIS 1 skin contact allergen II B 12 19
I/W/O/P48h -/-/-/- µg/m3 adults, Basel 1
HTP 25 ppm or 140 mg/m3 -
8hr, 50 ppm or 280 mg/m3 -
15 min
I/W/O/P48h 31.5/13.8/-/18.7
µg/m3
adults, Helsinki 1 23
Scented deodorisers,
polishes, cigarettes
I/W/O/P48h 46.6/10.5/6.2/-
µg/m3
adults, Milan 1
reactions with O
3
may form strong airway irritants, see
ref. 23
I/W/O/P48h
42.2/23.1/8.6/26.3 µg/m3
adults, Prague 1
The INDEX project Final report
268
Compound Health
relevance
Exposure Assessment Population Ref.
No.
Exposure/ Dose/Response Ass. or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
I/W/O/P-1week -/-/-/53.5
µg/m3
adults, Germany, GerESII 8
Aldehydes
Acetaldehyde C 2B, H I/W/O/P48h 10.1/2.6/1.5/7.9
µg/m3
adults, Helsinki,
EXPOLIS
1 unit risk for cancer 0.15 -0.9 #/Million 11;p56 HTP 25 ppm or 46 mg/m3 -
15min
19
EPA/Cal, cREL 9 µg/m3 10
Cigarettes, fireplaces,
b
uilding materials,
consumer products
Formaldehyde C 2A, H I/W/O/P48h
33.3/12.0/2.6/21.4 µg/m3
adults, Helsinki,
EXPOLIS
1 irritant above 0.1 mg/m3 2;p88 WHO 0.1 mg/m3 - 30 min 2;p90 Insulation, furnishing,
ETS
indoor air 30 - 60 µg/m3 2;p8
7
LOAEL 0.26 mg/m3 10 HTP 0.3 ppm or 0.37 mg/m3
- 8h
19
NOAEL 0.09 mg/m3 10 EPA/Cal, cREL 3 µg/m3 (2
ppb) (10 years)
10
living rooms 79 µg/m3 adults, East-Germany
Classical pollutants
CO I/W/O/P48h -/-/-/1.7 mg/m3 adults non-ETS, Athens 13 40 ppm (47 mg/m3) for 8-hours results in 5% COHb
for an adult
15;p33 2;p77
I/W/O/P48h -/-/-/0.82 mg/m3 adults, Basel 13 14; p88-
104
I/W/O/P48h 1.2/1.3/1.5/1.3
mg/m3
adults, Helsinki 21
I/W/O/P48h -/-/-/2.17 mg/m3 adults, Milan 13
WHO 10 mg/m3 (10 ppm) 8-
hour, 30 mg/m3 (25 ppm) 1-
hour, 60 mg/m3 (50 ppm)
30-minutes, 100 mg/m3 (90
ppm) 15-minutes
I/W/O/P48h -/-/-/1.5 mg/m3 adults, Prague 13
COHb in blood: 2.5% symtoms in patients with
cardiovascular diseases, headache with 20-30%,
dizziness with 30-50% and death with >50%
concentration of COHb
GV II = 15 mg/m3 8-hour 26
Badly flued gas cookers
and heaters,
environmental tobacco
smoke, attached garage.
GV II = 60 mg/m3 30-min 26
peak conc. 60 mg/m3 In the UK homes 14;p
19
GV I = 1.5 mg/m3 8-hour 26
GV I = 6 mg/m3 30-min 26
NO
2
Ir I/W/O/P48h 27/36/36/30
µg/m3
adults, Basel 16 complex and not fully understood, irritant 2;p 2;p179
I/W/O/P48h 18/27/24/25
µg/m3
adults, Helsinki 16 respiratory symptoms in asthmatics and COPD patients
WHO 40 µg/m3 - annual, 200
µg/m3 - 1hour
I/W/O/P48h 43/30/61/43
µg/m3
adults, Prague 16 LOAEL 375-565 µg/m3 2;p178 GV II = 60 µg/m3 1-week 26
GV II = 350 µg/m3 30-min 26
Badly flued gas cookers
and heaters,
environmental tobacco
smoke
The INDEX project Final report
269
Compound Health
relevance
Exposure Assessment Population Ref.
No.
Exposure/ Dose/Response Ass. or risk assessment for
stochastic effects
Ref. No. Risk Management
Regulatory standards
Ref. No. Sources inside the
buildings
home indoors 13-40 µg/m3 UK 17;p
43-
44
respiratory infection, 20% increase/15 ppb (28.3
µg/m3)
6;p46
2-week average
Inorganic comp.
NH
3
Ir Finland 18;p
137
effects on pulmonary function 10 19
home indoors (new building)
15-51 µg/m3
LOAEL 25 ppm (17.8 mg/m3) 10
HTP 20 ppm or 14 mg/m3 -
8h, 50 ppm or 36 mg/m3 -
15min
NOAEL 9.2 ppm (6.5 mg/m3) 10
Cleaning solutions,
meatbolic activity
EPA/Cal, cREL 200 µg/m3
(300 ppb) (12.2 years)
A = allergen
C = carcinogen classified by IARC; 1 = carcinogenic to humans, 2A = propably carcinogenic, 2B= possibly carcinogenic
H = hazardous air pollutant classified in Californian Clean Air Act Amendments of 1990
Ir = irritant
I/W/O/P(time) residential indoor/workplace/residential outdoor/ambient/personal exposure concentration (mean concentration in a given time
period)
HTP = Finnish occupational treshold limit value corresbonding to ¨TLV" in the USA
The INDEX project Final report
270
Annex 4. The EXPOLIS study: parameters describing the indoor air concentration distributions plotted to the
graphs of this report.
Sampling
period n
A
thens
n
Basel
n
Helsinki
n
Milan
n
Oxford
n
Prague
Aromatics
Benzene
30-hour
a
42
10.1 ± 7.8
47
2.7 ± 1.7
188
2.2 ± 1.9
41
17.0 ± 23.4
40
3.6 ± 3.4
46
8.0 ± 4.6
Naphtalene
30-hour
a
42
83.5 ± 197
47
0.7 ± 0.3
188
0.6 ± 0.5
41
21.0 ± 81.6
40
1.3 ± 1.5
46
2.0 ± 1.9
Styrene
30-hour
a
42
1.8 ± 1.5
47
0.7 ± 0.6
188
1.1 ± 1.5
41
30.3 ± 157.4
40
1.8 ± 2.3
46
2.0 ± 1.7
Toluene
30-hour
a
42
82.7 ± 141
47
19.5 ± 13.2
188
20.1 ± 24.4
41
77.6 ± 70.1
40
23.7 ± 41.5
46
86.2 ± 95.7
m,p,o-Xylenes
30-hour
a
42
30.4 ± 14.6
47
10.2 ± 8.2
188
10.1 ± 7.5
41
88.2 ± 184.1
40
12.5 ± 21.7
46
22.2 ± 12.2
Terpenes
alfa-Pinene
30-hour
a
42
10.7 ± 13.2
47
4.1 ± 6.9
188
15.7 ± 23.7
41
17.9 ± 52.3
40
16.5 ± 23.5
46
10.2 ± 15.9
d-Limonene
30-hour
a
42
78.3 ± 121
47
15.0 ± 19.4
188
30.7 ± 61.5
41
71.5 ± 150.5
40
19.0 ± 28.6
46
27.1 ± 26.4
Classical pollutants
Carbon monoxide 1-minute - - 258785
1.2 ± 0.6
4822
1.9 ± 1.3
b
----
Nitrogen dioxide 48-hour
-
50
23.8 (1.7)
175
14.7 (2.1)
-
-
43 26.9 ± 21.9 33
37.7 (1.9)
Aldehydes
Formaldehyde
30-hour
a
----15
41.4 ± 22.3
------
Acetaldehyde
30-hour
a
----15
18.2 ± 9.7
------
x±y = arithmetic mean ± standard deviation, x (y) = geometric mean (geometric standard deviation)
a
Nominal sampling time
b
15 min sampling time
City
The INDEX project Final report
271
Annex 5. The National Survey of air pollutants in English homes: parameters describing the indoor air
concentration distributions plotted to the graphs of this report.
Unit Samplin
g
period n Min Max GM 10% 50% 75% 95% Reference
Aromatics
Benzene
µg/m
3
28-day
796 < 0.1 93.5 3 1 3.3 5.8 14.6 Brown VM et al 2002
Toluene
µg/m
3
28-day
796 0.3 1783.5 15.1 4.4 14.9 27.9 74.9 Brown VM et al 2002
m,p -xylene
µg/m
3
28-day
796 0.1 152.8 3.8 0.9 3.7 7.5 30.3 Brown VM et al 2002
Brown VM et al 2002
Terpenes
d-Limonene
µg/m
3
28-day
796 < 0.1 308.4 6.2 1.3 7.1 15.5 51 Brown VM et al 2002
Classical pollutants
Carbon monoxide
mg/m
3
14-day 830 < 0.01 4.45 0.47 0.14 0.5 0.9 2.07 Raw et al 2002
Nitrogen dioxide
µg/m
3
14-day 845 0.8 620 21.8 7.2 21.8 40.1 90 Coward et al 2002
Aldeh
y
des
Formaldehyde
µg/m
3
3-day
833 1 171 22.2 9.8 24 35.2 61.2 Brown VM et al 2002
n = number of samples, min = minumum, max = maximum, GM = geometric mean, 10%, 50%, 75% and 95% = ith percentile of the cumulative distribution
The INDEX project Final report
272
Annex 6. The French National Survey on Indoor Air Quality (preliminary results from the ongoing project):
parameters describing the indoor air concentration distributions plotted to the graphs of this report (Golliot et
al 2003, Kirchner 2004).
Unit Sampling period n Mean STD Min Max
A
romatics
Benzene
µg/m
3
7-day 109
2.8
2.3 0.0 13.8
Styrene
µg/m
3
7-day 109 1.4
1.7
0.2 17.0
Toluene
µg/m
3
7-day 110 25.7 35.5 2.2 196
m+p+o-Xylenes
µg/m
3
7-day 102
17.2
24.7 1.4 143
Classical pollutants
Carbon monoxide
mg/m
3
5-min 550666 0.47 1.63 0.0 62.9
A
ldehydes
Acetaldehyde
µg/m
3
7-day 201 18.4 14.0 2.6 95.0
Formaldehyde
µg/m
3
7-day
201
23.1
11.8 2.9 63.7
n = number of samples, Mean = arithmetic mean, STD = standard deviation, Min = minumum, Max = maximum
The INDEX project Final report
273
Annex 7. Relevant National and International Guidelines and Recommendations
Formaldehyde CO NO
2
Naphthalene Toluene Styrene NH
3
Monoterpene
(α-Pinene)
µg/m
3
mg/m
3
µg/m
3
µg/m
3
µg/m
3
µg/m
3
µg/m
3
µg/m
3
Finland
1
S1
30 2 30
S2
50 3 30
S3
100 8 40
Germany
2,3,4
GV II 15 (8-h) 60 (1-w) 20 3000 (1-w) 300 (1-w) 2000 (1-w)
GV II 60 (30-min)
GV I 1.5 (8-h) 2 300 (1-w) 30 (1-w) 200 (1-w)
GV I 6 (30-min)
Norway
5
100 (30-min) 10 (8-h)
25 (1-h) 100 (1-h)
UK
6
100 (30-min) 100 (15-min) 300 (1-h)
60 (30-min 40 (1-y)
30 (1-h)
10 (8-h)
WHO
7
100 (30-min) 100 (15-min) 200 (1-h) 260 (1-w) 260 (1-w)
60 (30-min 40 (1-y)
30 (1-h)
10 (8-h)
Abbreviations for averaging times: (-min) = minute, (-h) = hour, (-w = week), and (-y) = year
1
Target values for indoor air quality and climate; S1 = very good indoor air climate (Individual Indoor Climate), S2 = good indoor air climate, S3 = satisfactory indoor
climate. Values given in the table are maximum values for S1, S2 and S3. Source: Finnish classification of indoor climate. Finnish Society of Indoor Air Quality and Climate
(FiSIAQ), 2000 (in English).
2
Guidelines values (GV) for indoor air pollutants; GV II is a health-related value based on current toxicological and epidemiological knowledge. If the concentration
corresponding to GV II is reached or exceeded immediate action must be taken because permanent stay in a room at this concentration level is likely to represent a threat to
health especially for sensitive people. GV I is the concentration level at which a substance, taken individually, does not give rise to adverse health effects even at life-long
exposure. An exceedance of GV I is linked with an exposure beyond normal which is undesirable from a hygienic viewpoint. GV I and GV II are given as 1-week average,
except carbon monoxide, which was given as 8-hour (8-h) and 30-minute (30-min) average. Source: Seifert B. et al. (1999). Guidelines values for indoor air pollutant,
Proceedings of Indoor Air ’99, Edimburgh, Vol. 1: 499-504.
3
Sagunski H, Heger W (2004). Richtwerte fur die Innenraumluft: Naphthalin. Bundesgesundheitsbl –
Gesundheitsforsch – Gesundheitsschutz. 47:705-712 (in German).
4
Sagunski H, Heinzow B (2003). Richtwerte fur die Innenraumluft: Bicyclische Terpene (Leitsubstanz α-
Pinen). Bundesgesundheitsbl – Gesundheitsforsch – Gesundheitsschutz. 46:346-352 (in German).
The INDEX project Final report
274
5
Becher R. (1999). Recommended guidelines for indoor air quality, Proceedings of Indoor Air ’99, Edimburgh, Vol. 1: 171-176.
6
COMEAP (2004). Guidance on the Effects on Health of Indoor Air Pollutants. Committee on the Medical Effects of Air Pollutants (COMEAP), December 2004.
7
WHO (2000). Air Quality Guidelines for Europe. WHO Regional Publications, European Series, No. 91, Regional Office for Europe, Copenhagen.
The INDEX project Final report 275
Bibliographic references
References in introductory chapters (chapters 1 to 4)
EU (2002a). European Union Risk Assessment Report. Styrene. European Chemicals Bureau. Joint Research Centre.
Institute for Health and Consumer Protection. Existing Substances, 1st Priority List. EUR 20541 EN. Volume 27, 2002.
Available at http://ecb.jrc.it/existing-chemicals/.
EU (2002b). A draft Risk Assessment Report (RAR) on benzene. Available at http://ecb.jrc.it/existing-chemicals/.
EU (2003a). European Union Risk Assessment Report. Naphthalene. European Chemicals Bureau. Joint Research
Centre. Institute for Health and Consumer Protection. Existing Substances, 1st Priority List. EUR 20763 EN, Volume:
33, p. 230. Available at http://ecb.jrc.it/existing-chemicals/.
EU (2003b). European Union Risk Assessment Report. Toluene. European Chemicals Bureau. Joint Research Centre.
Institute for Health and Consumer Protection. Existing Substances, 2
nd
Priority List. EUR 20539 EN, Volume 30, p.
320.
Golliot F, Annesi-Maesano I, Delmas MC et al. (2003): The French National Survey on Indoor Air Quality: sample
survey design. Proc Healthy building 7th International Conference;Vol 3; p712-717
Jantunen M, Hänninen O, Katsouyanni K, Knöppel H, Künzli N, Lebret E, Maroni M, Saarela K, Srám R, Zmirou D.
(1998). Air pollution exposure in European cities: the EXPOLIS study. J Expo Anal Environ Epidemiol. 8:495-518.
Kirchner S (2004). Preliminary results of the French National Survey on Indoor Air Quality. Personal communication.
Maroni M, Seifert B, Lindvall T (eds) (1995). A Comprehensive Reference Book. Air Quality Monographs Vol. - 3.
Elsevier Science B.V., Amsterdam, The Netherlands, p. 17.
Pellizzari E, Lioy P, Quackenboss J, Whitmore R, Clayton A, Freeman N, Waldman J, Thomas K, Rodes C, Wilcosky
T. (1995). Population-based exposure measurements in EPA region 5: a phase I field study in support of the National
Human Exposure Assessment Survey. J Expo Anal Environ Epidemiol. 5(3):327-58.
Raw GJ, Coward SKD, Llewellyn JW, Brown VM, DR Crump, Ross DI (2002). Indoor air quality in English homes –
introduction and carbon monoxide findings. In: Proceedings of the 9
th
International Conference on Indoor Air Quality
and Climate, vol 4, 461-466.
Schneider P, Lorinci G, Gebefugi IL, Heinrich J, Kettrup A, Wichmann HE (1999). Vertical and horizontal variability
of volatile organic compounds in homes in Eastern Germany. J Exposure Anal Environ Epidemiol 9:282-292.
Schneider P, Gebefugi IL, Richter K, Wolke G, Schnelle J, Wichmann HE, Heinrich J, INGA study group (2001).
Indoor and outdoor BTX levels in German cities. The Science of the Total Environment 267:41-51.
Seifert B, Becker K, Hoffmann K, Krause C, Schulz C (2000). The German Environmental Survey 1990/1992
(GerESII): a representative population study. J Expo Anal Environ Epidemiol. 10:103-114.
Sexton K, Kleffman DE, Callahan MA (1995). An introduction to the National Human Exposure Assessment Survey
(NHEXAS) and related phase I field studies. J Expo Anal Environ Epidemiol. 5(3):229-32.
WHO (1989). Indoor air quality: organic pollutants. Euro reports ans Studies 111. WHO Regional Office for Europe,
Copenhagen, Denmark.
Wallace L, Nelson W, Ziegenfus R, Pellizzari E, Michael L, Whitmore R, Zelon H, Hartwell T, Perritt R, Westerdahl D.
(1991). The Los Angeles TEAM Study: Personal Exposures, Indoor-Outdoor Air Concentrations, and Breath
Concentrations of 25 Volatile Organic Compounds. J. Expo Anal Environ Epidemiol 1: 157-192.
The INDEX project Final report 276
References by compounds (chapter 4)
Formaldehyde
Main sources:
Organization Shortcut Document link
World Health Organization - Air Quality
Guidelines for Europe – 2000
[WHO] Not available on web
Office of Environmental Health Hazard
Assessment of the Californian Environmental
Protection Agency
[OEHHA1] http://www.oehha.ca.gov/air/chronic_rels/pdf/50000.pdf
Office of Environmental Health Hazard
Assessment of the Californian Environmental
Protection Agency
[OEHHA2] http://www.oehha.ca.gov/air/acute_rels/allAcRELs.html
Agency for Toxic Substances and Disease
Registry
[ATSDR] http://www.atsdr.cdc.gov/phshome.html
National Institute for Working Life - The
Nordic Expert Group for Criteria
Documentation of Health Risks from
Chemicals and The Dutch Expert Committee
on Occupational Standards
[NIWL] www.arbetslivsinstitutet.se/
National Environment Protection Council -
Australia
[NEPC] www.ephc.gov.au/nepms/air/air_toxics.html
Ahlström, R. et al. Formaldehyde odor and its interaction with the air of a sick building. Environment international, 12:
289–295 1986.
AICE. American Institute of Chemical Engineers. Guidelines for chemical process quantitative risk analysis. New York
NY: Center for Chemical Process Safety of the American Institute of Chemical Engineers; 1989. p. 148-159.
Alexandersson, R. & Hedenstierna, G. Pulmonary function in wood workers exposed to formaldehyde: a prospective
study. Archives of environmental health, 44: 5–11 1998.
Alexandersson, R. & Hedenstierna, G. Repiratory hazards associated with exposure to formaldehyde and solvents in
acid-curing paints. Archives of environmental health, 43: 222–227 1988.
Andersen I, Mølhave L. 1983. Controlled human studies with formaldehyde. In: Gibson JE, ed. Formaldehyde toxicity.
Washington, DC: Hemisphere Publishing Corporation, 154-165.
Baez AP, Belmont R, Padilla H, 1995. Measurement of formaldehyde and acetaldehyde in the atmosphere of Mexico
City. Environmental Pollution 86, 166–187.
Bakeas EB, Argyris DI, Siskos PA 2003. Carbonyl compounds in the urban environment of Athens, Greece.
Chemosphere 52, 805-813.
Bender JR, Mullin LS, Graepel GJ, et al. 1983. Eye irritation response of humans to formaldehyde. Am Ind Hyg Assoc
J 44:463-465.
Blair A, Saracci R, Stewart PA, et al. 1990. Epidemiologic evidence on the relationship between formaldehyde
exposure and cancer. Scand J Work Environ Health 16:381-393.
Broder I, Corey P, Cole P, Lipa M, Mintz S, and Nethercott JR. 1988. Comparison of health of occupants and
characteristics of houses among control homes and homes insulated with urea formaldehyde foam: Parts I methodology,
II initial health and house variables and exposureresponse relationships, and III health and house variables following
remedial work. Environ. Res. 45 2:141-203.
Brown SK 2002. Volatile organic pollutants in new and established buildings in Melbourne, Australia. Indoor Air
12:55-63.
Brown SK, Cheng M, Mahoney KJ 2002. Room chamber assessment of pollutant emission properties of low-emission
unflued gas heaters. In: Proceedings of the 9
th
International Conference on Indoor Air Quality and Climate, vol 3, 637-
642.
The INDEX project Final report 277
Brown VM, Cockram AH, Crump DR, Gavin MA 1993. Indoor air assessments in the UK carried out by the building
research establishment advisory service. In: Proceedings of the 6
th
International Conference on Indoor Air Quality and
Climate, vol 2, 111-116.
Brown VM, Coward SKD, Crump DR, Llewellyn JW, Mann HS, Raw GJ 2002. Indoor air quality in English homes –
formaldehyde. In: Proceedings of the 9
th
International Conference on Indoor Air Quality and Climate, vol 4, 473-476.
California Air Resources Board CARB 1999. California ambient air quality data 1990–1998. Planningand Technical
Support Division, Los Angeles, USA.
Casanova M, Morgan KT, Steinhagen WH, et al. 1991. Covalent binding of inhaled formaldehyde to DNA in the
respiratory tract of Rhesus monkeys: Pharmacokinetics, rat-to-monkey interspecies scaling, and extrapolation to man.
Fundam Appl Toxicol 17:409-428.
Cassee FR, Groten JP, Feron VJ. 1996. Changes in the nasal epithelium of rats exposed by inhalation to mixtures of
formaldehyde, acetaldehyde, and acrolein. Fundam Appl Toxicol 29:208-218.
Cavallo D, Alcini D, De Bortoli M, Carrettoni D, Carrer P, Bersani M, Maroni M 1993. Chemical contamination of
indoor air in schools and office buildings in Milan, Italy. In: Proceedings of the 6
th
International Conference on Indoor
Air Quality and Climate, vol 2, 45-49.
CIIT 1998. Chemical Industry Institute of Toxicology. Formaldehyde risk assessment meeting. November 14, 1997.
Research Triangle Park, NC.
Clarisse B, Laurent AM, Seta N, Le Moullec Y, El Hasnaoui A, Momas I. 2003. Indoor aldehydes: measurement of
contamination levels and identification of their determinants in Paris dwellings. Environ Res. 92 3:245-53.
COMEAP Committee on the medical effects of air pollutants 1997: Handbook on air pollution and health. Department
of health, committee on the medical effects of air pollutants. Her Majesty’s Stationary Office, London, UK.
Connor TH, Theiss JC, Hanna HA, et al. 1985. Genotoxicity of organic chemicals frequently found in the air of mobile
homes. Toxicol Lett 25:33-40.
CRC. 1994. CRC Handbook of Chemistry and Physics, 75th edition. Lide DR, ed. Boca Raton,
Crosby RM, Richardson KK, Craft TR, et al. 1988. Molecular analysis of formaldehyde-induced mutations in human
lymphoblasts and e. coli. Environ Mol Mutagen 12:155-166.
Crump KS, Howe R. Probit-A computer program to extrapolate quantile animal toxicological data to low doses. Ruston
LA: Crump KS & Company, Inc; 1983.
Crump KS. A new method for determining allowable daily intakes. Fundam Appl Toxicol 1984;4:854-871.
Dallas CE, Scott MJ, Ward JB Jr, et al. 1992. Cytogenetic analysis of pulmonary lavage and bone marrow cells of rats
after repeated formaldehyde inhalation. J Appl Toxicol 12:199-203.
Day JH, Lees REM, Clark RH, et al. 1984. Respiratory response to formaldehyde and off-gas of urea formaldehyde
foam insulation. Can Med Assoc J 131:1061-1065.
Devos M, Patte F, Rouault J, Laffort P, Van Genert LJ, 1990. Standardized Human Olfactory Thresholds. IRL Press at
Oxforf University Press.
Dykewicz, M.S. et al. Serum IgE and IgG to formaldehyde–human serum albumin: lack of relation to gaseous
formaldehyde exposure and symptoms. Journal of allergy and clinical immunology, 87: 48–57 1991.
ECETOC. European Centre for Ecotoxicology and Toxicology of Chemicals. editor. 65. cancer risk, Formaldehyde and
human 1995. Brussels. ECETOC technical report 65.
Edling C, Hellquist H, Odkvist L. 1988. Occupational exposure to formaldehyde and histopathological changes in the
nasal mucosa. Br. J. Ind. Med. 45 11:761-765.
EPA 2002. The fact sheet on Formaldehyde. Environmental Health Center. The National Safety Council, USA.
Available at http://www.nsc.org/ehc/indoor/formald.htm.
EPA California, USA. OEHHA Office of Environmental Health Hazard Assessment,
http://www.oehha.ca.gov/air/chronic_rels/. Accessed in November, 2003.
European Concerted Action 1990. Indoor pollution by formaldehyde in European countries. ECA, Indoor air quality &
its impact on man, COST Project 613, report no 7. Brussels-Luxemburg, Commission of the European Communities.
Feinman SE, editor. Formaldehyde sensitivity and toxicity. Boca Raton FL: CRC Press Inc; 1988.
Feng Y, Wen S, Wang, Sheng G, He Q, Tang J, Fu J 2004. Indoor and outdoor carbonyl compounds in the hotel
ballrooms in Guangzhou, China. Atmospheric Environment 38, 103–112.
FL: CRC Press Inc.
Franklin P, Dingle P, Stick S. 2000. Raised exhaled nitric oxide in healthy children is associated with domestic
formaldehyde levels. Am J Respir Crit Care Med. 2000 May;161 5:1757-9.
The INDEX project Final report 278
Garrett MH, Hooper MA, Hooper BM, Rayment PR, Abramson MJ. 1999. Increased risk of allergy in children due to
formaldehyde exposure in homes. Allergy 1999 Dec;54 12:1327.
Gordon SM, Callahan PJ, Nishioka MG, Brinkman MC, O'Rourke MK, Lebowitz MD, Moschandreas DJ 1999.
Residential environmental measurements in the national human exposure assessment survey NHEXAS pilot study in
Arizona: preliminary results for pesticides and VOCs. J Expo Anal Environ Epidemiol, 9:456-470.
Gorski P, and Krakowiak A. 1991. Formaldehyde induced bronchial asthma - Does it really exist? Polish J. Occup.
Med. 4 4:317-320.
Gorski P, Tarkowski M, Krakowiak A, et al. 1992. Neutrophil chemiluminescence following exposure to formaldehyde
in healthy subjects and in patients with contact dermatitis. Allergol Immunopathol Madr 20:20-23.
Grafström RC, Fornace A, Harris CC. 1984. Repair of DNA damage caused by formaldehyde in human cells. Cancer
Res 44:4323-4327.
Grammer LC, Harris KE, Shaughnessy MA, Sparks P, Ayars GH, Altman LC, and Patterson R. 1990. Clinical and
immunologic evaluation of 37 workers exposed to gaseous formaldehyde. J. Allergy Clin. Immunol. 86 2:177-188.
Granby K, Christensen CS, 1997. Urban and semi-rural observations of carboxylic acids and carbonyls. Atmospheric
Environment 31, 1403–1415.
Green DJ, Bascom R, Healey EM, et al. 1989. Acute pulmonary response in healthy, nonsmoking adults to inhalation of
formaldehyde and carbon. J Toxicol Environ Health 28:261-275.
Green DJ, Sauder LR, Kulle TJ, et al. 1987. Acute response to 3.0 ppm formaldehyde in exercising health nonsmokers
and asthmatics. Am Rev Respir Dis 135:1261-1266.
Grosjean D, Grosjean E, Moreira LFR, 2002. Speciated ambient carbonyls in Rio de Janeiro, Brazil. Environmental
Science and Technology 36, 1389–1395.
Grosjean E, Williams II, Grosjean, D, 1993. Ambient levels of formaldehyde and acetaldehyde in Atlanta, Georgia.
Journal of Air and Waste Management Association 43, 469–474.
Gustafson P, Barregard L, Lindahl R, Sallsten G 2004. Formaldehyde levels in Sweden: personal exposure, indoor, and
outdoor concentrations. J Expo Anal Environ Epidemiol, online publication, 1-9.
Hansen, J. & Olsen, J.H. Formaldehyde and cancer morbidity among male employees in Denmark. Cancer causes and
control, 6: 354–360 1995.
Harving H, Korsgaard J, Dahl R, et al. 1986. Low concentrations of formaldehyde in bronchial asthma: a study of
exposure under controlled conditions. Br Med J 293:310.
Harving H, Korsgaard J, Pedersen OF, et al. 1990. Pulmonary function and bronchial reactivity in asthmatics during
low-level formaldehyde exposure. Lung 168:15-21.
Health Canada, Environment Canada. 2001. Priority substances list assessment report: Formaldehyde. Ottawa. Ministry
of Public Works and Government Services. Available at http://www.hc-sc.gc.ca/hecs-sesc/exsd/psl2.htm
Heck Hd’A, Casanova M, Steinhagen WH et al. 1989. DNA-protein cross-linking studies in rats and nonhuman
primates. In: Feron VJ and Bosland MC, eds. Nasal carcinogenesis in rodents: Relevance to human health risk. The
Netherlands: Pudoc Wageningen, 159-164. Cited in EPA, 1991a
Ho KF, Lee SC, Louie PKK, Zou SC, 2002. Seasonal variation of carbonyls compound concentrations in urban area of
Hong Kong. Atmospheric Environment 36, 1259–1265.
Hodgson AT, Beal D, McIlvane JER 2002. Sources of formaldehyde, other aldehydes and terpenes in a new
manufactured house. Indoor Air 12:235-242.
Hodgson AT, Rudd AF, Beal D, Chandra S 2000. Volatile organic compound concentrations and emission rates in new
manufactured and site-built houses. Indoor Air 10:178-192.
Holmstrom M, Wilhelmsson B, Hellquist H, et al. 1989. Histological changes in the nasal mucosa in persons
occupationally exposed to formaldehyde alone and in combination with wood dust. Acta Otolaryngol Stockh 107:120-
129.
Holness, D.L. & Nethercott, J.R. Health status of funeral service workers exposed to formaldehyde. Archives of
environmental health, 44: 222–228 1989.
HSDB 2003. Hazardous Substances Data Bank. Specialised Information Services. National Library of Medicine.
Available online at http://toxnet.nlm.nih.gov/.
IARC 1995 Formaldehyde. In: Wood dust and formaldehyde. Monographs on the Evaluation of the Carcinogenic Risk
of Chemicals to Humans, Vol. 62, pp. 217–362 Lyon, 1995.
IPCS/WHO. Formaldehyde. In: Environmental Health Criteria 89. Geneva: International Programme on Chemical
Safety, World Health Organisation, 1989.
The INDEX project Final report 279
Jurvelin J, Vartiainen M, Jantunen M, Pasanen P. 2001. Personal Exposure Levels and Microenvironmental
Concentrations of Formaldehyde and Acetaldehyde in the Helsinki metropolitan Area, Finland. J.Air &Waste Manage.
Assoc. 51:17-24.
Kelly TJ, Smith DL, Satola J 1999. Emission rates of formaldehyde form materials and consumer products found in
California homes. Environmental Science and Technology 33, 81–88.
Khamgaonkar, M.B. & Fulare, M.B. Pulmonary effects of formaldehyde exposure. An environmental–epidemiological
study. Indian journal of chest diseases and allied sciences, 33: 9–13 1991.
Kirchner S 2004. Preliminary results of the French National Survey on Indoor Air Quality. Personal communication.
Krakowiak A, Gorski P, Pazdrak K, et al. 1998. Airway response to formaldehyde inhalation in asthmatic subjects with
suspected respiratory formaldehyde sensitization. Am J Ind Med 33:274-281.
Kramps, J.A. et al. Measurement of specific IgE antibodies in individuals exposed to formaldehyde. Clinical and
experimental allergy, 19: 509–514 1989.
Krzyzanowski, M, J.J. Quackenboss and M.D. Lebowitz. 1990. Chronic respiratory effects of indoor formaldehyde
exposure. Environ. Res. 52: 117-125
Kulle TJ, Sauder LR, Hebel JR, et al. 1987. Formaldehyde dose-response in healthy nonsmokers. J Air Pollut Control
Assoc 37:919-924.
Kulle TJ. 1993. Acute odor and irritation response in health nonsmokers with formaldehyde exposure. Inhal Toxicol
5:323-332.
Lam, CH.-W, Casanova, M, and Heck, H.D'A. 1985 Depletion of nasal mucosal glutathione to acrolein and
enhancement of formaldehyde induced DNA-protein cross linking by simultaneous exposure to acrolein. Arch. Toxicol,
58: 67-71.
Lee SC, Wang B. 2004. Characteristics of emissions of air pollutants from burning of incense in a large environmental
chamber. Atmospheric Environment 38:941-951.
Levine, R.J. et al. The effects of occupational exposure on the respiratory health of West Virginia morticians. Journal of
occupational medicine, 26: 91–98 1984.
Li YY, Wu PC, Su HJ, Chou PC, Chiang CM 2002. Effects of HVAC ventilation efficiency on the concentrations of
formaldehyde and total volatile organic compounds in office buildings. In: Proceedings of the 9th International
Conference on Indoor Air Quality and Climate, vol 3, 376-381.
Lindstrom AB, Proffitt D, Fortune CR 1995. Effects of modified residential construction on indoor air quality. Indoor
Air 5:258-269.
Liu KS, Huang FY, Hayward SB, et al. 1991. Survey of formaldehyde concentrations and health effects in mobile
homes [Abstract]. Arch Environ Health 46:181-182.
Main, D.M. & Hogan, T.J. Health effects of low-level exposure to formaldehyde. Journal of occupational medicine, 25:
896–900 1983.
Malaka, T. & Kodama, A.M. Respiratory health of plywood workers occupationally exposed to formaldehyde. Archives
of environmental health, 45: 288–294 1990.
McLaughlin, J.K. Formaldehyde and cancer: a critical review. International archives of occupational and environmental
health, 66: 295–301 1994.
Migliore L, Ventura L, Barale R, et al. 1989. Micronuclei and nuclear anomalies induced in the gastrointestinal
epithelium of rats treated with formaldehyde. Mutagenesis 4:327-334.
Moncrieff, R.W. 1955 A technique for olfactory, trigeminal, and ocular irritations. Q. J. exp. Psychol, 7: 128-132.
National Institute for Working Life NIWL – Sweden. The Nordic Expert Group for Criteria Documentation of Health
Risks from Chemicals and The Dutch Expert Committee on Occupational Standards. 132. Formaldehyde Anton
Wibowo. ISBN 91-7045-687-9 ISSN 0346-7821 available in the Internet at: www.arbetslivsinstitutet.se/
NIOSH. National Institute for Occupational Safety and Health. Health Hazard Evaluations: Formaldehyde. CDROM for
1981-1989 and 1990-1996
Nordman H, Keskinen H, Tuppurainen M. 1985. Formaldehyde asthma - Rare or overlooked? J Allergy Clin Immunol
75:91-99.
Norppa, H. et al. Increased frequency of micronucleated cells in buccal mucosa but not in lymphocyte cultures of
formaldehyde-exposed workers. In: Abstracts of the Sixth International Conference on Environmental Mutagens,
Melbourne, 1993, Abstract 110.
Nunn, A.J. et al. Six year follow up of lung function in men occupationally exposed to formaldehyde. British journal of
industrial medicine, 47: 747–752 1990.
The INDEX project Final report 280
Olsen JC, and Dossing M. 1982. Formaldehyde induced symptoms in day care centers. Am. Ind. Hyg. Assoc. J. 43
5:366-370.
Partanen, T. Formaldehyde exposure and respiratory cancer – a meta-analysis of the epidemiologic evidence.
Scandinavian journal of work, environment and health, 19: 8–15 1993.
Patterson, R. et al. IgG antibody against formaldehyde human serum proteins: a comparison with other IgG antibodies
against inhalant proteins and reactive chemicals. Journal of allergy and clinical immunology, 84: 359–366 1989.
Pazdrak K, Gorski P, Krakowiak A, Ruta U. Changes in nasal lavage fluid due to formaldehyde inhalation. Int Arch
Occup Environ Health 1993;64 7:515-519
Possanzini M, Di Palo V, Petricca R, Fratarcangeli R, Brocco D, 1996. Measurements of lower carbonyls in Rome
ambient air. Atmospheric Environment 30, 3757–3764.
Pross, J.F. et al. Immunologic studies of subjects with asthma exposed to formaldehyde and urea–formaldehyde foam
insulation UFFI off products. Journal of allergy and clinical immunology, 79: 797–810 1987.
Puhakka E and Karkkainen J 1993. Formaldehyde concentrations in indoor air in new buildings; factors that affect the
prevalence of formaldehyde. In: Proceedings of the 6
th
International Conference on Indoor Air Quality and Climate, vol
2, 147-152.
Recio L, Sisk S, Pluta L, et al. 1992. p53 mutations in formaldehyde-induced nasal squamous cell carcinomas in rats.
Cancer Res 52:6113-6116.
Risby, T.H. et al. Model to estimate effective doses of adsorbed pollutants on respirable particles and their subsequent
release into alveolar surfactant. I. Validation of the model for the adsorption and release of formaldehyde on a
respirable carbon black. Inhalation toxicology, 2: 223–239 1990.
Ritchie IM, and Lehnen RG. 1987. Formaldehyde-related health complaints of residents living in mobile and
conventional homes. Am. J. Public Health 77:323-328.
Rothenberg, S.J. et al. Surface area, adsorption, and desorption studies on indoor dust samples. American Industrial
Hygienists Association journal, 50: 15–23 1989.
Sakai K, Norback D, Mi Y, Shibata E, Kamijima M, Yamada T, Yasuhiro T 2004. A comparison of indoor air
pollutants in Japan and Sweden: formaldehyde, nitrogen dioxide, and chlorinated volatile organic compounds. Environ
Res 94:75-85.
Salkie, M.L. The prevalence of atopy and hypersensitivity to formaldehyde in pathologists. Archives of pathology and
laboratory medicine, 115: 614–616 1991.
Satsumabayashi H, Kurita H, Chang YS, Carmichael GR, Ueda H, 1995. Photochemical formations of lower aldehydes
and lower fatty acids under long-range transport in central Japan. Atmospheric Environment 29, 255–266.
Sauder, L.R. et al. Acute pulmonary response to formaldehyde exposure in healthy nonsmokers. Journal of occupational
medicine, 28: 420–424 1986.
Schachter EN, Witek TJJ, Brody DJ, et al. 1987. A study of respiratory effects from exposure to 2.0 ppm formaldehyde
in occupationally exposed workers. Environ Res 44:188-205.
Schachter EN, Witek TJJ, Tosun T, et al. 1986. A study of respiratory effects from exposure to 2 ppm formaldehyde in
healthy subjects. Arch Environ Health 41:229-239.
Sexton K, Petreas MX, Liu K-S 1989. Formaldehyde exposures inside mobile homes. Environmental Science and
Technology 23:985-988.
SIS 2003 Specialised Information Services. US National Library of Medicine. ChemlPlus Record. Available in
http://chem2.sis.nlm.nih.gov/chemidplus/ProxyServlet?objectHandle=DBMaint&actionHandle=default&nextPage=jsp/
chemidlite/ResultScreen.jsp&TXTSUPERLISTID=000050000 last updated October 1, 2003 .
Slooff et al. 1992 Exploratory report formaldehyde. National Institute of Public Heatlh and Environmental,
editor.RIVM report 710401018. Bilthoven. Ref Type: Report
U.S. EPA 1991. Environmental Protection Agency. Formaldehyde risk assessment update - final draft. Washington,
DC: U.S. Environmental Protection Agency, Office of Toxic Substances Disease Registry.
U.S. EPA 2002. Environmental Protection Agency. Integrated Risk Information System: Formaldehyde. Office of
Research and Development, National Centre for Environmental Assessment, Washington, DC
Uba et al. 1989 Prospective study of respiratory effects of formaldehyde among healthy and asthmatic medical students.
American journal of industrial medicine, 15: 91–101.
Ullrich D, Gleue C, Krause C, Lusansky C, Nagel R, Schultz C, Seifert B 2002. German Environmental Survey of
children and teenagers 2000 GerES IV: a representative population study including indoor pollutants. In: Proceedings of
the 9
th
International Conference on Indoor Air Quality and Climate, vol 1, 209-214.
The INDEX project Final report 281
Verschueren K 2001. Handbook of Environmental Data on Organic Chemicals. Fourth edition. John Wiley & Sons, Inc,
New York. Vol. 1, 24.
Viskari E, Vartiainen M, Pasanen P, 2000. Seasonal and diurnal variation in formaldehyde and acetaldehyde
concentrations along a highway in Eastern Finland. Atmospheric Environment 34, 917–923.
Wantke F, Demmer CM, Tappler P, Gotz M, Jarisch R. 1996. Exposure to gaseous formaldehyde induces IgE-mediated
sensitization to formaldehyde in school-children. Clin Exp Allergy. 1996 Mar;26 3:276-80.
Weber-Tschopp A, Fischer T, Grandjean E. 1977. Irritating effects of formaldehyde on men. Int Arch Occup Environ
Health 39:207-218. German
WHO 1987. World Health Organization. Air quality guidelines for Europe. WHO Regional Office for Europe, WHO
Regional Publications, European Series, No. 23, Copenhagen,1987.
WHO 1989. Environmental health criteria for acetaldehyde, 89. WHO, Geneva, 1989. Available in the Internet
http://www.inchem.org/documents/ehc/ehc/ehc89.htm.
WHO 1989. World Health Organization, Formaldehyde. Environmental Health Criteria, No. 89, Geneva, 1989
International Agency for Research on Cancer IARC, Formaldehyde. In: Wood dust and formaldehyde.
WHO 2000. Guidelines for Air Quality. WHO Geneva, Switzerland. Available in the Internet at
http://www.who.int/peh/air/Airqualitygd.htm.
WHO 2001. World Health Organization, Air quality guidelines for Europe 2000. WHO Regional Office for Europe,
WHO Regional Publications, European Series, No. 91, Copenhagen,2001.
Wilhelmsson B, and Holmstrom M. 1992. Possible mechanisms of formaldehyde-induced discomfort in the upper
airway. Scand. J. Work. Environ. Health 18 6:403-407.
Wilmer JWG, Woutersen RA, Appelman LM, et al. 1987. Subacute 4-week inhalation toxicity study of formaldehyde in
male rats: 8-hour intermittent versus 8-hour continuous exposures. J Appl Toxicol 7:15-16.
Witek TJ, Schachter EN, Tosun T, et al. 1986. Controlled human studies on the pulmonary effects of indoor air
pollution: Experiences with sulfur dioxide and formaldehyde. Environ Int 12:129-135.
Zwiener G, Wortmann G, Wortmann FJ, Sweredjuk R, Doppelmayer F 1999. Sorption of indoor formaldehyde by wool.
In: Proceedings of the 8
th
International Conference on Indoor Air Quality and Climate, vol 1, 414-419.
The INDEX project Final report 282
Carbon monoxide
Main sources:
Authority Shortcut Link to original document
WHO- Air Quality Guidelines for
Europe - 2000
[WHO] no Internet link
WHO – IPCS Environmental
Health Criteria 213 - 1999
[IPCS] http://www.inchem.org/documents/ehc/ehc/ehc213.htm
Office of Environmental Health
Hazard Assessment of the
Californian Environmental
Protection Agency
Acute Toxicity summary 1999
[OEHHA1] http://www.oehha.ca.gov/air/acute_rels/pdf/630080A.pdf
OEHHA - Evaluation of current
California Air Quality Standards
with respect to protection of
children 2000
[OEHHA2] http://www.oehha.ca.gov/air/pdf/oehhaco.pdf
ACGIH 1991a. Chemical Substances TLV Committee. Notice of intended change - carbon monoxide. Applied
occupational and environmental hygiene, 6: 621–624.
ACGIH 1991b. Chemical Substances TLV Committee. Notice of intended change – carbon monoxide. Applied
occupational and environmental hygiene, 6: 896–902.
Adams, K.F, G. Koch, B. Chatterjee, G.M. Goldstein, J.J. O'Neil, P.A. Bromberg, D.S. Sheps, S. McAllister, C.J. Price,
and J. Bissette. 1988. Acute elevation of blood carboxyhemoglobin to 6% impairs exercise performance and aggravates
symptoms in patients with ischemic heart disease. J. Am. Coll. Cardiol. 12:900-909.
Akland GG, Hartwell TD, Johnson TR, Whitmore RW 1985. Measuring human exposure to carbon monoxide in
Washington, DC, and Denver, Colorado, during the winter of 1982-1983. Environ Sci Technol 19:911-918.
Alderman, B.W, A.E. Baron, and D.A. Savitz. 1987. Maternal exposure to neighborhood carbon monoxide and risk of
low infant birth weight, Public Health Rep. 102:410-414.
Allred, E.N, E.R. Bleeker, B.R. Chaitman, T.E. Dahms, S.O. Gottlieb, J.D. Hackney, M. Pagano, R.H. Selvester, S.M.
Walden, and J. Warren. 1989. Short-term effects of carbon monoxide on the exercise performance of subjects with
coronary artery disease New England J. Med. 321:1426-1432.
Alm S, Mukala K, Jantunen MJ 2000. Personal carbon monoxide exposures of preschool children in Helsinki, Finland:
levels and determinants. Atmos Environ 34 2:277-285.
Alm S. Personal exposures of pre-school children to carbon monoxide and nitrogen dioxide in urban air. Academic
dissertation. Publications of the National Public Health Institute A19/99. National Public Health Institute, Kuopio,
Finland; 1999.
Altman, P. L. & Dittmen, D. S, ed. 1971 Respiration and circulation, Bethesda, MD, Federation of the American
Society of Experimental Biology.
Anderson, E.W. et al. Effect of low-level carbon monoxide exposure on onset and duration of angina pectoris, a study
in ten patients with ischemic heart disease. Annals of internal medicine, 79: 46–50 1973.
Aronow, W.S. & Isbell, M.W. Carbon monoxide effect on exercise-induced angina pectoris. Annals of internal
medicine, 79: 392–395 1973.
Aronow, W.S. et al. Effect of freeway travel on angina pectoris. Annals of internal medicine, 77: 669–676 1972.
Atkinson RW, Bremner SA, Anderson HR, Strachan DP, Bland JM, de Leon AP 1999. Short-term associations between
emergency hospital admissions for respiratory and cardiovascular disease and outdoor air pollution in London. Arch
Environ Health. 1999 Nov-Dec;54 6:398-411.
Balraj, E.K, 1984. Atherosclerotic coronary artery disease and "low" levels of carboxyhemoglobin: Report of fatalities
Bascom, R. et al. Health effects of outdoor air pollution Part 2. American journal of respiratory and critical care
medicine, 153: 477–498 1996.
Benignus, V.A. Behavioral effects of carbon monoxide: meta analyses and extrapolations. Journal of applied
physiology, 76:1310–1316 1994.
Benignus, V.A. et al. Effect of low level carbon monoxide on compensatory tracking and event monitoring.
Neurotoxicology and teratology, 9: 227–234 1987.
Benignus, V.A. Importance of experimenter-blind procedure in neurotoxicology. Neurotoxicology and teratology, 15:
45–49 1993.
The INDEX project Final report 283
Berk PD, Rodkey FL, Blaschke TF, Collison HA, & Waggoner JG 1974 Comparison of plasma bilirubin turnover and
carbon monoxide production in man. J Lab Clin Med, 83: 29-37.
Brown S.D. and Piantadosi, C.S. 1992 Recovery of energy metabolism in rat brain after carbon monoxide hypoxia. J.
Clin Invest. 89:666-672.
Brown SK, Cheng M, Mahoney KJ 2002. Room chamber assessment of pollutant emission properties of low-emission
unflued gas heaters. In: Proceedings of the 9
th
International Conference on Indoor Air Quality and Climate, vol 3, 637-
642.
Bruinen de Bruin Y 2002. Exposure of an urban adult population to carbon monoxide. Direct and indirect exposure
assessment, methods, determinants and sources. Academic dissertation. University of Milan, Department of
Occupational Health, Milan, Italy.
Bruinen de Bruin Y, Carrer P, Jantunen M, Hanninen O, Scotto di Marco G, Kephalopoulos S, Cavallo D, Maroni M
2004. Personal carbon monoxide exposure levels; contribution of local sources to exposures and microenvironment
concentrations in Milan. Jouranl of Expo Analysis and Environmental Epidemiology vol 14, in press.
Bunnell, D.E. & Horvath, S.M. Interactive effects of physical work and carbon monoxide on cognitive task
performance. Aviation, space, and environmental medicine, 59: 1133–1138 1988.
Burnett RT, Smith-Doiron M, Stieb D, Cakmak S, Brook JR. 1999. Effects of particulate and gaseous air pollution on
cardiorespiratory hospitalizations. Arch Environ Health. 1999 Mar-Apr;54 2:130-9.
Chaloulakou A, Mavroidis I, Duci A 2003. Indoor and outdoor carbon monoxide concentration relationships at different
microenvironments in the Athens area. Chemosphere 52:1007-1019.
Coburn RF 1970 Enhancement by phenobarbital and diphenylhydantoin of carbon monoxide production in normal man.
N Engl J Med, 283: 512-515.
Coburn RF, Forster RE, & Kane PB 1965 Considerations of the physiological variables that determine the blood
carboxyhemoglobin concentration in man. J Clin Invest, 44: 1899-1910.
Coburn RF, Williams WJ, & Forster RE 1964 Effect of erythrocyte destruction on carbon monoxide production in man.
J Clin Invest, 43: 1098-1103.
Coburn RF, Williams WJ, & Kahn SB 1966 Endogenous carbon monoxide production in patients with hemolytic
anemia. J Clin Invest, 45: 460-468.
Costa, D.L. and Amdur, M.O. 1996 Air Pollution in Cassarett and Doull’s Toxicology, Klassen, ed. McGraw-Hill. New
York p 878.
Dahms TE, Younis LT, Wiens RD, Zarnegar S, Byers SL, & Chaitman BR 1993 Effects of carbon monoxide exposure
in patients with documented cardiac arrhythmias. J Am Coll Cardiol, 21 2: 442-450.
Delivoria-Papadopoulos M, Coburn RF, & Forster RE 1970 Cyclical variation of rate of heme destruction and carbon
monoxide production Func {V dot}
CO
in normal women. Physiologist, 13: 178.
EC 2000. Official Journal of the European Communities L 313/13 of the 13.12.2000. DIRECTIVE 2000/69/EC OF
THE EUROPEAN PARLIAMENT AND OF THE COUNCIL of 16 November 2000 relating to limit values for
benzene and carbon monoxide in ambient air.
El Fadel M, Alameddine I, Kazopoulo M, Hamdan M, Nasrallah R 2001. Carbon monoxide and volatile organic
compounds as indicators of indoor air quality in underground parking facilities. Indoor Built Environ 10:70-82.
Ellenhorn MJ & Barceloux DG ed 1988 Medical toxicology diagnosis and treatment of human poisoning. New York,
Elsenin, pp 820-829.
EPA 2000. Air quality criteria for carbon monoxide. USEPA EPA 600/P-99/001F. 01 Jun 2000. U.S. Environmental
Protection Agency, Office of Research and Development, National Center for Environmental Assessment, Washington,
DC. Available in the Internet at http://www.epa.gov/ncea/pdfs/coaqcd.pdf.
EPA 2003. Indoor Air Quality. Available accessed in November, 2003 in the Internet at
http://www.epa.gov/iaq/co.html.
EXPOLIS 2002. Combined International Database CIDB, KTL- National Public Health Institute, Kuopio Finland.
Georgoulis LB, Hänninen O, Samoli E, Katsouyanni K, Kuenzli N, Polanska L, Bruinen de Bruin Y, Alm S, Jantunen
M 2002. Personal carbon monoxide exposure in five European cities and its determinants. Atmos Environ 36:963-974.
Grace, T.W. and F.W. Platt, 1981. Subacute carbon monoxide poisoning. J. Am. Med. Assoc. 246, 1698-1700.
Guo H, Lee, SC, Chan LY 2004. Indoor air quality in ice skating rinks in Hong Kong. Environ Respectively 94:327-
335.
Hinderliter AL, Adams KF Jr, Price CJ, Herbst MC, Koch G, & Sheps DS 1989 Effects of low-level carbon monoxide
exposure on resting and exercise-induced ventricular arrhythmias in patients with coronary artery disease and no
baseline ectopy. Arch Environ Health, 44: 89-93.
The INDEX project Final report 284
IEH 1998. Indoor air quality in the home 2: carbon monoxide. Assessment A5. Leicester UK: Institute of of
Environment and Health 1998. p. 4-5, 19, 41.
Jetter J, Guo Z, Jenia, McBrian JA, Flynn MR 2002. Characterization of emissions from burning incense. The Science
of The Total Environment 295:51-67.
Junker M, Koller T, Monn C 2000. An assessment of indoor air contaminants in buildings with recreational activity.
The Science of the Total Environment 246:139-152.
Kirchner S 2004. Preliminary results of the French National Survey on Indoor Air Quality. Personal communication.
Klees, M, M. Heremans and S. Dougan, 1985. Psychological sequelae to carbon monoxide intoxication in the child. Sci.
Total Environm. 44, 165-176.
Kleinman M.T, D.M. Davidson, R.B. Vandagriff, V.J. Caiozzo, and J.L. Whittenberger. 1989. Effects of short-term
exposure to carbon monoxide in subjects with coronary artery disease. Arch. Environ. Health 44:361-369.
Kleinman MT, Leaf DA, Kelly E, Caiozzo V, Osann K, O'Niell T. 1998. Urban angina in the mountains: effects of
carbon monoxide and mild hypoxemia on subjects with chronic stable angina. Arch Environ Health. 1998 Nov-Dec;53
6:388-97.
Laties, V.G. & Merigan, W.H. Behavioral effects of carbon monoxide on animals and men. Annual review of
pharmacology and toxicology, 19: 357–392 1979.
Lebret E, Noy D, Boley J, Brunekreef B 1987. Real-time concentrations of CO and NO
2
in twelve homes. In:
Proceedings of the 4
th
International Conference on Indoor Air Quality and Climate, vol 1, 435-439.
Lee SC, Wang B. 2004. Characteristics of emissions of air pollutants from burning of incense in a large environmental
chamber. Atmospheric Environment 38:941-951.
Leichter, J. 1993. Fetal growth retardation due to exposure of pregnant rats to carbon monoxide. Biochem. Arch. 9:267-
272.
Linn WS, Szlachcic Y, Gong H Jr, Kinney PL, Berhane KT. 2000. Air pollution and daily hospital admissions in
metropolitan Los Angeles. Environ Health Perspect. 2000 May;108 5:427-34.
Longo, L.D. 1977. The biological effects of carbon monoxide on the pregnant woman, fetus, and newborn infant.
American journal of obstetrics and gynecology, 129: 69–103.
Lynch SR & Moede AL 1972 Variation in the rate of endogenous carbon monoxide production in normal human
beings. J Lab Clin Med, 79: 85-95.
Mann JK, Tager IB, Lurmann F, Segal M, Quesenberry CP Jr, Lugg MM, Shan J, Van Den Eeden SK. 2002. Air
pollution and hospital admissions for ischemic heart disease in persons with congestive heart failure or arrhythmia.
Environ Health Perspect. 2002 Dec;110 12:1247-52.
Maroni M, Carrer P, Cavallo D, Alcini D, Basso A, Preti G, Verselli F, Visigalli F, Bersani M 1996. Characterisation of
personal exposure to air pollutants of subjects living in Milan. Proceedings of the 7th International Conference on
Indoor Air Quality and Climate. Nagoya, Japan. 1:501-505.
Maroni M, Seifert B, Lindvall T eds 1995. A Comprehensive Reference Book. Air Quality Monographs Vol. - 3.
Elsevier Science B.V, Amsterdam, The Netherlands, p. 17.
Maynard RL & Waller R 1999 Carbon Monoxide. In: Holgate ST, Samet JM, Koren HS & Maynard RL eds Air
Pollution and Health Academis Press: Harcourt Brace & Company, Publishers, pp. 749-796.
Morris RD, Naumova EN, Munasinghe RL. 1995. Ambient air pollution and hospitalization for congestive heart failure
among elderly people in seven large US cities. Am J Public Health. 1995 Oct;85 10:1361-5.
OEHHA 2000. Office of Environmental Health Hazard Assessment of the Californian Environmental Protection
Agency. Evaluation of current California Air Quality Standards with respect to protection of children available on the
Internet at http://www.oehha.ca.gov/air/pdf/oehhaco.pdf
Peterson, J.E. & Stewart, R.D, 1975, Predicting thecarboxyhemoglobin levels resulting from carbon monoxide
exposures. J. Appl. Physiol, 39,
633-638
Peterson, J.E. Postexposure relationship of carbon monoxide in blood and expired air. Archives of environmental
health, 21: 172–173 1970.
Putz, V.R. The effects of carbon monoxide on dual-task performance. Human factors, 21:13–24 1979.
Raub J.A. and Benignus V.A. 2002. Review: Carbon monoxide and the nervous system. Neuroscience and
Biobehavioral Reviews 26 2002 925–940
Raub J.A, Mathieu-Nolf M, Hampson N.B, Thom S.R. 2000. Review: Carbon monoxide poisoning — a public health
perspective. Toxicology 145 2000 1–14
Raw GJ, Coward SKD, Llewellyn JW, Brown VM, DR Crump, Ross DI 2002. Indoor air quality in English homes –
introduction and carbon monoxide findings. In: Proceedings of the 9
th
International Conference on Indoor Air Quality
and Climate, vol 4, 461-466.
The INDEX project Final report 285
Ritz,B. and Yu,F, 1999, The effect of ambient carbon monoxide on low birth weight among children born in southern
California between 1989 and 1993. Environ Health Perspect 107:17-25.
Rodgers PA, Vreman HJ, Dennery PA, & Stevenson DK 1994 Sources of carbon monoxide CO in biological systems
and applications of CO detection technologies. Semin Perinatol, 18: 2-10.
Ross D. 1996. Continuous monitoring of NO
2
, CO, temperature and humidity in UK homes. In: Proceedings of the 7
th
International Conference on Indoor Air Quality and Climate, vol 1, Tokyo, Institute for Public Health, 513-518.
Schwartz J. 1999. Air pollution and hospital admissions for heart disease in eight U.S. counties. Epidemiology. 1999
Jan;10 1:17-22.
Le Tertre A, Medina S, Samoli E, Forsberg B, Michelozzi P, Boumghar A, Vonk JM, Bellini A, Atkinson R, Ayres JG,
Sunyer J, Schwartz J, Katsouyanni K. 2002. Short-term effects of particulate air pollution on cardiovascular diseases in
eight European cities. J Epidemiol Community Health. 2002 Oct;56 10:773-9.
Scotto di Marco G, Kephalopoulos S,Ruuskanen J. and Jantunen M. 2003 Personal carbon monoxide exposure in
Helsinki, Finland. Submitted to Atmospheric Environment.
Sheps DS, Herbst MC, Hinderliter AL, Adams KF, Ekelund LG, O’Neil JJ, Goldstein GM, Bromberg PA, Dalton JL,
Ballenger MN, Davis SM, & Koch GG 1990 Production of arrhythmias by elevated carboxyhemoglobin in patients with
coronary artery disease. Ann Intern Med, 113: 343-351.
Sheps, D.S, K.F. Adams, P.A. Bromberg, G.M. Goldstein, J.J. O'Neil, D. Horstman, and G. Koch. 1987. Lack of effect
of low levels of carboxyhemoglobin on cardiovascular function in patients with ischemic heart disease, Arch. Environ.
Health 42:108-116.
Smith RP 1986 Toxic Responses of the Blood. In: Klaassen DD, Amadur MO and Doull J eds Casarett and Doulss's
Toxicology, 3rd edn. New York: Mc Millan Publishing Company, pp. 223-240.
Smith, C.J. & Steichen, T.J. The atherogenic potential of carbon monoxide. Atherosclerosis, 99: 137–149 1993.
Solanki DL, McCurdy PR, Cuttitta FF, & Schechter GP 1988 Hemolysis in sickle cell disease as measured by
endogenous carbon monoxide production: a preliminary report. Am J Clin Pathol, 89: 221-225.
Stern FB, Halperin WE, Hornung RW, Ringenburg VL, & McCammon CS 1988 Heart disease mortality among bridge
and tunnel officers exposed to carbon monoxide. Am J Epidemiol, 128: 1276-1288.
Stewart, R.D, J.E. Peterson, E.D. Baretta, R.T. Bachand, M.J. Hosko and A.A. Herrmann, 1970. Experimental human
exposure to carbon monoxide. Arch. Environ. Health 21, 154-164.
Storm, J.E. and L.D. Fechter. 1985. Alteration in the postnatal ontogeny of cerebellar norepinephrine content following
chronic prenatal carbon monoxide. J. Neurochem. 45:965-969.
U.S.EPA 1984. Revised Evaluation of Health Effects Associated with Carbon Monoxide Exposure: An Addendum to
the l979 EPA Air Quality Criteria Document for Carbon Monoxide, EPA Report No. EPA 600/9-83-033F, U.S.
Environmental Protection Agency, Environmental Criteria and Assessment Office, Research Triangle Park, NC, NTIS
No. PB85-103471.
U.S.EPA 1991. Air quality criteria for carbon monoxide. Washington, DC. US Environmental Protection Agency,
Office of Research and Development, 1991 publication no. EPA-600/B-90/045F.
U.S.EPA 2000. Air quality criteria for carbon monoxide. EPA 600/P-99/001F, U. S. Environmental Protection Agency,
Office of Research and Development, Washington, D.C.
Valerio F, Pala M, Lazzarotto A, Balducci D 1997. Preliminary evaluation, using passive tubes of carbon monoxide
concentrations in outdoor and indoor air at street level shops in Genoa Italy. Atmos Environ 31 17:2871-2876.
Varon J, Marik PE, Fromm RE, Gueler A. 1999 Carbon Monoxide Poisoning: A Review for Clinicians. The Journal of
Emergency Med, 17: 87-93.
Verschueren K 2001. Handbook of Environmental Data on Organic Chemicals. Fourth edition. John Wiley & Sons, Inc,
New York. Vol. 1, 24.
WHO 1979. Environmental Health Criteria For Carbon Monoxide 13. WHO, Geneva, 1979. Available in the Internet
http://www.inchem.org/documents/ehc/ehc/ehc013.htm.
WHO 1999. Environmental Health Criteria 213, Carbon Monoxide Second Edition, IPCS, International Programme on
Chemical Safety; World Health Organization, Geneva, Switzerland, 1999.
WHO 2000: Air Quality Guidelines for Europe. WHO Regional Publications, European Series, No. 91, Regional Office
for Europe, Copenhagen.
Winter PM & Miller JN 1976 Carbon monoxide poisoning. J Am Med Assoc, 236: 1503.
Zhang J, Smith KR, Uma R, Ma Y, Kishore VVN, Lata K, Khalil MAK, Rasmussen RA, Thorneloe STYRENE 1999.
Carbon monoxide from cookstoves in developing countries: 2. Exposure potentials, Chemosphere – Global Change
Science 1:367-375.
The INDEX project Final report 286
Nitrogen dioxide
Main sources:
Authority Shortcut Link to original document
WHO- Air Quality Guidelines for
Europe - 2000
[WHO] no Internet link
WHO – IPCS
ENVIRONMENTAL HEALTH
CRITERIA 188
[EHC 188] http://www.inchem.org/documents/ehc/ehc/ehc188.htm
Office of Environmental Health
Hazard Assessment of the
Californian Environmental
Protection Agency
Acute Toxicity summary 1999
[OEHHA1] http://www.oehha.ca.gov/air/acute_rels/pdf/
OEHHA - NITROGEN DIOXIDE:
EVALUATION OF CURRENT
CALIFORNIA AIR QUALITY
STANDARDS WITH RESPECT
TO PROTECTION OF
CHILDREN
[OEHHA2]
http://www.oehha.ca.gov/air/pdf/oehhano2.pdf
U.S.EPA - Integrated Risk
Information System
[IRIS] http://www.epa.gov/iris/subst/0080.htm
AIHA - American Industrial Hygiene Association. Odor thresholds for chemicals with established occupational health
standards. Akron OH: AIHA; 1989. p. 29.
Alm S. 1999. Personal exposures of pre-school children to carbon monoxide and nitrogen dioxide in urban air.
Academic dissertation. Publications of the National Public Health Institute A19/99. National Public Health Institute,
Kuopio, Finland; 1999.
Amoore JE & Hautala E 1983 Odor as an aid to chemical safety: odor thresholds compared with threshold limit values
and volatilities for 214 industrial chemicals in air and water dilution. J Appl Toxicol, 3 6: 272-290.
Anderson, H.R. et al. Air pollution and daily mortality in London: 1987-92. British medical journal, 312: 665–669
1996.
Azadniv, M, Utell, M. J, Morrow, P. E, Gibb, F. R, Nichols, J, Roberts, N. J. Jr, Speers, D. M, Torres, A, Tsai, Y,
Abraham, M. K, Voter, K. Z, and Frampton, M.W. 1998. Effects of nitrogen dioxide exposure on human host defense.
Inhalation Toxicol. 10:585-602.
Bauer, M. A, Utell, M. J, Morrow, P. E, Speers, D. M, and Gibb, F. R. 1986. Inhalation of 0.30 ppm nitrogen dioxide
potentiates exercise-induced bronchospasm in asthmatics. Am Rev Respir Dis. 134:1203-1208.
Berglund M, Braback L, Bylin G, Gustafsson L, Moldeus P, Norberg S, Pershagen G, Victorin K 1994. Personal NO2
exposure monitoring shows high exposure among ice-skating schoolchildren. Arc Environ Health 49: 17-24.
Berglund, M. et al. Health risk evaluation of nitrogen oxides. Scandinavian journal of work, environment and health, 19
Suppl. 2 1993.
Bermudez, E, Ferng, S. F, Castro, C. E, and Mustafa, M. G. 1999. DNA strand breaks caused by exposure to ozone and
nitrogen dioxide. Environ Res. 81:72-80.
Blomberg, A, Krishna, M. T, Bocchino, V, Biscione, G. L, Shute, J. K, Kelly, F. J, Frew, A. J, Holgate, S. T, and
Sandstrom, T. 1997. The inflammatory effects of 2 ppm NO2 on the airways of healthy subjects. Am J Respir Crit Care
Med. 156:418-424.
Brown SK, Cheng M, Mahoney KJ 2002. Room chamber assessment of pollutant emission properties of low-emission
unflued gas heaters. In: Proceedings of the 9
th
International Conference on Indoor Air Quality and Climate, vol 3, 637-
642.
Bylin, G, Lindvall, T, Rehn, T, and Sundin, B. 1985. Effects of short-term exposure to ambient nitrogen dioxide
concentrations on human bronchial reactivity and lung function. European Journal of Respiratory Disease. 66:205-217.
CARB 1992. California Air Resources Board. Review of the one-hour ambient air quality standard for nitrogen dioxide
technical support document. Sacramento: State of California Air Resources Board Technical Support Division;
December 1992.
COMEAP Committee on the medical effects of air pollutants 1997: Handbook on air pollution and health. Department
of health, committee on the medical effects of air pollutants. Her Majesty’s Stationary Office, London, UK.
Coward SKD, Raw GJ, Llewellyn JW, Ross DI 2002. Indoor air quality in English homes – nitrogen dioxide. In:
Proceedings of the 9
th
International Conference on Indoor Air Quality and Climate, vol 4, 467-472.
The INDEX project Final report 287
Dab, W. et al. Short term respiratory health effects of ambient air pollution: results of the APHEA project in Paris.
Journal of epidemiology and community health, 50 Suppl. 1: S42–S46 1996.
Damji, K.S.and Richters, A. 1989. Reduction in T lymphocyte subpopulations following acute exposure to 4 ppm
nitrogen dioxide. Environ Res. 49:217-224.
Evans, M.J, Johnson, L.V, Stephens, R.J, and Freeman, G. 1976. Renewal of the terminal bronchiolar epithelium in the
rat following exposure to NO2 or O3. Lab Invest. 35:246-257.
EXPOLIS 2002. Combined International Database CIDB, KTL- National Public Health Institute, Kuopio Finland.
Farman, C. A, Watkins, K, van Hoozen, B, Last, J. A, Witschi, H, and Pinkerton, K. E. 1999. Centriacinar remodeling
and sustained procollagen gene expression after exposure to ozone and nitrogen dioxide. Am J Respir Cell Mol Biol.
20:303-11.
Frampton, M. W, Smeglin, A. M, Roberts, N. J. Jr, Finkelstein, J. N, Morrow, P. E, and Utell, M. J. 1989. Nitrogen
dioxide exposure in vivo and human alveolar macrophage inactivation of influenza virus in vitro. Environ Res. 48:179-
192.
Garcia-Algar O, Zapater M, Figueroa C, Vall O, Basagana X, Sunyer J, Freixa A, Guardino X, Pichini S 2003. Sources
and Concentrations of Indoor Nitrogen Dioxide in Barcelona, Spain. Atmos Environ 53:1312-1317.
Garrett, M. H, Hooper, M. A, Hooper, B. M, and Abramson, M. J. 1998. Respiratory symptoms in children and indoor
exposure to nitrogen dioxide and gas stoves. Am J Respir Crit Care Med. 158:891-895.
Gavras, J. B, Frampton, M. W, Ryan, D. H, Levy, P. C, Looney, R. J, Cox, C, Morrow, P. E, and Utell, M. J. 1994.
Expression of membrane antigens on human alveolar macrophages after exposure to nitrogen dioxide. Inhalation
Toxicol. 6:633-646.
Goings, S.A.J, Kulle, T.J, Bascom, R, Sauder, L.R, Green, D.J, Hebel, J.R, and Clements, M.L. 1989. Effect of nitrogen
dioxide exposure on susceptibility to influenza A virus infection in healthy adults. Am Rev Respir Dis. 139:1075-1081.
Goldstein E, Peek NF, Parks NJ, Hines HH, Steffey P and Tarkington B. Fate and distribution of inhaled nitrogen
dioxide in rhesus monkeys. Am Rev Respir Dis, 1977; 115: 403-12.
Hänninen OO, Alm S, Katsouyanni K, Künzli N, Maroni M, Nieuwenhuijsen MJ, Saarela K, Srám RJ, Zmirou D,
Jantunen MJ 2004. The EXPOLIS study: implications for exposure research and environmental policy in Europe.
Journal of Exposure Analysis and Environmental Epidemiology, advance online publication, 1-17.
Hasselblad, V, Kotchmar, D. J, and Eddy, D. M. 1992. Synthesis of environmental evidence: nitrogen dioxide
epidemiology studies. Journal of Air & Waste Management. 42:662-671.
Hazenkamp-von Arx ME, Gotschi T, Ackermann-Liebrich U, Bono R, Burney P, Cyrys J, Jarvis D, Lillienberg L,
Luczynska C, Maldonado JA, Jaen A, de Marco R, Mi Y, Modig L, Bayer-Oglesby L, Payo F, Soon A, Sunyer J,
Villani S, Weyler J, Kunzli N 2004. PM
2.5
and NO
2
assessment in 21 European study centres of ECRHS II: annual
means and seasonal differences. Atmos Environ 38:1943-1953.
Hazucha, M.J, Folinsbee, L.J, Seal, E, and Bromberg, P.A. 1994. Lung function response of healthy women after
sequential exposures to NO2 and O3. Am J Respir Crit Care Med. 150:642-647.
Hazucha, M.J, Ginsberg, J.F, McDonnell, W.F, Haak Jr, E.D, Pimmel, R.L, Salaam, S.A, House, D.E, and Bromberg,
P.A. 1983. Effects of 0.1 ppm nitrogen dioxide on airways of normal and asthmatic subjects. J Appl Physiol. 54:730-
739.
HSDB 2003. Hazardous Substances Data Bank. Specialised Information Services. National Library of Medicine.
Available online at http://toxnet.nlm.nih.gov/.
Jarvis, D, Chinn, S, Luczynska, C, and Burney, P. 1996. Association of respiratory symptoms and lung function in
young adults with use of domestic gas applicances. Lancet. 347:426-431.
Johnson, D.A, Frampton, M.W, Winters, R.S, Morrow, P.E, and Utell, M.J. 1990. Inhalation of nitrogen dioxide fails to
reduce the activity of human lung alpha-1-proteinase inhibitor. Am Rev Respir Dis. 142:758-762.
Jorres, R.and Magnussen, H. 1991. The effect of resting ventilation of 0.25 ppm nitrogen dioxide on the airway
response to hyperventilation of sulfur dioxide in smokers. Am Rev Respir Dis. 143 Suppl. April:
Katsouyanni, K. et al. Short term effects of air pollution on health: a European approach using epidemiology time series
data: the APHEA protocol. Journal of epidemiology and community health, 50 Suppl. 1: S12–S18 1996.
Kleinman, M.T, Bailey, R.M, Linn, W.S, Anderson, K.R, Whynot, J.D, Shamoo, D.A, and Hackney, J.D. 1983. Effects
of 0.2 ppm nitrogen dioxide on pulmonary function and response to bronchoprovocation in asthmatics. J Toxicol
Environ Health. 12:815-826.
Koenig, J.Q, Covert, D.S, Morgan, M.S, Horike, M, Horike, N, Marshall, S.G, and Pierson, W.E. 1985. Acute effects of
0.12 ppm ozone or 0.12 ppm nitrogen dioxide on pulmonary function in healthy and asthmatic adolescents. Am Rev
Respir Dis. 132:648-651.
The INDEX project Final report 288
Koenig, J.Q, Pierson, W.E, Covert, D.S, Marshall, S.G, Morgan, M.S, and van Belle, G. 1988. The effects of ozone and
nitrogen dioxide on lung function in healthy and asthmatic adolescents. Health Effects Institute Research Report. 5-24.
Kousa A, Monn C, Rotko T, Alm S, Oglesby L. and Jantunen M. 2001: Personal exposures to NO2 in the Expolis-
study: Relation to residential indoor, outdoor and workplace concentrations in Basel, Helsinki and Prague. Atmos
Environ 35 20:3405-3412.
Kuraitis, K.V.and Richters, A. 1989. Spleen cellularity shifts from the inhalation of 0.25-0.35 ppm nitrogen dioxide.
Journal of Environmental Pathology and Toxicology. 9:1-11.
Lafuma, C, Harf, A, Lange, F, Bozzi, L, Ponci, J.L, and Bignon, J. 1987. Effect of low-level NO2 chronic exposure on
elastase-induced emphysema. Environ Res. 43:75-84.
Lebret E, Noy D, Boley J, Brunekreef B 1987. Real-time concentrations of CO and NO2 in twelve homes. In:
Proceedings of the 4th International Conference on Indoor Air Quality and Climate, vol 1, 435-439.
Lee SC, Wang B. 2004. Characteristics of emissions of air pollutants from burning of incense in a large environmental
chamber. Atmospheric Environment 38:941-951.
Levesque B, Allaire S, Gauvin D, Koutrakis P, Gingras S, Rhainds M, Prud’Homme H, Duchesne J-F 2001. Wood-
burning appliances and indoor air quality. The Science of the Total Environment 281:47-62.
Levy JI, Lee K, Spengler JD, Yanagisawa Y 1998. Impact of Residential Nitrogen Dioxide Exposure on Personal
Exposure: An International Study. JAWMA 48:553-560.
Linn, W. S, Avol, E. L, Shamoo, D. A, Whynot, J. D, Anderson, K. R, and Hackney, J. D. 1986. Respiratory responses
of exercising asthmatic volunteers exposed to sulfuric acid aerosol. J Air Poll Contr Assoc. 36:1323-1328.
Miller, F. J, Overton, J. H, Myers, E. T, and Graham, J. A. 1982. Pulmonary dosimetry of nitrogen dioxide in animals
and man. in Air pollution by nitrogen oxides. Schneider, T.and Grant, L, Eds. pp 377-386, Amsterdam, Elsevier
Scientific Publishing Company.
Mohsenin, V. 1987. Airway responses to nitrogen dioxide in asthmatic subjects. J Toxicol Environ Health. 22:371-380.
Mohsenin, V.and Gee, J.B.L. 1987. Acute effect of nitrogen dioxide exposure on the functional activity of alpha-1-
protease inhibitor in bronchoalveolar lavage fluid of normal subjects. Am Rev Respir Dis. 136:646-650.
Mohsenin, V. 1991. Lipid peroxidation and antielastase activity in the lung under oxidant stress: role of antioxidant
defenses. J Appl Physiol. 70:1456-1462.
Morrow, P. E. and Utell, M. J. Responses of susceptible subpopulations to nitrogen dioxide. 23. 89. Montpelier, VT,
Capital City Press. Health Effects Institute Research Report.
Mukala, K, Pekkanen, J, Tiittanen, P, Alm, S, Salonen, R. O, and Tuomisto, J. 1999. Personally measured weekly
exposure to NO2 and respiratory health among preschool children. Eur Respir J. 13:1411-7.
Neas, L. M, Dockery, D. W, Ware, J. H, Spengler, J. D, Speizer, F. E, and Ferris, B. G. Jr. 1991. Association of indoor
nitrogen dioxide with respiratory symptoms and pulmonary function in children. Am J Epidemiol. 134:204-219.
Noy D, Brunekreef B, Boleij JSM, Houthuijs D, De Koning R 1990. The assessment of personal exposure to nitrogen
dioxidein epidemiological studies. Atmos Environ 24A:2903-2909.
Orehek, J, Massari, J.P, Gayrard, P, Grimaud, C, and Charpin, J. 1976. Effect of short-term, low-level nitrogen dioxide
exposure on bronchial sensitivity of asthmatic patients. J Clin Invest. 57:301-307.
Overton, J.H. et al. Significances of the variability of airway paths and their air flow rates to dosimetry model
predictions of the absorption of gases. Research Triangle Park, NC, US Environmental Protection Agency, 1987, EPA
Report No. EPA-600/D-87-364.
Overton, J.H, Jr. Physicochemical processes and the formulation of dosimetry models. In: Miller, F.J. & Menzel, D.B,
ed. Fundamentals of extrapolation modeling of inhaled toxicants: ozone and nitrogen dioxide. Washington, DC,
Hemisphere, 1984, pp. 93–114.
Pennanen A, Salonen RO, Alm S, Jantunen M, Pasanen P 1998. Characterization of air quality problems in five finnish
indoor ice arenas. JAWMA 47:1079-1086.
Pennanen A, Vahteristo M, Salonen RO 1998. Contribution of techinal and operational factors to nitrogen dioxide
concentration in indoor ice arenas. Env Int 24:381388.
Pilotto, L. S, Douglas, R. M, Attewell, R. G, and Wilson, S. R. 1997. Respiratory effects associated with indoor
nitrogen dioxide exposure in children. Int J Epidemiol. 26:788-96.
Pönkä, A. & Virtanen, M. Asthma and ambient air pollution in Helsinki. Journal of epidemiology and community
health, 50 Suppl. 1: S59–S62 1996.
Pönkä, A. Asthma and low level air pollution in Helsinki. Archives of environmental health, 46: 262–270 1991.
Postlethwait EM and Bidani A. Pulmonary disposition of inhaled NO2-nitrogen in isolated rat lungs. Toxicol Appl
Pharmacol, 1989; 98: 303-312.
The INDEX project Final report 289
Postlethwait, E. M, Langford, S. D, and Bidani, A. 1990. Reactive absorption of nitrogen dioxide by pulmonary
epithelial lining fluid. J Appl Physiol. 68:594-603.
Rajini, P, Gelzleichter, T. R, Last, J. A, and Witschi, H. 1993. Alveolar and airway cell kinetics in the lungs of rats
exposed to nitrogen dioxide, ozone, and a combination of the two gases. Toxicol Appl Pharmacol. 121:186-192.
Richters, A.and Damji, K.S. 1988. Changes in T-lymphocyte subpopulations and natural killer cells following exposure
to ambient levels of nitrogen dioxide. J Toxicol Environ Health. 25:247-256.
Richters, A.and Richters, V. 1989. Nitrogen dioxide NO2 inhalation, formation of microthrombi in lungs and cancer
metastasis. Journal of Environmental Pathology and Toxicology. 9:45-51.
Roger, L.J, Horstman, D.H, McDonnell, W, Kehrl, H, Ives, P.J, Seal, E, Chapman, R, and Massaro, E. 1990. Pulmonary
function, airway responsiveness, and respiratory symptoms in asthmatics following exercise in NO2. Toxicol Ind
Health. 6:155-171.
Ross D. 1996. Continuous monitoring of NO
2
, CO, temperature and humidity in UK homes. In: Proceedings of the 7
th
International Conference on Indoor Air Quality and Climate, vol 1, Tokyo, Institute for Public Health, 513-518.
Rubinstein, I, Bigby, B.G, Reiss, T.F, and Boushey, H.A.Jr. 1990. Short-term exposure to 0.3 ppm nitrogen dioxide
does not potentiate airway responsiveness to sulfur dioxide in asthmatic subjects. Am Rev Respir Dis. 141:381-385.
Rubinstein, I, Reiss, T.F, Bigby, B.G, Stites, D.P, and Boushey, H.A.Jr. 1991. Effects of 0.60 PPM nitrogen dioxide on
circulating and bronchoalveolar lavage lymphocyte phenotypes in healthy subjects. Environ Res. 55:18-30.
Sakai K, Norback D, Mi Y, Shibata E, Kamijima M, Yamada T, Yasuhiro T 2004. A comparison of indoor air
pollutants in Japan and Sweden: formaldehyde, nitrogen dioxide, and chlorinated volatile organic compounds. Environ
Res 94:75-85.
Samet, J. M, Lambert, W. E, Skipper, B. J, Cushing, A. H, Hunt, W. C, Young, S.A, McLaren, L. C, Schwab, M, and
Spengler, J. D. 1993. Nitrogen dioxide and respiratory illnesses in infants. Am Rev Respir Dis. 148:1258-1265.
Sandstrom, T, Helleday, R, Bjermer, L, and Stjernberg, N. 1992a. Effects of repeated exposure to 4 ppm nitrogen
dioxide on bronchoalveolar lymphocyte subsets and macrophages in healthy men. Eur Respir J. 5:1092-1096
Sandstrom, T, Ledin, M.C, Thomasson, L, Helleday, R, and Stjernberg, N. 1992b. Reductions in lymphocyte
subpopulations after repeated exposure to 1.5 ppm nitrogen dioxide. Br J Ind Med. 49:850-854.
Sandstrom, T, Stjernberg, N, Eklund, A, Ledin, M. C, Kolmodin-Hedman, B, Lindstrom, K, Rosenhall, L, and
Angstrom, T. 1991. Inflammatory cell response in bronchoalveolar lavage fluid after nitrogen dioxide exposure of
healthy subjects: a dose-response study. Eur Respir J. 4:332-339.
Saul RL and Archer MC. Nitrate formation in rats exposed to nitrogen dioxide. Toxicol Appl Pharmacol, 1983; 67: 284-
291.
Schouten, J.P. et al. Short term effects of air pollution on emergency hospital admissions for respiratory disease: results
of the APHEA project in two major cities in the Netherlands, 1977–89. Journal of epidemiology and community health,
50 Suppl. 1: S22–S29 1996.
Simoni M, Carrozzi L, Baldacci S, Scognamiglio A, Angino A, Pistelli F, Di Pede F, Viegi G 2002. Acute respiratory
effects of indoor pollutants in two general population samples living in a rural and in an urban area of Italy. In:
Proceedings of the 9
th
International Conference on Indoor Air Quality and Climate, vol 1, 119-124.
Spengler, J.D, Brauer, M, and Koutrakis, P. 1990. Acid air and health. Environ Sci Technol. 24:946-956.
Spix, C. & Wichmann, H.E. Daily mortality and air pollutants: findings from Köln, Germany. Journal of epidemiology
and community health, 50 Suppl. 1: S52–S58 1996.
Strand, V, Salomonsson, P, Lundahl, J, and Bylin, G. 1996. Immediate and delayed effects of nitrogen dioxide exposure
at an ambient level on bronchial responsiveness to histamine in subjects with asthma. Eur Respir J. 9:733-740.
U.S.EPA 1993. Air quality criteria for oxides of nitrogen. Research Triangle Park, NC, US Environmental Protection
Agency 1993 EPA Report No. EPA/600/8-91/049aF-cF. 3v.
VDI 1985. Verein Deutscher Ingenieure. Maximale Immissions-Werte zum Schutze des Menschen: Maximale
Immissions-Konzentrationen für Stickstoffdioxid [Maximum emission values for the protection of human health:
maximum emmission concentrations for nitrogen dioxide]. In: VDI-Handbuch Reinhaltung der Luft, Vol. 1. Düsseldorf,
VDI-Verlag, 1985, Part 12.
Verschueren K 2001. Handbook of Environmental Data on Organic Chemicals. Fourth edition. John Wiley & Sons, Inc,
New York. Vol. 1, 24.
Victorin, K. Review of the genotoxicity of nitrogen oxides. Mutation research,. 317: 43–55 1994.
Wagner, H. M. Update of a study for establishing criteria dose/effect relationships for nitrogen oxides. Luxembourg,
Office for Official Publications of the European Communities, 1985 Report No. EUR 9412 EN.
The INDEX project Final report 290
WHO 1997. Environmental Health Criteria. Nitrogen Oxides, 2nd ed. 188. WHO, Geneva, 1997. Available in the
Internet http://www.inchem.org/documents/ehc/ehc/ehc188.htm.
WHO 2000. Air Quality Guidelines for Europe. WHO Regional Publications, European Series, No. 91, Regional Office
for Europe, Copenhagen.
Witschi, H. Ozone, nitrogen dioxide and lung cancer: a review of some recent issues and problems. Toxicology, 48: 1–
20 1988.
Zmirou, D. et al. Short term effects of air pollution on mortality in the city of Lyon, France, 1985-90. Journal of
epidemiology and community health, 50 Suppl. 1: S30–S35 1996.
The INDEX project Final report 291
Benzene
Main sources:
Organization Shortcut Document link
WHO- Air Quality Guidelines for Europe - 2000 [WHO] no Internet link
EPA - Integrated Risk Information System 2003 [IRIS] http://www.epa.gov/iris/subst/0276.htm
Office of Environmental Health Hazard
Assessment of the Californian Environmental
Protection Agency
Acute and Chronic Toxicity summary 1999
[OEHHA] http://www.oehha.ca.gov/
OEHHA: Benzene In Drinking Water 2001
OEHHA: Reproductive Toxicity for Benzene 2001
OEHHA: Cancer Potencies and NSRLs for
Benzene 2003
Agency for Toxic Substances and Disease
Registry
[ATSDR] http://www.atsdr.cdc.gov/phshome.html
European Chemical Bureau - Institute for Health
and Consumer Protection IHCP – draft 2003
[ECB] http://ecb.jrc.it
EPA - Toxicological review of benzene –
noncancer effects 2002
[EPA1] http://www.epa.gov/iris/toxreviews/0276-tr.pdf
EPA - Carcinogenic Effects of Benzene: An
Update 1998
[EPA2] http://www.epa.gov/ncea/pdfs/benzenef.pdf
ATSDR 1989. Toxicological profile for benzene. Atlanta, Georgia, Agency for Toxic Substances and Disease Registry,
173 pp ATSDR/TP-88/03; PB/89/209464/AS.
ATSDR 1991. Toxicological profile for benzene. Atlanta, Georgia, Agency for Toxic Substances and Disease Registry,
193 pp.
ATSDR Agency for Toxic Substances and Disease Registry. 1997 Toxicological profile for benzene update. Public
Health Service, U.S. Department of Health and Human Services, Atlanta, GA.
Bolcsak LE, Nerland DE. Inhibition of erythropoiesis by benzene and benzene metabolites. Toxicol Appl Pharmacol
1983;69:363-368.
Brown SK 2002. Volatile organic pollutants in new and established buildings in Melbourne, Australia. Indoor Air
12:55-63.
Brown VM, Coward SKD, Crump DR, Llewellyn JW, Mann HS, Raw GJ 2002. Indoor air quality in English homes –
VOCs. In: Proceedings of the 9th International Conference on Indoor Air Quality and Climate, vol 4, 477-482.
Carrer P, Maroni M, Alcini D, Cavallo D, Fustinoni S, Lovato L, Visigalli F 2000. Assessment through environmental
and biological measurements of total daily exposure to volatile organic compounds of office workers in Milan, Italy.
Indoor Air 10 4, 258-268.
Coate WB, Hoberman AM, Durloo RS. Inhalation teratology study of benzene in rats. In: MacFarland HN, editor.
Advances in modern environmental toxicology, Vol VI. Applied toxicology of petroleum hydrocarbons. Princeton NJ:
Princeton Scientific Publishers, Inc; 1984. p. 187-198.
Cocheo V, Sacco P, Boaretto C, de Saeger E, Ballesta P, Skov H, Goelen E, Gonzalez N, Caracena A 2000. Urban
benzene and population exposure. Nature 404:141.
Dowty, BJ; Laseter, JL; Storer, J. 1976 The transplacental migration and accumulation in blood of volatileorganic
constituents. Pediatr Res 10:696-701.
Edwards R, Jantunen M 2001. Benzene exposure in Helsinki, Finland. Atmos Environ 35:1411-1420.
Edwards R, Jurvelin J, Saarela K, Jantunen M. 2001. VOC concentrations measured in personal samples and residential
indoor, outdoor and workplace microenvironments in EXPOLIS-Helsinki, Finland. Atmos Environ 35:4531-4543.
Elke K, Jermann E, Begerow J, Dunemann L 1998. Determination of benzene, toluene, ethylbenzene and xylenes in
indoor air at environmental levels using diffusive samplers in combination with headspace solid-phase microextraction
and high-resolution gas chromatography-flame ionization detection. J of Chromatography A 826:191-200.
EPA. 1988e. Recommendations for and documentation of biological values for use in risk assessment. Environmental
Criteria and Assessment Office, Office of Health and Environmental Assessment, Environmental Protection Agency,
Cincinnati, OH. NTIS No. PB88-179874.
The INDEX project Final report 292
EPA. 1990h. Interim methods for development of inhalation reference concentrations. Office of Research and
Development. U. S. Environmental Protection Agency. EPA 600 8-90/066A.
Erexson GL, Wilmer JL, Steinhagen WH, Kligerman AD. Induction of cytogenetic damage in rodents after short-term
inhalation of benzene. Environ Mutagen 1986;8:29-40.
EU 2003. A draft Risk Assessment Report RAR on benzene. Available at http://ecb.jrc.it/existing-chemicals/.
EXPOLIS 2002. Combined International Database CIDB, KTL- National Public Health Institute, Kuopio Finland.
Farris GM, Robinson SN, Gaido KW, Wong BA, Wong VA, Hahn WP, Shah R. Benzeneinduced hematotoxicity and
bone marrow compensation in B6C3F1 mice. Fundam Appl Toxicol 1997;36 2:119-129.
Farris GM, Robinson SN, Gaido KW, Wong BA, Wong VA, Leonard L, Shah R. Effects of low concentrations of
benzene on mouse hematopoietic cells in vivo: a preliminary report. Environ Health Perspect 1996;104 6:1275-1276.
GDCh Society of German Chemists 1988 [Advisory Committee on Existing Chemicals of Environmental Relevance.
Benzene.] Weinheim, VCH Verlagsgesellschaft, 82 pp BUA Report 24 in German with English summary.
Golliot F, Annesi-Maesano I, Delmas M.C. et al. 2003. The French National Survey on Indoor Air Quality: sample
survey design. Proc Healthy building 7th International Conference 2003;Vol 3; p712-717
Green JD, Snyder CA, LoBue J, Goldstein BD, Albert RE. Acute and chronic dose/response effect of benzene
inhalation on the peripheral blood, bone marrow, and spleen cells of CD-1 male mice. Toxicol Appl Pharmacol
1981;59:204-214.
Greenlee WF, Sun JD, Bus JS. A proposed mechanism of benzene toxicity: formation of reactive intermediates from
polyphenol metabolites. Toxicol Appl Pharmacol 1981;59:187-195.
Haley TJ. Evaluation of the health effects of benzene inhalation. Clin Toxicol 1977;11 5:531-548.
Hattemer-Frey HA, Travis CC, & Land ML 1990 Environmental partitioning and human exposure. Environ Res, 53:
221-232.
Henderson, R.F, Sabourin, P.J, Bechtold, W.E, Griffith, W.D, Medinsky, M.A, Birnbaum, L.S, Lucier, G. 1989: The
effect of dose, dose rate, route of administration, and species on tissue and blood levels of benzene metabolites.
Environ. Health Persp. 82: 9-17
Hoffmann K, Krause C, Seifert B, Ullrich D. The German Environmental Survey 1990/1992 GerES II 2000: Sources of
Personal Exposure to Volatile Organic Compounds. J. Exp. And Environ. Epid. 10: 115-125.
Holmberg B, and Lundberg P. 1985. Benzene: Standards, occurrence, and exposure. Am. J. Ind.
HSDB 2003. Hazardous Substances Data Bank. Specialised Information Services. National Library of Medicine.
Available online at http://toxnet.nlm.nih.gov/.
IARC - International Agency for Research on Cancer. IARC monographs on the evaluation of the carcinogenic risk of
chemicals to humans. Vol. 29. Some industrial chemicals and dyestuffs. Lyon: IARC; 1982. p. 93-148.
IARC 1989. Occupational exposures in petroleum refining; crude oil and major petroleum fuels. Lyon, International
Agency for Research on Cancer, 322 pp IARC Monographs on the Evaluation of Carcinogenic Risks to Humans,
Volume 45.
Irons, R.D. 1992. Benzene and other hemotoxins Chapter 63. In: Hazardous Materials Toxicology, Clinical Principles
of Environmental Health, J. B. Sullivan, Jr. and G. R. Krieger eds. Baltimore, MD: Williams & Wilkins, pp. 718-728,
1992.
Jantunen MJ, Katsouyanni K, Knöppel H, Künzli N, Lebret E, Maroni M, Saarela K, Srám R, Zmirou D. 1999. Final
Report: Air Pollution Exposure in European Cities: the EXPOLIS Study. Kuopio, Finland: National Public Health
Institute B16, 1999 Annex II: Table 18.
Keller KA, Snyder CA. Mice exposed in utero to 20 ppm benzene exhibit altered numbers of recognizable
hematopoeitic cells up to seven weeks after exposure. Fundam Appl Toxicol 1988;10:224-232.
Keller KA, Snyder CA. Mice exposed in utero to low concentrations of benzene exhibit enduring changes in their
colony forming hematopoietic cells. Toxicology 1986;42:171-181.
Kirchner S, Pasquier N, Derbez M,Golliot F, Ramalho O,Iannaccone C,Cochet C. 2004. Rapport exécutif Démarrage de
la campagne nationale dans les logements », Rapport CSTB N° DDD/SB 2004-03, Juillet 2004.
Kuna R, Kapp RW. The embryotoxic/teratogenic potential of benzene vapor in rats. Toxicol Appl Pharmacol
1981;57:1-7.
Lee SC, Wang B. 2004. Characteristics of emissions of air pollutants from burning of incense in a large environmental
chamber. Atmospheric Environment 38:941-951.
Med. 7:375-383.
Murray FJ, John JA, Rampy L, Kuna RA, Schwetz BA. Embryotoxicity of inhaled benzene in mice and rabbits. Am Ind
Hyg Assoc J 1979;40:993-998.
The INDEX project Final report 293
Nielsen GD, Alarie Y. Sensory irritation, pulmonary irritation, and respiratory stimulation by alkyl benzene and
alkylbenzenes: prediction of safe industrial exposure levels and correlation with their thermodynamic properties.
Toxicol Appl Pharmacol 1982;65:459-477.
OEHHA 1999. Office of Environmental Health Hazard Assessment of the Californian Environmental Protection
Agency Acute and Chronic Toxicity summary. Available in the Internet at http://www.oehha.ca.gov/
.
OEHHA 2001. Reproductive Toxicity for Benzene. Available in the Internet at http://www.oehha.ca.gov/
.
Pekari, K; Vainiotalo, S; Heikkila, P; et al. 1992 Biological monitoring of occupational exposure to low levels of
benzene. Scand J Work Environ Health 18:317-322.
Reprotext® System. Dabney BJ, editor. Denver CO: Micromedex, Inc.; 1993. Edition expires 11/31/93.
Rickert, DE; Baker, TS; Bus, JS; et al. 1979 Benzene disposition in the rat after exposure by inhalation. Toxicol Appl
Pharmacol 49:417-423.
RIVM 1988 Integrated criteria document on benzene. Bilthoven, The Netherlands, National Institute for Public Health
and Environmental Protection, 138 pp Report No. 758476003.
Rosenthal GJ, Snyder CA. Modulation of the immune response to Listeria monocytogenes by benzene inhalation.
Toxicol Appl Pharmacol 1985;80:502-510.
Ross, D. 1996 Metabolic basis of benzene toxicity. Eur J Haematol 57:111-118.
Rozen MG, Snyder CA, Albert RE. 1984. Depression in B- and T-lymphocyte mitogen-induced blastogenesis in mice
exposed to low concentrations of benzene. Toxicol Lett 20:343-349.
Sawahata, T, Rickerts, D.E, Greenlee, W.F. 1985: Metabolism of benzene and its metabolites in bone marrow. In
Toxicology of the blood and bone marrow, RD Irons, editor, Raven Press New York
Schneider P, Gebefugi IL, Richter K, Wolke G, Schnelle J, Wichmann HE, Heinrich J, INGA study group 2001. Indoor
and outdoor BTX levels in German cities. The Science of the Total Environment 267:41-51.
Schneider P, Lorinci G, Gebefugi IL, Heinrich J, Kettrup A, Wichmann HE 1999. Vertical and horizontal variability of
volatile organic compounds in homes in Eastern Germany. J Exposure Anal Environ Epidemiol 9:282-292.
SIS 2003 Specialised Information Services. US National Library of Medicine. ChemlPlus Record. Available in
http://chem2.sis.nlm.nih.gov/chemidplus/jsp/chemidlite/ChemInfo.jsp?type=notes
last updated October 1, 2003 .
Smith, M.T, Yager, J.W, Steinmetz, K.L, Eastmond, D.A. 1989: Peroxidase-dependent metabolism of benzene`s
phenolic metabolites and its potential role in benzene toxicity and carcinogenicity. Environ. Health Persp. 82: 23-29
Snyder, R, Dimitriadis, E, Guy, R. et al. 1989: Studies on the mechanisms of benzene toxicity. Environ. Health Persp.
82: 31-35
Snyder, R; Hedli, CC. 1996 An overview of benzene metabolism. Environ Health Perspect 104 suppl 6:1165-1171.
Snyder, R; Witz, G; Goldstein, BD. 1993b The toxicology of benzene. Environ Health Perspect 100:293-306.
Tatrai E, Ungvary GY, Hudak A, Rodics K, Lorincz M, Barcza GY. Concentration dependence of the embryotoxic
effects of benzene inhalation in CFY rats. J Hyg Epidemiol Microbiol Immunol 1980;24 3:363-371.
Ullrich D, Gleue C, Krause C, Lusansky C, Nagel R, Schultz C, Seifert B 2002. German Environmental Survey of
children and teenagers 2000 GerES IV: a representative population study including indoor pollutants. In: Proceedings of
the 9
th
International Conference on Indoor Air Quality and Climate, vol 1, 209-214.
Valentine, J.L, Lee, S.S.-T, Seaton, M.J, Asgharian, B, Farris, G, Corton, J.C, Gonzales, F.J, Medinsky, M. 1996:
Reduction of Benzene Metabolism and Toxicity in Mice That Lack CYP2E1 Expression. Toxicol. and Appl.
Pharmacol. 141: 205-213
Verschueren K 2001. Handbook of Environmental Data on Organic Chemicals. Fourth edition. John Wiley & Sons, Inc,
New York. Vol. 1, 24.
Wallace LA 1989. Major sources of benzene exposure. Environ Health Perspect, 82: 165-169.
Watanabe KH, Bois FY, Daisey JM, Auslander DM, Spear RC. Benzene toxicokinetics in humans: exposure of bone
marrow to metabolites. Occup Environ Med 1994;51 6:414-420.
WHO 1979. Carbon monoxide - Environmental Health Criteria, No. 13. World Health Organization, Geneva
WHO 1993. Environmental health criteria 150, benzene. WHO, Geneva, 1995. Available in the Internet
http://www.inchem.org/documents/ehc/ehc/ehc150.htm.
WHO 2001. World Health Organization, Air quality guidelines for Europe 2000. WHO Regional Office for Europe,
WHO Regional Publications, European Series, No. 91, Copenhagen,2001.
Witz, G; Z. Zhang, Z; Goldstein, BD. 1996 Reactive ring-opened aldehyde metabolites in benzene hematotoxicity.
Environ Health Perspect 104 suppl 6:1195-1199.
The INDEX project Final report 294
Naphthalene
Main sources:
Authority Shortcut Link to original document
Environmental Protection Agency [EPA] http://www.epa.gov/safewater/ccl/pdf/naphthalene.pdf
Environmental Protection Agency - Integrated
Risk Information System
[IRIS] http://www.epa.gov/iris/subst/0436.htm
Office of Environmental Health Hazard
Assessment of the Californian Environmental
Protection Agency
[OEHHA] http://www.oehha.ca.gov/air/chronic_rels/pdf/91203.pdf
European Chemical Bureau
Risk Assessment Report: Naphthalene EUR
20763 EN
[ECB] http://ecb.jrc.it/
Agency for Toxic Substances and Disease
Registry
[ATSDR] http://www.atsdr.cdc.gov/toxprofiles/tp67.html
Abdo KM, Grumbein S, Chou BJ, Herbert R. 2001. Toxicity and carcinogenicity study in F344 rats following 2 years of
whole-body exposure to naphthalene vapors. Inhal Toxicol. 2001 Oct;13 10:931-50.
ACGIH. 2000. Threshold Limit Values for Chemical Substances and Physical Agents and Biological Exposure Indices.
American Conference of Government Industrial Hygienists, Cincinnati, OH.
Adkins, B, E.W. Van Stee, J.E. Simmons, et al. 1986. Oncogenic response of strain A/J mice to inhaled chemicals. J.
Toxicol. Environ. Health 17:311-322 as cited in U.S. EPA, 1998a.
Amoore JE and Hautala E 1983. Odor as an Aid to Chemical Safety: Odor Thresholds Compared with Threshold Limit
Values and Volatiles for 214 Industrial Chemicals in Air and Water Dilution. J. Appl. Toxicol. 3 6, 272-290.
Anziulewicz JA, Dick HJ, Chiarulli EE. 1959. Transplacental naphthalene poisoning. Am J Obstet Gynecol 78:519-521.
Athanasious M, Tsantali C, Trachana M, et al. 1995. Hemolytic anemia in a female newborn infant whose mother
inhaled naphthalene before delivery. J Pediatr 130 4:680-681.
ATSDR. 1995. Agency for Toxic Substances and Disease Registry. Toxicological Profile for Naphthalene, Department
of Health and Human Services. CRC Press, Boca Raton, FL.
ATSDR. 2003 Agency for Toxic Substances and Disease Registry. Toxicological profile for naphthalene 1-
methylnapthalene 2-methylnapthalene. Atlanta, GA: U.S. Department of Health and Human Services, Public Health
Service. Available on the Internet at: http://www.atsdr.cdc.gov/phshome.html
Beutler E, Westwood B, Melemed A, Dal Borgo P, Margolis D: Three new exon 10 glucose-6-phosphate
dehydrogenase mutations. Blood Cells Mol Dis 21:64, 1995
Beutler E.1994. G6PD deficiency. Blood 84:3613,
Beutler, E. 1991. Glucose-6-phosphate dehydrogenase deficiency. New Engl. J. Med. 324 3:169-174.
Bieniek, G. 1994. The presence of 1-naphthol in the urine of industrial workers exposed to naphthalene. Occup.
Environ. Med. 51:357-359 as cited in ATSDR, 1995.
Brown VM, Cockram AH, Crump DR and Gardiner D 1990. Investigations of the volatile organic compound content of
indoor air in homes with an odorous damp proof membrane. In: Proc. 'Indoor Air '90', Toronto, Vol. 3, 575-580.
CCOHS. 2003. Canadian Centre for Occupational Health & Safety. CHEMINFO database. Available on the Internet at:
http://www.ccohs.ca/
Chan CC Vainer L, Martin JW, Williams DT 1990. Determination of Organic Contaminants in Residential Indoor Air
Using an Adsorption-thermal Desorption Technique. J Air Waste Manage Assoc 40: 62-67.
CRC 1995. CRC Handbook of Chemistry and Physics, 75th edition. Lide DR, ed. Boca Raton,
Dean BS, Lopez G, Krenzelok EP. 1992. Environmentally-induced methemoglobinemia in an infant. Clin Toxicol
30:127-133.
DeBortoli M, Knöppel H, Pecchio E, Peil A, Rogora L, Schauenburg H, Schlitt H, Vissers H 1986. Concentrations of
selected organic pollutants in indoor and outdoor air in Northern Italy Environ Int 12: 343-350.
Devos M, Patte F, Rouault J, Laffort P, Van Gemert L.J, 1990. Standardized Human Olfactory Thresholds. IRL Press,
Oxford.
ECB 2003. European Chemical Bureau - European Union Risk Assessment Report: Naphthalene EUR 20763 EN.
Office for Official Publications of the European Communities, 2003 Available on the Internet at: http://ecb.jrc.it/
The INDEX project Final report 295
EPA/California 2003, Chronic Reference Exposure Levels Adopted Office of Environmental Health Hazard
Assessment OEHHA as of August 2003. Available in the Internet
http://www.oehha.ca.gov/air/chronic_rels/AllChrels.html
.
EU 2003. European Union Risk Assessment Report. Naphthalene. European Chemicals Bureau. Joint Research Centre.
Institute for Health and Consumer Protection. Existing Substances, 1st Priority List. EUR 20763 EN, Volume: 33. p.
230.
EXPOLIS 2002. Combined International Database CIDB, KTL- National Public Health Institute, Kuopio Finland.
Fait DW, Nachreiner RW. 1985. Naphthalene acute inhalation toxicity study. Report to Texaco, Inc, Beacon, NY, by
Bushy Run Research Center, Union Carbide, Export, PA. Project No. 48-511.
Hoffmann K, Krause C, Seifert B, Ullrich D. The German Environmental Survey 1990/1992 GerES II: Sources of
Personal Exposure to Volatile Organic Compounds. J. Exp. And Environ. Epid. 2000; 10: 115-125.
Horning, M.G, W.G. Stillwell, G.W. Griffin, et al. 1980. Epoxide intermediates in the metabolism of naphthalene by the
rat. Drug Metab. Dispos. 8:404-414 as cited in ATSDR, 1995.
HSDB 2003. Hazardous Substances Data Bank. Specialised Information Services. National Library of Medicine.
Available online at http://toxnet.nlm.nih.gov/.
Huntingdon Research Centre 1993a. Naphthalene 13-week inhalation study in rats. Report LDA 2/930704 unpublished,
as cited in ECB, 2003.
Huntingdon Research Centre 1993b. Naphthalene 4-week inhalation study in rats. Report LDA 1/921559 unpublished,
as cited in ECB, 2003.
IARC. 2002. Naphthalene. IARC Monogr Eval Carcinog Risks Hum 82:367-435.
Iyer P, Martin JE, Irvin TR. 1991. Role of biotransformation in the in vitro preimplantation embryotoxicity of
naphthalene. Toxicology 66:257-270.
Jantunen MJ, Katsouyanni K, Knöppel H, Künzli N, Lebret E, Maroni M, Saarela K, Srám R, Zmirou D. Final Report:
Air Pollution Exposure in European Cities: the EXPOLIS Study. Kuopio, Finland: National Public Health Institute B16,
1999 Annex II: Table 18.
Kanekal, S, C. Plopper, D. Morin, et al. 1990. Metabolic activation and bronchiolar Clara cell necrosis from
naphthalene in the isolated perfused mouse lung. J. Pharmacol. Exp. Ther. 252:428-437 as cited in ATSDR, 1995.
Kostiainen R. Volatile Organic Compounds in the Indoor Air of Normal and Sick Houses. Atmos Environ 1995; 29:
693-702.
Linick M. 1983. Illness associated with exposure to naphthalene in mothballs - Indiana. MMWR 32:34-35.
Long PH, Herbert RA, Peckham JC, Grumbein SL, Shackelford CC, Abdo K. 2003. Morphology of nasal lesions in
F344/N rats following chronic inhalation exposure to naphthalene vapors. Toxicol Pathol. 2003 Nov-Dec;31 6:655-64.
Luzzatto, L. and A. Mehta. 1989. Glucose-6-phosphate dehydrogenase deficiency. In: the Metabolic Basis of Inherited
Disease. Scriver, C, Beaudet, A.L, Sly, W.S, and D. Valle, eds. New York: McGraw Hill Information Services Co.
pp.2237-2239.
Maroni M, Seifert B, Lindvall T eds 1995. A Comprehensive Reference Book. Air Quality Monographs Vol. - 3.
Elsevier Science B.V, Amsterdam, The Netherlands.
New Jersey Department of Health and Senior Services – Hazardous Substances Fact Sheets available on web
http://www.state.nj.us/health/eoh/rtkweb
.
NIH Publication No. 92-3141. Institute of Environmental Health Sciences, Research Triangle Park, NC. NTIS PB92-
219831.
NTP. 1980a. Subchronic toxicity study: Naphthalene C52904, B6C3F1 mice. Research Triangle Park, NC: U.S.
Department of Health and Human Services, National Toxicology Program.
NTP. 1980b. Subchronic toxicity study: Naphthalene C52904, Fischer 344 rats. Research Triangle Park, NC: U.S.
Department of Health and Human Services, National Toxicology Program.
NTP. 1991. Developmental toxicity of naphthalene CAS No. 91-20-3 administered by gavage to Sprague-Dawley CD
rats on gestational days 6 through 15. Final study report and appendix. Research Triangle Park, NC: National
Toxicology Program, National Institute of Environmental Health Sciences, U.S. Department of Health and Human
Services, Public Health Service, National Institutes of Health. TER91006.
NTP. 1992a. Toxicology and carcinogenesis studies of naphthalene CAS no. 91-20-3 in B6C3F1 mice inhalation
studies. National Toxicology Program, U.S. Department of Health and Human Services, National Institutes of Health,
Rockville, MD. Technical report series No. 410.
NTP. 1992b. Developmental toxicity of naphthalene CAS No. 91-20-3 administered by gavage to New Zealand white
rabbits on gestational days 6 through 9. Research Triangle Park, NC: National Toxicology Program, National Institute
The INDEX project Final report 296
of Environmental Health Sciences, U.S. Department of Health and Human Services, Public Health Service, National
Institutes of Health. TER91021.
NTP. 2000. Toxicology and carcinogenesis studies of naphthalene CAS No. 91-20-3 in F344/N rats inhalation studies.
National Toxicology Program. NTP TR 500, NIH Publ. No. 01-4434.
NTP. 2002. Report on Carcinogens Background Document for Naphthalene. Durham, NC: Technology Planning and
Management Corporation. http://ntpserve
. niehs.nih.gov/newhomeroc/rocII/naphthalenePub.pdf. June 6, 2002.
O'Brien, KAF; Smith, LL; Cohen, GM. 1985 Differences in naphthalene-induced toxicity in the mouse and rat. Chem
Biol Interact 55 1-2:109-122.
Pellizzari ED, Hartwell TD, Benjamin SH, et al. 1982. Purgeable organic compounds in mother's milk. Bull Environ
Contam Toxicol 28:322-328.
Plasterer MR, Bradshaw WS, Booth GM, et al. 1985. Developmental toxicity of nine selected compounds following
prenatal exposure in the mouse: Naphthalene, p-nitrophenol, sodium selenite, dimethyl phthalate, ethylenethiourea and
four glycol ether derivatives. Toxicol Environ Health 15:25-38.
Plopper, CG; Suverkropp, C; Morin, D; et al. 1992 Relationship of cytochrome P-450 activity to Clara cell cytotoxicity.
I. Histopathologic comparison of the respiratory tract of mice, rats and hamsters after parenteral administration of
naphthalene. J Pharmacol Exp Ther 261 1:353-363.
PRI. 1985. Dose-range-finding-developmental toxicity in rabbits: Naphthalene. Waverly, PA: Pharmakon Research
International, Inc. PH 329DR-TX-001-85.
PRI. 1986. Developmental toxicity study in rabbits: Naphthalene. Waverly, PA: Pharmakon Research International, Inc.
PH 329-TX-001 85.
Schmeltz I, Tosk J, Hilfrich J, et al. 1978. Bioassays of naphthalene and alkylnaphthalenes for cocarcinogenic activity.
Relation to tobacco carcinogenesis. Carcinogenesis 3:47-60.
Shopp GM, White KL JR, Holsapple MP, et al. 1984. Naphthalene toxicity in CD-1 mice: General toxicology and
immunotoxicology. Fundam Appl Toxicol 4:406-419.
Tong, SS; Hirokata, Y; Trush, MA; et al. 1981 Clara cell damage and inhibition of pulmonary mixed-function oxidase
activity by naphthalene. Biochem Biophys Res Commun 100 3:944-950.
U.S. EPA. 1986. Broad scan analysis of the FY82 national human adipose tissue survey specimens: Volume I -
executive summary. Washington, DC: U.S. Environmental Protection Agency, Office of Toxic Substances.
U.S. EPA. 1987. Summary Review of Health Effects Associated with Naphthalene. U.S. Environmental Protection
Agency, Office of Health and Environmental Assessment, Washington, D.C. EPA/600/8-87/055F.
U.S. EPA. 1994. U.S. Environmental Protection Agency Methods for Derivation of Inhalation Reference
Concentrations and Application of Inhalation Dosimetry.
U.S. EPA. 1996. Exposure Factors Handbook. U.S. Environmental Protection Agency, Office of Research and
Development, Washington, D.C. EPA/600/8-89/043.
U.S. EPA. 1998. Toxicological Review of Naphthalene CAS No. 91-20-3 in Support of Summary Information on the
Integrated Risk Information System IRIS. U.S. Environmental Protection Agency. August, 1998. Available on the
Internet at http:/www.epa.gov/iris.
U.S. EPA. 2000. U.S. Environmental Protection Agency. Integrated Risk Information System IRIS. Naphthalene.
Available on-line at http://www.epa.gov/ngispgm3/iris
U.S. EPA. 2002. Health effects support document for naphthalene: External review draft. Alexandria, VA: U.S.
Environmental Protection Agency Office of Water. EPA822R02031. EPA/600/8-90/066F. Office of Research and
Development. Washington, DC: U.S.EPA.
Valaes T, Doxiadis SA, Fessas P. 1963. Acute hemolysis due to naphthalene inhalation. J Pediatr 63:904-915.
Verschueren K 2001. Handbook of Environmental Data on Organic Chemicals. Fourth edition. John Wiley & Sons, Inc,
New York. Vol. 1, 24.
Vulliamy T, Beutler E, Luzzatto L: Variants of glucose-6-phosphate dehydrogenase are due to missense mutations
spread throughout the coding region of the gene. Hum Mutat 2:159, 1993
Warren DL, Brown DL Jr, Buckpitt AR. 1982. Evidence for cytochrome P-450 mediated metabolism in the bronchiolar
damage of naphthalene. Chem Biol Interact 40:287-303.
Wells PG, Wilson B, Lubek BM. 1989. In vivo murine studies on the biochemical mechanism of naphthalene
cataractogenesis. Toxicol Appl Pharmacol 99:466-473.
West JAA, Pakehham G, Morin D, et al. 2001. Inhaled naphthalene causes dose dependent cell cytotoxicity in mice but
not in rats. Toxicol Appl Pharmacol 173 2:114-119.
Wolf O. 1978. Cancer of the larynx in naphthalene cleaner. Z. Gesamte. Hyg. 24 10:737-739.
The INDEX project Final report 297
Xu GT, Zigler JS, Lou MF. 1992a. Establishment of a naphthalene cataract model in vitro. Exp Eye Res 54:73-81.
Xu GT, Zigler JS, Lou MF. 1992b. The possible mechanism of naphthalene cataract in rat and its prevention by an
aldose reductase inhibitor AL01576. Exp Eye Res 54:63-72.
Zinkham WH, Childs B. 1957. Effect of vitamin K and naphthalene metabolites on glutathione metabolism of
erythrocytes from
normal newborns and patients with naphthalene hemolytic anemia. Am J Dis Child 94
6:420-423.
Zinkham WH, Childs B. 1958. A defect of glutathione metabolism of erythrocytes from patients with naphthalene-
induced hemolytic anemia. Pediatrics 22:461-471.
The INDEX project Final report 298
Acetaldehyde
Main sources :
Organization Shortcut Document link
World Health Organization –
International Program on Chemical
Safety
[WHO] http://www.inchem.org/documents/ehc/ehc/ehc167.htm
U.S.EPA - Integrated Risk
Information System
[IRIS] http://www.epa.gov/iris/subst/0290.htm
Office of Environmental Health
Hazard Assessment of the
Californian Environmental
Protection Agency
[OEHHA] http://www.oehha.ca.gov/air/chronic_rels/index.html
National Institute of Public Heatlh
and Environmental – The
Netherlands
[RIVM] http://www.rivm.nl/bibliotheek/rapporten/650270003.pdf
Environment Canada - Healt
Canada
[HC] http://www.ec.gc.ca/substances/ese/eng/psap/final/acetaldehyde.cfm
AIHA - American Industrial Hygiene Association. Odor thresholds for chemicals with established occupational health
standards. Akron OH: AIHA; 1989. p. 29.
Amoore JE & Hautala E 1983 Odor as an aid to chemical safety: odor thresholds compared with threshold limit values
and volatilities for 214 industrial chemicals in air and water dilution. J Appl Toxicol, 3 6: 272-290.
Anderson RA, Quigg JM, Oswald C, & Zaneveld LJD 1985 Demonstration of a functional blood-testis barrier to
acetaldehyde; evidence for lack of acetaldehyde effect on ethanol-induced depression of testosterone in vivo. Biochem
Pharmacol, 34 5: 685-695.
Appelman, L.M, R.A. Woutersen and V.J. Feron. 1982. Inhalation toxicity of acetaldehyde in rats. I. Acute and
subacute studies. Toxicology 23: 293–307.
Appelman, L.M, R.A. Woutersen, V.J. Feron, R.N. Hoofman and W.R.F. Notten. 1986. Effect of variable versus fixed
exposure levels on the toxicity of acetaldehyde in rats. J. Appl. Toxicol. 6: 331–336.
Aranyi, C, W. O’Shea, J. Graham and F. Miller. 1986. The effects of inhalation of organic chemical air contaminants on
murine lung host defences. Fundam. Appl. Toxicol. 6: 713–720.
Asmussen E, Hald J, & Larsen V 1948 The pharmacological action of acetaldehyde on the human organism. Acta
Pharmacol, 4: 311-320.
Badr FM & Hussain F 1977 Action of ethanol and its metabolite acetaldehyde in human lymphocytes. In vivo and in
vitro study. Genetics, 86: s2-s3.
Bakeas EB, Argyris DI, Siskos PA 2003. Carbonyl compounds in the urban environment of Athens, Greece.
Chemosphere 52, 805-813.
Baraona E, Di Padova C, Tabasco J, & Lieber CS 1987 Red blood cells:a new major modality for acetaldehyde
transport from liver to other tissues. Life Sci, 40: 253-258.
Blakley PM & Scott WR Jr 1984 Determination of the proximate teratogen of the mouse fetal alcohol syndrome. II.
Pharmacokinetics of the placental transfer of ethanol and acetaldehyde. Toxicol Appl Pharmacol, 72: 364-371.
Boehlke JU, Singh S, & Goedde HW 1983 Cytogenetic effects of acetaldehyde in lymphocytes of Germans and
Japanese: SCE, clastogenic activity, and cell cycle delay. Hum Genet, 63: 285-289
Bogdanffy MS, Randall HW, & Morgan KT 1986 Histochemical localization of aldehyde dehydrogenase in the
respiratory tract of the Fisher 344 rat. Toxicol Appl Pharmacol, 82: 560-567.
Bostrom CE et al 1994. Human exposure to urban air pollution. Environmental Health Perspectives Suppl 4:39-47.
Brien, J. and C. Loomis. 1983. Pharmacology of acetaldehyde. Can. J. Physiol. Pharmacol. 61: 1–22.
Casanova, M, K. Morgan, E. Gross, O. Moss and H. Heck. 1994. DNA–protein cross-links and cell replication at
specific sites in the nose of F344 rats exposed subchronically to formaldehyde. Fundam. Appl. Toxicol. 23: 525–536.
Cassee, F, J. Groten and V. Feron. 1996. Changes in the nasal epithelium of rats exposed by inhalation to mixtures of
formaldehyde, acetaldehyde, and acrolein. Fundam. Appl. Toxicol. 29: 208–218.
Cederbaum, A. and E. Rubin. 1976. Protective effect of cysteine on the inhibition of mitochondrial functions by
acetaldehyde. Biochem. Pharmacol. 25: 963–973.
The INDEX project Final report 299
Cina SJ, Russell RA, Conradi SE. Sudden death due to metronidazole/ethanol interaction. Am J Forensic Med Pathol
1996; 17 4:343-346.
Clarisse B, Laurent AM, Seta N, Le Moullec Y, El Hasnaoui A, Momas I. 2003. Indoor aldehydes: measurement of
contamination levels and identification of their determinants in Paris dwellings. Environ Res. 92 3:245-53.
Dalhamn, T, M. Edfors and R. Rylander. 1968. Retention of cigarette smoke components in human lungs. Arch.
Environ. Health 17: 746–748.
David, R. and H. Heck. 1983. Disposition of inhaled [14C]-acetaldehyde. Toxicologist 3: 331 Abstract.
DECOS Dutch Expert Committee on Occupational Standards. 1993. Health-based recommended occupational exposure
limit for acetaldehyde. Directorate-General of Labour, The Netherlands.
De Bortoli M, Knöppel H, Pecchio E, Peil A, Rogora L, Schauenburg H, Schlitt H and Vissers H 1986. Concentrations
of selected organic pollutants in indoor and outdoor air in Northern Italy. Environ Internat 12: 343-350.
Devos M, Patte F, Rouault J, Laffort P, Van Genert LJ, 1990. Standardized Human Olfactory Thresholds. IRL Press at
Oxforf University Press.
Egle JL 1970 Retention of inhaled acetaldehyde in man. J Pharmacol Exp Ther, 174 1: 14-19.
EPA 1990. Indoor Air Quality and Work Environment Study. EPA Headquarters Buildings Vol. 2: Results of Indoor
Air Environmental Monitoring Study. Research Triangle Park, NC: Atmos Res Expos Asses Lab.
EPA/California 2003, Chronic Reference Exposure Levels Adopted Office of Environmental Health Hazard
Assessment OEHHA as of August 2003. Available in the Internet
http://www.oehha.ca.gov/air/chronic_rels/AllChrels.html
.
Feng Y, Wen S, Wang, Sheng G, He Q, Tang J, Fu J 2004. Indoor and outdoor carbonyl compounds in the hotel
ballrooms in Guangzhou, China. Atmospheric Environment 38, 103–112.
Feron, V. 1979. Effects of exposure to acetaldehyde in Syrian golden hamsters simultaneously treated with benzo
apyrene or dimethylnitrosamine. Prog. Exp. Tumour Res. 24: 162–176.
Feron, V, A. Kruysse and R. Woutersen. 1982. Respiratory tract tumours in hamsters exposed to acetaldehyde vapour
alone or simultaneously to benzo apyrene or diethylnitrosamine. Eur. J. Cancer Clin. Oncol. 18: 13–31.
Feron, V, C. Kuper, B. Spit, P. Reuzel and R. Woutersen. 1985. Glass fibres and vapour phase components of cigarette
smoke as cofactors in experimental respiratory tract carcinogenesis. Carcinogenesis 8: 93–118.
Feron, V, R. Woutersen and L. Appelman. 1984. Epithelial damage and tumours of the nose after exposure to four
different aldehydes by inhalation. In: Grosdanoff et al. eds, Problems of inhalatory toxicity studies. MMV Medizin
Verlag Munchen. pp. 587–604.
Flemming RC. Upper respiratory tract toxicity of mixtures of aldehydes In vivo and in vitro studies. 1995. Wageningen,
Ponsen en Looyen B.V. Ref Type: Report
Goedde HW & Agarwal DP 1986 Aldehyde oxidation: ethnic variations in metabolism and response. In: Kalow W,
Goedde W, & Agarwal DP ed.Ethnic differences in reactions to drugs and xenobiotics. New York, Alan R. Liss Inc, pp
113-138.
Goedde HW, Agarwal DP, & Harada S 1979 Alcohol metabolizing enzymes:studies of isozymes in human biopsies and
cultured fibroblasts. Clin Genet, 16: 29-33.
Grosjean D 1991 Ambient levels of formaldehyde, acetaldehyde and fumaric acid in Southern California. Results of a
one-year base-line study. Environ Sci Technol, 25:710-715.
Guicherit R & Schulting FL 1985. The occurrence of organic chemicals in the atmosphere of the Netherlands. Sci Total
Environ, 43:193-219.
Gustaffson P, Barregard L, Strandberg B, Sallsten G. Domestic wood burning and exposure to benzene 1,3-butadiene
and aldehydes. In: Proceedings of the 13th annual conference of ISEA, Stresa, Italy, 2003.
Hagemeyer HJ 1978 Acetaldehyde. In: Grayson M ed. Kirk-Othmer encyclopedia of chemical technology, 3rd ed. New
York, John Wiley and Sons, vol 1, pp 97-112.
He SM & Lambert B 1990 Acetaldehyde-induced mutation at the hprt locus in human lymphocytes in vitro. Environ
Mol Mutagen, 16: 57-63.
Health Canada 2000. Environment Canada. Priority substances list assessment report: acetaldehyde Priority substances
list assessment report Canadian Environmental Protection Act, 1999. Co-published by Health Canada. ISBN 0-662-
28654-5 Cat. no. En40-215/50E Environment Canada’s Web site: www.ec.gc.ca
Heelon MW, White M. Disulfiram-cotrimoxazole reaction. Pharmacotherapy 1998; 18 4:869-870.
Hellström-Lindahl E & Weiner H 1985 Effects of disulfiram on the oxidation of benzaldehyde and acetaldehyde in rats
liver. Biochem Pharmacol, 34 9: 1529-1535.
The INDEX project Final report 300
Hemminiki, K. 1982. Urinary sulfur containing metabolites after administration of ethanol, acetaldehyde and
formaldehyde to rats. Toxicol. Lett. 11: 1–6.
Hobara N, Watanabe A, Kobayashi M, Nakatsukasa H, Nagshima H, Fukuda T, & Araki Y 1985 Tissue distribution of
acetaldehyde inhalation and intragastric ethanol administration. Bull Environ Contam Toxicol, 35: 393-396.
Hodgson AT, Rudd AF, Beal D, Chandra S 2000. Volatile organic compound concentrations and emission rates in new
manufactured and site-built houses. Indoor Air 10:178-192.
Howe, R. 1995. THRESH: A computer program to compute a reference dose from quantal animal toxicity data using
the benchmark dose method. ICF Kaiser Engineers, Inc, Ruston, Lousiana.
Hsu LC, Bendel RE, & Yoshida A 1978 Genomic structure of the human mitochondrial aldehyde dehydrogenase gene.
Genomics, 2 1: 57-65.
IARC 1985. International Agency for Research on Cancer. IARC Monographs on the Evaluation of the carcinogenic
Risk of Chemicals to Humans. Allyl Compounds, Aldehydes, Epoxides, and Peroxides. Vol. 36. 369 pp. Lyon, France:
IARC, 1985.
IARC 1987. International Agency for Research on Cancer. IARC Monographs on the Evaluation of Carcinogenic Risks
to Humans. Overall Evaluations of Carcinogenicity. Supplement 7. 440 pp. Lyon, France: IARC, 1987.
IARC 1999. International Agency for Research on Cancer. IARC Monographs on the Evaluation of Carcinogenic Risks
to Humans. Re-evaluation of Some Organic Chemicals, Hydrazine, and Hydrogen Peroxide. Vol. 71. 1589 pp. Lyon,
France: IARC, 1999.
IPCS/CEC 1990 International Chemical Safety Card No. 0009:Acetaldehyde. Luxembourg, Commission of the
European Communities First series.
Jones, A. 1995. Measuring and reporting the concentration of acetaldehyde in human breath. Alcohol Alcohol. 30: 271–
285.
Jurvelin J, Vartiainen M, Jantunen M, Pasanen P. 2001. Personal Exposure Levels and Microenvironmental
Concentrations of Formaldehyde and Acetaldehyde in the Helsinki metropolitan Area, Finland. J.Air &Waste Manage.
Assoc. 51:17-24.
Karl PI, Gordon BHJ, Lieber CS, & Fisher SE 1988 Acetaldehyde production and transfer by perfused human placental
cotyledon. Science, 242: 273-275.
Kirchner S 2004. Preliminary results of the French National Survey on Indoor Air Quality. Personal communication.
Koivula T 1975 Subcellular distribution and characterization of human liver aldehyde dehydrogenase fractions. Life
Sci, 16: 1563-1570.
Kruysse, A, V.J. Feron and H.P. Til. 1975. Repeated exposure to acetaldehyde vapour. Studies in Syrian golden
hamsters. Arch. Environ. Health 30: 449–452.
Lam CW, Casanova M, & Heck HD 1986 Decreased extractability of DNA from proteins in the rat nasal mucosa after
acetaldehyde exposure. Fundam Appl Toxicol, 6 3: 541-550
Lee SC, Wang B. 2004. Characteristics of emissions of air pollutants from burning of incense in a large environmental
chamber. Atmospheric Environment 38:941-951.
Maarse H, Visscher CA 1992. Volatile compounds in food. Qualitative and quantitative data. Supplement 3 and
cumulative index. Zeist, Central Institute for Nutrition and Food Research, TNO, Biotechnology and Chemistry
Institute, pp XXXIII-XLI.
Marjanen LA 1973 Comparison of aldehyde dehydrogenases from cytosol and mitochondria of rat liver. Biochim
Biophys Acta, 327: 238-246.
Michoudet C & Baverel G 1987a Characteristics of acetaldehyde metabolism in isolated dog, rat and guinea pig kidney
tubules. Biochem Pharmacol, 36 22: 3987-3991.
Michoudet C & Baverel G 1987b Metabolism of acetaldehyde in human and baboon renal cortex:ethanol synthesis by
isolated baboon kidney-cortex tubules. FEBS Lett, 216 1: 113-117.
Morris, J. 1997. Uptake of acetaldehyde vapour and aldehyde dehydrogenase levels in the upper respiratory tracts of the
mouse, rat, hamster, and guinea-pig. Fundam. Appl. Toxicol. 35: 91–100.
Morris, J, D. Robinson, T. Vollmuth, R. Brown and B. Domeyer. 1996. A parallelogram approach for safety evaluation
of ingested acetaldehyde. Regul. Toxicol. Pharmacol. 24: 251–263.
Nazaroff WW and Singer BC 2002. Inhalation of hazardous air pollutants from environmental tobacco smoke in US
residences. In: Proceedings of the 9
th
International Conference on Indoor Air Quality and Climate, vol 2, 477-482.
New Jersey Department of Health and Senior Services – Hazardous Substances Fact Sheets available on web at:
http://www.state.nj.us/health/eoh/rtkweb
.
The INDEX project Final report 301
Nicholls, R, J. De Jersey, S. Worrall and P. Wilce. 1992. Modification of proteins and other biological molecules by
acetaldehyde: adduct structure and functional significance. Int. J. Biochem. 24: 1899–1906.
Obe G, Brodmann R, Fleischer R, Engeln H, Gobel D, & Herha J 1985[Mutagenic and carcinogenic activity of adduct
products.] In: Keup W ed. [Biology of addiction.] Berlin, Heidelberg, New York, Springer-Verlag, pp 31-43 in German.
Obe G, Natarajan AT, Meyers M, & Den Hertog A 1979 Induction of chromosomal aberrations in peripheral
lymphocytes of human blood in vitro, and of SCE's in bone-marrow cells of mice in vivo by ethanol and its metabolite
acetaldehyde. Mutat Res, 68: 291-294.
Obe G, Ristow H, & Herha J 1978 Mutagenic activity of alcohol in man. In: Mutations: their origin, nature and
potential relevance to genetic risk in man. Proceedings Yearly Conference 1977 of German Research Society. Boppard,
Harald Boldt, pp 151-161.
OEHHA 1999a. Office of Environmental Health Hazard Assessment of the Californian Environmental Protection
Agency. Air Toxics Hot Spots Program Risk Assessment Guidelines. Part I. The Determination of Acute Reference
Exposure Levels for Airborne Toxicants.
OEHHA 1999b. Office of Environmental Health Hazard Assessment of the Californian Environmental Protection
Agency. REPORT ON ETHANOL IN GASOLINE - 02/15/00 Toxicology and Epidemiology Section, March 1999.
OEHHA 1999c. Office of Environmental Health Hazard Assessment of the Californian Environmental Protection
Agency. Chronic Reference Exposure Levels Adopted by OEHHA available on the Internet at
http://www.arb.ca.gov/toxics/tac/factshts/acetalde.pdf
Ortiz, A, P. Griffiths and J. Littleton. 1974. A comparison of the effects of chronic administration of ethanol and
acetaldehyde to mice: evidence for a role of acetaldehyde in ethanol dependence. J. Pharm. Pharmacol. 26: 249–260.
Parrilla R, Ohkawa K, Lindros KO, Zimmernman UP, Kobayashi K, & Williamson JR 1974 Functional
compartmentation of acetaldehyde oxidation in rat liver. J Biol Chem, 249: 4926-4933.
Priscott PK & Ford JR 1985 An in vitro model of acetaldehyde metabolism by rodent conceptuses. in vitro Cell Dev
Biol,21 2: 88-92.
Roe, F. and D. Wood. 1992. Acetaldehyde and formaldehyde: is there a cancer risk for man? Indoor Environ. 1: 8–15.
Roumec, C, Y. Lamboeuf and G. Blanquat. 1988. Synaptosomal phospholipids in rats chronically treated with
acetaldehyde. Adv. Biosci. 71: 201–205.
Saldiva, P, M. Caldiera, E. Massad, D. Calheiros, L. Cardoso, G. Bohm and C. Saldiva. 1985. Effects of formaldehyde
and acetaldehyde inhalation on rat pulmonary mechanics. J. Appl. Toxicol. 5: 288–292.
Shiohara, E, M. Tsukada, S. Chiba, H. Yamazaki, K. Nishiguchi, R. Miyamoto and S. Nakanishi. 1985. Effect of
chronic administration of acetaldehyde by inhalation on Na+ + K+-activated adenosine triphosphate activity of rat brain
membranes. Toxicology 34: 277–284.
Silverman L, Schulte HF, & First MW 1946 Further studies on sensory response to certain industrial solvent vapors. J
Ind Hyg Toxicol, 28: 262-266.
Sim VM & Pattle RE 1957 Effect of possible smoke irritation on human subjects. J Am Med Assoc, 165: 1908-1913.
SIS 2003 Specialised Information Services. US National Library of Medicine. ChemlPlus Record. Available in the
Internet at http://chem2.sis.nlm.nih.gov/chemidplus/jsp/chemidlite/ChemFull.jsp
last updated October 1, 2003 .
Sprince, H, C. Parker, P. Smith, G. Smith and L. Gonzales. 1974. Protection against acetaldehyde toxicity in the rat by
L-cysteine, thiamine, and L-2-methylthiazolidine-4-carboxylic acid. Agents Actions 4: 125–130.
Suokas A, Kupari M, Pettersson J, Lindros K. The nitrefazole-ethanol interaction in man: cardiovascular responses and
the accumulation of acetaldehyde and catecholamines. Alcohol Clin Exp Res 1985; 9 3:221-227.
Teschke R, Hasumura Y, & Lieber CS 1977 Hepatic pathways of ethanol and acetaldehyde metabolism and their role in
the pathogenesis of alcohol-induced liver injury. Nutr Metab, 21 Suppl 1: 144-147.
U.S. EPA 1987. Health assessment document for acetaldehyde. Environmental Criteria Assessment Office, Research
Triangle Park, North Carolina EPA/600/8-86/105A.
U.S. EPA 1998. Integrated Risk Information System IRIS. Acetaldehyde. Online. National Center for Environmental
Assessment, Cincinnati, OH. Available at: http://www.epa.gov/iris/subst/0290.htm
Veghelyi PV & Osztovics M 1978 The alcohol syndromes: the intrarecombigenic effect of acetaldehyde. Experientia
Basel, 34: 195-196.
Verschueren K 2001. Handbook of Environmental Data on Organic Chemicals. Fourth edition. John Wiley & Sons, Inc,
New York. Vol. 1, 24.
von Wartburg, J.-P. 1987. Acute aldehyde syndrome and chronic aldehydism. Mutat. Res. 186: 249–259.
The INDEX project Final report 302
Watanabe A, Hobara N, & Nagashima H 1986 Blood and liver acetaldehyde concentration in rats following
acetaldehyde inhalation and intravenous and intragastric ethanol administration. Bull Environ Contam Toxicol, 37: 513-
516.
Watanabe I 1987 [Distribution of lower oxygenated organic compounds in urban air.] Preprint of 54th Annual Meeting
of the Chemical Society of Japan, vol 1, p 129 in Japanese.
WHO 1995. Environmental health criteria for acetaldehyde, 167. WHO, Geneva, 1995. Available in the Internet
http://www.inchem.org/documents/ehc/ehc/ehc167.htm.
WHO. World Health Organization Regional Office for EuropeGuidelines for Air Quality 1999. WHO Geneva,
Switzerland. http://www.euro.who.int/document/aiq/5_8formaldehyde.pdf.
Woutersen, R. and V. Feron. 1987. Inhalation toxicity of acetaldehyde in rats. IV. Progression and regression of nasal
lesions after discontinuation of exposure. Toxicology 47: 295–305.
Woutersen, R, L. Appelman, A. Van Garderen-Hoetmer and V. Feron. 1986. Inhalation toxicity of acetaldehyde in rats.
III. Carcinogenicity study. Toxicology 41: 213–231.
Woutersen, R, L. Appelman, V. Feron and C. Van der Heijden. 1984. Inhalation toxicity of acetaldehyde in rats. II.
Carcinogenicity study: interim results after 15 months. Toxicology 31: 123–133.
Yin, S, C. Liao, C. Chen, F. Fan and S. Lee. 1992. Genetic polymorphism and activities of human lung alcohol and
aldehyde dehydrogenases: implications for ethanol metabolism and cytotoxicity. Biochem. Genet. 30: 203–215.
Yoshida A, Huang IY, & Ikawa M 1984 Molecular abnormality of an inactive aldehyde dehydrogenase variant
commonly found in Orientals. Proc Natl Acad Sci USA, 81: 258-261.
The INDEX project Final report 303
Toluene
Main sources :
Organization Shortcut Document link
World Health Organization - Air Quality
Guidelines for Europe – 2000
[WHO] Not available on web
U.S.EPA - Integrated Risk Information
System
[IRIS] http://www.epa.gov/iris/subst/0118.htm
Office of Environmental Health Hazard
Assessment of the Californian Environmental
Protection Agency
[OEHHA1] http://www.oehha.ca.gov/air/chronic_rels/index.html
[OEHHA2] http://www.oehha.ca.gov/air/acute_rels/allAcRELs.html
European Chemical Bureau - Institute for
Health and Consumer Protection IHCP
[ECB] http://ecb.jrc.it
Agency for Toxic Substances and Disease
Registry
[ATSDR] http://www.atsdr.cdc.gov/phshome.html
Abbate C, Giorgianni C, Munao F, and Brecciaroli R. 1993. Neurotoxicity induced by exposure to toluene. An
electrophysiologic study. Int. Arch. Occup. Environ. Health 64:389-92.
Abou-El-Makarem MM, Millburn P, Smith RL, Williams RT 1967. Biliary excretion of foreign compounds. Benzene
and its derivatives in the rat. Biochem. J. 105, 1269-1274.
AIHA - American Industrial Hygiene Association. Odor thresholds for chemicals with established occupational health
standards. Akron OH: AIHA; 1989. p. 29.
Andersen MD, Lundqvist GR, Molhave L, Pedersen OF, Proctor DF, Vaeth M, et al. Human response to controlled
levels of toluene in six-hour exposures. Scand J Work Environ Health 1983;9:405-418.
Askergren A 1982. Organic solvents and kidney function. Adv. Mod. Environ. Toxicol. 2, 157-172.
Åstrand I 1975. Uptake of solvents in the blood and tissues of man. A review. Scand. J. Work Environ. Health, 1 199-
218.
Åstrand I, Ehrner-Samuel H, Kilbom Å, Övrum P 1972. Toluene exposure I. Concentration in alveolar air and blood at
rest and during exercise. Work Environ. Health 9, 119-30.
ATSDR - Agency for Toxic Substances and Disease Registry 1999. Toxicological Profile for Toluene Update.
PB/95/100228/AS. U.S. Department of Health and Human Services, Public Health Service.
ATSDR - Agency for Toxic Substances and Disease Registry. Cases in environmental medicine: toluene toxicity.
Atlanta GA: US Dept of Health and Human Services; 1993.
ATSDR - Agency for Toxic Substances and Disease Registry. Toluene. Atlanta GA: US Public Health Service; 1989.
Baelum J, Andersen LB, Lundqvist GR, Molhave L, Pedersen OF, Vaeth M, et al. Response of solvent-exposed printers
and unexposed controls to six-hour toluene exposure. Scand J Work Environ Health 1985;11:271-280.
Bælum J, Lundqvist GR, Mølhave L, Andersen NT 1990. Human response to varying concentrations of toluene. Int.
Arch. Occup. Environ. Health 62, 65-71.
Ballesta PP, Connolly R, Boix A, Cancelinha J, Cao N, Baldan A, Payrissat M 1998. Benzene, Toluene and Xylene
Monitoring Campaign in Bologna. European Commission, Joint Research Center, Environment Institute, EUR 18093
EN, Ispra, Italy.
Ballesta PP, Payrissat M, Saeger E De, Cancelinha J, Galata R 1997. BTX Monitoring Campaign in the City of Catania.
European Commission, Joint Research Center, Environment Institute, EUR 17272 EN, Ispra, Italy.
Bauchinger, M. et al. Chromosome changes in lymphocytes after occupational exposure to toluene. Mutation research,
102: 439–445 1982.
Benignus VA, Muller KE, Barton CN, Bittikofer JA 1984. Toluene blood level following subcutaneous injection of
toluene in the rat. Environ. Res. 33, 441-453.
Bennett RH, Forman HR 1980. Hypokalemic periodic paralysis in chronic toluene exposure. Arch. Neurol. 37, 673.
Bergman K 1979. Whole-body autoradiography and allied tracer techniques in distribution and elimination studies of
some organic solvents. Scand. J. Work. Environ. Health. 5, suppl 1, 1-263.
Bergman K 1983. Application and results of whole-body autoradiography in distribution studies of organic solvents.
CRC Crit. Rev. Toxicol. 12, 59-118.
The INDEX project Final report 304
Boey KW, Foo SC, and Jeyaratnam J. 1997. Effects of occupational exposure to toluene: a neuropsychological study on
workers in Singapore. Ann. Acad. Med. Singapore 26:184-7.
Brown SK 2002. Volatile organic pollutants in new and established buildings in Melbourne, Australia. Indoor Air
12:55-63.
Brown VM, Coward SKD, Crump DR, Llewellyn JW, Mann HS, Raw GJ 2002. Indoor air quality in English homes –
VOCs. In: Proceedings of the 9
th
International Conference on Indoor Air Quality and Climate, vol 4, 477-482.
Carlsson A 1982. Exposure to toluene. Uptake, distribution and elimination in man. Scand. J. Work. Environ. Health 8,
43-55.
Carlsson A, Lindqvist T 1977. Exposure of animals and man to toluene. Scand. J. Work. Environ. Health 3, 135-143.
Carlsson A, Ljungquist E 1982. Exposure to toluene. Concentration in subcutaneous adipose tissue. Scand. J. Work
Environ. Health 8, 56-62.
Carrer P, Maroni M, Alcini D, Cavallo D, Fustinoni S, Lovato L, Visigalli F 2000. Assessment through Environmental
and Biological Measurements of Total Daily Exposure to Volatile Organic Compounds of Office Workers in Milan,
Italy. Indoor Air 10 4, 258-268.
Chang MJW, Hsu KH, Chen YC, Hsieh LL, Luo JJ 1996. Biological monitoring of urinary hippuric acid in a Taiwanese
semiconductor company. Abstracts of the International Symposium on Biological Monitoring in Occupational and
Environmental Health. Sept 11-13, Espoo, Helsinki. Finnish Institute of Occupational Health.
Chapman, D.E. et al. Metabolism and covalent binding of [14C]toluene by human and rat liver microsomal fractions
and liver slices. Drug metabolism and disposition, 18: 929–936 1990.
Cherry, N, H. Hutchins, T. Pace and H.A. Waldron. 1985. Neurobehavioral effects of repeated occupational exposure to
toluene and paint solvents. Br. J. Ind. Med. 42 5: 291-300.
Dees, C. & Travis, C. Hyperphosphorylation of p53 induced by benzene, toluene and chloroform. Cancer letters, 84:
117–123 1994.
Donald, J.M, Hooper, K. & Hopenhayn-Rich, C. Reproductive and developmental toxicity of toluene: A Review.
Environmental health perspectives, 94: 237–244 1991.
Dudek, B. et al. Neurobehavioural effects of experimental exposure to toluene, xylene and their mixture. Polish journal
of occupational medicine, 3: 109–116 1990.
Echeverria D, Fine L, Langolf G, Schork A, Sampaio C. Acute neurobehavioral effects of toluene. Br J Ind Med
1989;46:483-495.
Elke K, Jermann E, Begerow J, Dunemann L 1998. Determination of benzene, toluene, ethylbenzene and xylenes in
indoor air at environmental levels using diffusive samplers in combination with headspace solid-phase microextraction
and high-resolution gas chromatography-flame ionization detection. J of Chromatography A 826:191-200.
EPA/California 2003, Chronic Reference Exposure Levels Adopted Office of Environmental Health Hazard
Assessment OEHHA as of August 2003. Available in the Internet
http://www.oehha.ca.gov/air/chronic_rels/AllChrels.html.
EU 2003. European Union Risk Assessment Report. Toluene. European Chemicals Bureau. Joint Research Centre.
Institute for Health and Consumer Protection. Existing Substances, 2
nd
Priority List. EUR 20539 EN, Volume: 30. p.
320.
EXPOLIS 2002. Combined International Database CIDB, KTL- National Public Health Institute, Kuopio Finland.
Fischman CM, Oster JR 1979. Toxic effects of toluene: A new cause of high anion gap metabolic acidosis. J. Am. Med.
Ass. 241, 1713-1715.
Foo SC, Jeyaratnam, J, and Koh D. 1990. Chronic neurobehavioral effects of toluene. Br. J. Ind. Med. 47 7:480-484.
Fornazzari L, Wilkinson DA, Kapur BM, and Carlen PL. 1983. Cerebellar cortical and functional impairment in toluene
abusers. Acta Neurol. Scand. 67:319-329.
Franchini I, Cavatorta A, Falzoi M, Lucertini S, Mutti A 1983. Early indicators of renal damage in workers exposed to
organic solvents. Int. Arch. Occup. Environ. Health 52, 1-9.
Gamberale F, Hultengren M. Toluene exposure. II Psychophysiological functions. Work Environ Health 1972;9:131-
139.
Ghantout H, Danielsson BRG 1986. Placental transfer and distribution of toluene, xylene and benzene, and their
metabolites during gestation in mice. Biological Research in Pregnancy 7, 98-106.
Hass U, Lund SP, Hougaard KS, Simonsen L 1999. Developmental neurotoxicity after toluene inhalation exposure in
rats. Neurotoxicol. Teratol. 21 4, 349-57.
Helmig, D, Arey J 1992. Organic chemicals in the air at Whitaker’s Forest/Sierra Nevada
The INDEX project Final report 305
Hillefors-Berglund M, Liu Y, and von Euler G. 1995. Persistent, specific and dose-dependent effects of toluene
exposure on dopamine D2 agonist binding in the rat caudate-putamen. Toxicology 100:185-94.
Hoshika Y, Imamura T, Muto G, Van Gemert LJ, Don JA, Walpot JI. 1993. International comparison of odor threshold
values of several odorants in Japan and in The Netherlands. Environ Res. 1993 Apr;61 1:78-83.
HSDB 2003. Hazardous Substances Data Bank. Specialised Information Services. National Library of Medicine.
Available online at http://toxnet.nlm.nih.gov/.
IPCS 1985. Toluene. World Health Organixation WHO, International Programme on Chemical Safety IPCS,
Environmental Health Criteria 52, Geneva.
Iregren, A. 1982. Effects on psychological test performance of workers exposed to a single solvent toluene - a
comparison with effects of exposure to a mixture of organic solvents. Neurobehav. Toxicol. Teratol. 4 6: 695- 701.
Iregren, A. et al. Experimental exposure to toluene in combination with ethanol intake. Scandinavian journal of work &
environmental health, 12: 128–136 1986.
Jensen AA, Slorach SA 1991. Chemical Contaminants in Human Milk. CRC Press, Bocan Raton.
Kawai T, Yasugi T, Mizunuma K, et al. 1992b. Monitoring of workers exposed to a mixture of toluene, styrene and
methanol vapours by means of diffusive air sampling, blood analysis and urinalysis. Int Arch Occup Environ Health
63:429-435.
Kelly TW 1975. Prolonged cerebellar dysfunction associated with paintsniffing. Pediatrics 56, 605-606.
Kemp K, Palmgren F, Manscher OH 1998. The Danish Air Quality Monitoring Programme. Annual Report for 1997.
National Environmental Research Institute, NERI Technical Report No.245, Roskilde, Denmark.
Kishi R, Harabuchi I, Ikeda T, Yokota H, Miyake H 1988. Neurobehavioural effects and pharmakokinetics of toluene in
rats and their relevance to man. Br. J. Ind. Med. 45, 396-408.
Kotzias D 2004. Preliminary results of the Airmex-project. Personal communication.
Kroeger RM, Moore RJ, Lehman TH, Giesy JD, Skeeters CE 1980. Recurrent urinary calculi associated with toluene
sniffing. J. Urol. 123, 89-91.
Lauwerys R 1983. Human Biological Monitoring of Industrial Chemicals Series. Toluene. Industrial Health and Safety.
Alessio L, Berlin A, Roi R, Boni M eds. Ispra, Italy, 175 pp.
Lee SC, Wang B. 2004. Characteristics of emissions of air pollutants from burning of incense in a large environmental
chamber. Atmospheric Environment 38:941-951.
Lindbohm, M.-L. et al. Spontaneous abortions among women exposed to organic solvents. American journal of
industrial medicine, 17: 449–463 1990.
Lindqvist T 1977. Fördelingskoefficienterna blod/luft och vatten/luft för några vanliga lösningsmedel. Arbete och Hälsa
8, 1-5.
Löf A, Wigaeus Hjelm E, Colmsjö A, Lundmark BO, Norström Å, Sato A 1993. Toxicokinetics of toluene and urinary
excretion of hippuric acid after human exposure to 2H8-toluene. Br. Journ. Ind. Med. 50, 55-59.
Massengale ON, Glaser HH, LeLievre RE, Dodds MJB, Klock ME 1963. Physical and psychologic factors in glue
sniffing. New Engl. J. Med. 269, 1340-1344.
Moss AH, Gabow PA, Kaehny WD, Goodman SI, Haut LL, Haussler MR 1980. Fanconi's syndrome and distal renal
tubular acidosis after glue sniffing. Ann. Intern. Med. 92, 69-70.
Mountains, California. Science of the total environment, 112: 233–250.
Muttray A, Wolters V, Jung D, and Konietzko J. 1999. Effects of high doses of toluene on color vision. Neurotoxicol.
Teratol. 21:41-5.
Ng, T.P, Foo, S.C. & Yoong, T.. Menstrual function in workers exposed to toluene. British journal of industrial
medicine, 49: 799–803 1992.
Ng, T.P, Foo, S.W. & Yoong, T. Risk of spontaneous abortion in workers exposed to toluene. British journal of
industrial medicine, 49: 804–808 1992.
Nielsen HK, Krusell L, Bælum J, Lundqvist G, Omland Ø, Væth M, Husted SE, Mogensen CE, Geday E 1985. Renal
effects of acute exposure to toluene. A controlled clinical trial. Acta Med. Scand. 218, 317-321.
Nise, G. et al. Cytogenetic effects in rotogravure printers exposed to toluene and benzene. Mutation research, 261: 217–
223 1991.
Nomiyama K, Nomiyama H 1974. Respiratory retention, uptake and excretion of organic solvents in man. Benzene,
toluene, n-hexane, trichloroethylene, acetone, ethyl acetate and ethyl alcohol. Int. Arch. Arbeitsmed. 32, 75-83.
NTP. 1990. National Toxicology Program. Toxicology and carcinogenesis studies of toluene CAS No. 108-88-3 in
F344/N rats and B6C3F1 mice inhalation studies. NTP-TR-371.
The INDEX project Final report 306
Nylen, P. Hagman, M. & Johnson, A.-C. Function of the auditory system, the visual system, and peripheral nerve and
long-term combined exposure to toluene and ethanol in rats. Pharmacology and toxicology, 76: 107–111 1995.
O'Brian ET, Yeoman WB, Hobby JAE 1971. Hepatorenal damage from toluene in a "glue sniffer". Br. Med. J. 2, 29-30.
Orbaek P and Nise G. 1989. Neurasthenic complaints and psychometric function of toluene-exposed rotogravure
printers. Am. J. Ind. Med. 16:67-77.
Patel R, Benjamin J 1986. Renal disease associated with toluene inhalation. Clin. Toxicol. 24, 213-223.
Pelclova, D, Rossner, P. & Pickova, J. Chromosome aberrations in rotogravure printing plant workers. Mutation
research, 245: 299–303 1990.
Periago, F, Luna, A. & Morente, A. Determinacion de disolventes en el aire exhalado de una poblacion laboralmente
expuesta. Toxicologia, 92: 103–106 1992.
Plappert, U, Baryhel, E. & Seidel, H.J. Reduction of benzene toxicity by toluene. Environmental and molecular
mutation, 24: 283–292 1994.
Rees DC, Wood RW, McCormick JP, Cox C 1985. Toxicokinetics of toluene in the rat. Scand. J. Work Environ. Health
11, 301-306.
Reisin E, Teicher A, Jaffe R, Eliahou HE 1975. Myoglobinuria and renal failure in toluene poisoning. Br. J. Ind. Med.
32, 163-168.
Reprotext® System. Dabney BJ, editor. Denver CO: Micromedex, Inc.; 1999. Edition expires 1/31/1999.
Richer, C.L. et al. Cytogenetic effects of low-level exposure to toluene, xylene, and their mixture on human blood
lymphocytes. International archives of occupational and environmental health, 64: 581–585 1993.
Russ G, Clarkson AR, Woodroffe AJ, Seymour AE, Cheng IKP 1981. Renal failure from glue sniffing. Med. J. Aust. 2,
121-123.
Saarela K, Tirkkonen T, Laine-Ylijoki J, Jurvelin J, Nieuwenhuijsen MJ, Jantunen M 2003. Exposure of population and
microenvironmental distributions of volatile organic compound concentrations in the EXPOLIS study. Atmospheric
Environment 37, 5563-5575.
Saeger E De, Gerbola M, Ballesta PP, Amantini L, Payrissat M 1995. Air Quality Measurements in Brussels 1993-
1994. NO
2
and BTX Monitoring Campaigns by Diffusive Samplers. European Commission, Joint Research Center,
Environment Institute, EUR 16310 EN, Ispra, Italy.
Sato A, Fujiwara Y, Hirosawa K 1972. Solubility of benzene, toluene and m-xylene in blood. In Japanese with English
summary Jap. J. Ind. health 14, 3-8.
Sato A, Nakajima T 1977. Vatten/luft, blod/luft, olja/luft, olja/vatten och olja/blod fördelingskoefficientar av några
aromatiska kolväten. Original in Japanese, translated to Swedish. Jap. J. Ind. Health 19, 132-133.
Sato A, Nakajima T, Fujiwara Y, Hirosawa K 1974. Pharmacokinetics of benzene and toluene. Int. Arch. Arbeitsmed.
33, 169-182.
Sherwood RJ 1976. Ostwald solubility coefficients of some industrially important substances. Br. J. Ind. Med. 33, 106-
107.
Spencer PS, Schaumberg HH. Organic solvent neurotoxicity: Facts, and research needs. Scand J Work Environ Health
II 1985;11 Suppl 1:53-60 [cited in NRC, 1987].
Streicher HZ, Gabow PA, Moss AH, Kono D, Kaehny WD 1981. Syndromes of toluene sniffing in adults. Ann. Intern.
Med. 94, 758-762.
Svensson BG, Nise G, Erfurth EM, Olsson H 1992b. Neuroendocrine effects in printing workers exposed to toluene. Br.
J. Ind. Med. 49, 402-08.
Svensson, B.G. et al. Deaths and tumours among rotogravure printers exposed to toluene. British journal of industrial
medicine, 47: 372–379 1990.
Svensson, B.G. et al. Hormone status in occupational toluene exposure. American journal of industrial medicine, 22:
99–107 1992.
Svensson, B.G. et al. Neuroendocrine effects in printing workers exposed to toluene. British journal of industrial
medicine, 49: 402–408 1992.
Taher SM, Anderson RJ, McCartney R, Popovtzer MM, Schrier RW 1974. Renal tubular acidosis associated with
toluene "sniffing". New Engl. J. Med. 290, 765-768.
Takahashi, S. et al. Determination of S-benzyl-N-acetyl-L-cysteine by gas chromatography/mass spectrometry as a new
marker of toluene exposure. Journal of analytical toxicology, 18: 78–80 1994.
Tardif R, Lapare S, Plaa GL, et al. 1991. Effect of simultaneous exposure to toluene and xylene on their respective
biological exposure indices in humans. Int Arch Occup Environ Health 63:279-284.
The INDEX project Final report 307
Tardif R, Plaa GL, Brodeur J. 1992. Influence of various mixtures of inhaled toluene and xylene on the biological
monitoring of exposure to these solvents in rats. Can J Physiol Pharmacol 70:385-395.
Thiel R, Chahoud I 1997. Postnatal development and behaviour of Wistar rats after prenatal toluene exposure. Arch.
Toxicol. 71, 258-265.
Ulfvarson U, Övrum P 1976. Fördelning av lösningsmedel mellan blod och luft. I. Bestämning av
fördelningskoefficienten mellan blod och luft för några lättflyktiga lösningsmedel. Arbete och Hälsa 7, 1-7.
Ullrich D, Gleue C, Krause C, Lusansky C, Nagel R, Schultz C, Seifert B 2002. German Environmental Survey of
children and teenagers 2000 GerES IV: a representative population study including indoor pollutants. In: Proceedings of
the 9
th
International Conference on Indoor Air Quality and Climate, vol 1, 209-214.
Unger E, Alexander A, Fritz T, Rosenberg N and Dreisbach J. 1994. Toluene abuse: physical basis for hypointensity of
the basal ganglia on T2-weighted MR images. Radiology 193:473-6.
Verschueren K 2001. Handbook of Environmental Data on Organic Chemicals. Fourth edition. John Wiley & Sons, Inc,
New York. Vol. 1, 24.
Veulemans H, Masschelein R 1978a. Experimental human exposure to toluene. I. Factors influencing the individual
respiratory uptake and elimination. Int. Arch. Occup. Environ. Health 42, 91-103.
Veulemans H, Masschelein R 1978b. Experimental human exposure to toluene. II. Toluene in venous blood during and
after exposure. Int. Arch. Occup. Environ. Health 42, 105-17.
von Euler G, Ogren SO, Eneroth P, Fuxe K, and Gustafsson JA. 1994. Persistent effects of 80 ppm toluene on
dopamine-regulated locomotor activity and prolactin secretion in the male rat. Neurotoxicology 15:621-4.
von Euler G, Ogren SO, Li XM, Fuxe K, and Gustafsson JA. 1993. Persistent effects of subchronic toluene exposure on
spatial learning and memory, dopamine-mediated locomotor activity and dopamine D2 agonist binding in the rat.
Toxicology 77:223-32.
Von Oettingen WF, Neal PA, Donahue DD. The toxicity and potential dangers of toluene. JAMA 1942;118 8:579-584.
Vrca A, Bozicevic D, Bozikov V, Fuchs R, Malinar M 1997a. Brain stem evoked potentials and visual evoked
potentials in relation to the length of occupational exposure to low levels of toluene. Acta Med. Croatica 51 4-5, 215-9.
Vrca A, Karacic V, Bozicevic D, Fuchs R, Malinar M 1997b. Cognitive evoked potentials VEP P300 persons
occupationally exposed to low concentrations of toluene. Arh. Hig. Rada. Toksikol. 48 3, 277-85.
Waldron HA, Cherry N, Venables H 1982. Solvent exposure and liver function. Lancet 2, 1276.
Weinstein S, Scottolini AG, Bhagavan NV 1985. Low neutrophil alkaline phosphatase in renal tubular acidosis with
hypophosphatemia after toluene sniffing. Clin. Chem. 31, 330-331.
WHO - Recommended health-based limits in occupational exposure to selected organic solvents: Report of a Study
Group. Geneva, World Health Organization, 1981 Technical Report Series, No. 664.
WHO 1986. Environmental Health Criteria, Toluene 52. WHO, Geneva, 1983. Available in the Internet
http://www.inchem.org/documents/ehc/ehc/ehc52.htm.
WHO 1987. Air quality guidelines for Europe. Copenhagen, World Health Organization Regional Office for Europe,
1987 WHO Regional Publications, European Series, No. 23.
WHO 2000. Guidelines for Air Quality. WHO Geneva, Switzerland. Available in the Internet at
http://www.who.int/peh/air/Airqualitygd.htm.
WHO 2001. World Health Organization, Air quality guidelines for Europe 2000. WHO Regional Office for Europe,
WHO Regional Publications, European Series, No. 91, Copenhagen,2001.
Will AM, McLaren EH 1981. Reversible renal damage due to glue sniffing. Br. Med. J. 283, 525-526.
Williams RT 1959. Detoxication Mechanisms. The Metabolism and Detoxication of Drugs, Toxic Substances and other
Organic Compounds. 2
nd Ed. Chapman and Hall Ltd, London.
Wilson RH. Toluene poisoning. JAMA 1943;123 17:1106-1108.
Woiwode W, Drysch K 1981. Experimental exposure to toluene: further consideration of cresol formation in man. Br. J.
Ind. Med. 38, 194-197.
Woiwode W, Wodarz R, Drysch K, Weichardt H 1979. Metabolism of toluene in man: Gas-chromatographic
determination of o-, m- and p-cresol in urine. Arch. Toxicol. 43, 93-98.
Yin S, Li G, Hu Y, Zhang XM, Jin C, Inoue O, Seiji K, Kasahara M, Nakatsuka H, and Ikeda M. 1987. Symptoms and
signs of workers exposed to benzene, toluene, or the combination. Ind. Health. 25 3:113-130.
Zahlsen K, Eide I, Nilsen AM, Nilsen OG 1992. Inhalation kinetics of C6 to C10 aliphatic, aromatic and naphthenic
hydrocarbons in rat after repeated exposures. Pharm. Toxicol. 71, 144-149.
The INDEX project Final report 308
Zavalic, M, Mandic, Z, Turk, R et al. 1998a. Quantitative assessment of color vision impairment in workers exposed to
toluene. Am J Ind Med 32: 297-304.
Zavalic M, Mandic Z, Turk R, Bogadi-Sare A, Plavec D, Gomzi M, and Skender LJ. 1998b. Assessment of colour
vision impairment in male workers exposed to toluene generally above occupational exposure limits. Occup. Med.
Lond. 48:175-80.
The INDEX project Final report 309
Xylenes
Main sources:
Organization Shortcut Document link
EPA – Toxicological Review of Xylenes EPA
635/R-03/001 - In Support of Summary
Information on the Integrated Risk
Information System IRIS
[EPA] http://www.epa.gov/iris/toxreviews/0270.htm
Environmental Protection Agency - Integrated
Risk Information System
[IRIS] http://www.epa.gov/iris/subst/0270.htm
Office of Environmental Health Hazard
Assessment of the Californian Environmental
Protection Agency
[OEHHA1] http://www.oehha.ca.gov/air/chronic_rels/index.html
“ [OEHHA2] http://www.oehha.ca.gov/air/acute_rels/allAcRELs.html
Agency for Toxic Substances and Disease
Registry
[ATSDR] http://www.atsdr.cdc.gov/phshome.html
AIHA - American Industrial Hygiene Association. Odor thresholds for chemicals with established occupational health
standards. Akron OH: AIHA; 1989. p. 29.
Anderson, BE; Zeiger, E; Shelby, MD; et al. 1990 Chromosome aberration and sister chromatid exchange test results
with 42 chemicals. Environ Mol Mutagen. 16: Suppl. 18:55–137.
Andersson, K; Fuxe, K; Nilsen, OG; et al. 1981 Production of discrete changes in dopamine and noradrenaline levels
and turnover in various parts of the rat brain following exposure to xylene, ortho-, meta-, and para-xylene, and
ethylbenzene. Toxicol Appl Pharmacol. 60:535–548.
Arp, EW, Jr; Wolf, PH; Checkoway, H. 1983 Lypmphocytic leukemia and exposures to benzene and other solvents in
the rubber industry. J Occup Med 25:598-602.
Askergren A. 1981. Studies on kidney function in subjects exposed to organic solvents: III. Excretion of cells in the
urine. Acta Med. Scand. 210:103-106.
Askergren A. 1982. Organic solvents and kidney function. In: Mehlman MA, ed. Advances in Modern Environmental
Toxicology. Vol. 2. Princeton Junction, NJ: Senate Press. Pp. 157-172.
Astrand, I; Engström, J; Övrum, P. 1978 Exposure to xylene and ethylbenzene. I. Uptake, distribution, and elimination
in man. Scand J Work Environ Health. 4:185–194.
ATSDR. 1995. Agency for Toxic Substances and Disease Registry. Toxicological Profile for Xylenes Update. Atlanta,
GA: ATSDR. U.S. Printing Office 1995-639-298.
Backes WL, Sequeira DJ, GF Cawley, et al. 1993. Relationship between hydrocarbon structure and induction of P-450:
Effects on protein levels and enzyme activities. Xenobiotica 23 12:1352-1366.
Blanchard KT, Morris JB. 1994. Effects of m-xylene on rat nasal cytochrome P -450 mixed function oxidase activity.
Toxicol Lett 70:253-259.
Bos, RP; Brouns, RME; van Doorn, R; et al. 1981 Non-mutagenicity of toluene, o-, m-, and p-xylene, o-
methylbenzylalcohol and o-methylbenzylsulfate in the Ames assay. Mutat Res. 88:273–279.
Bray, HG; Humphris, BG; Thorpe, WV. 1949 Metabolism of derivatives of toluene: 3. o-, m-, and p-Xylenes. Biochem
J. 45:241–244.
Brown SK 2002. Volatile organic pollutants in new and established buildings in Melbourne, Australia. Indoor Air
12:55-63.
Campbell L, Wilson HK, Samuel AM, et al. 1988. Interactions of m-xylene and aspirin metabolism in man. Br J Ind
Med 45:127-132.
Carlsson, A. 1981 Distribution and elimination of 14C-xylene in rat. Scand J Work Environ Health. 7:51–55.
Carpenter CP, Kinkead ER, Geary DJ Jr, Sullivan LJ, King JM. 1975. Petroleum hydrocarbon toxicity studies: V.
Animal and human response to vapors of mixed xylenes. Toxicol. Appl. Pharmacol. 33:543-558.
Condie, LW; Hill, JR; Borzelleca, JF. 1988 Oral toxicology studies with xylene isomers and mixed xylene. Drug Chem
Toxicol. 11:329–354.
David, A; Flek, J; Frantik, E; et al. 1979 Influence of phenobarbital on xylene metabolism in man and rats. Int Arch
Occup Environ Health. 44:117–125.
The INDEX project Final report 310
Desi, I; Kovacs, F; Zahumenszky, Z; et al. 1967 Maze learning in rats exposed to xylene intoxication.
Psychopharmacologia. 11:224-230.
Donner, M; Maki-Paakkanen, J; Norppa, H; et al. 1980 Genetic toxicology of xylenes. Mutat Res. 74:171–172.
Dudek B, Gralewicz K, Jakubowski M, Kostrzewski P, Sokal J. 1990. Neurobehavioral effects of experimental
exposure to toluene, xylene and their mixture. Pol. J. Occup. Med. 3:109-116.
ECETOC 1986 Joint assessment of commodity chemicals No. 6: Xylenes. Brussels, European Chemical Industry
Ecology and Toxicology Centre.
ECETOC. 1986. European Chemical Industry Ecology and Toxicology Centre. Joint assessment of commodity
chemicals: No 6. Xylenes. Brussels, Belgium: ECETOC.
Edelfors S, Hass U, Ravn-Jonsen A, Lund SP. The effect of ageing and in vitro exposure to xylene and KCl on [Ca2+]i
in synaptosomes from rats exposed prenatally to xylene. Pharmacol Toxicol 1996;78 6:409-412.
Elke K, Jermann E, Begerow J, Dunemann L 1998. Determination of benzene, toluene, ethylbenzene and xylenes in
indoor air at environmental levels using diffusive samplers in combination with headspace solid-phase microextraction
and high-resolution gas chromatography-flame ionization detection. J of Chromatography A 826:191-200.
Elovaara E, Collan Y, Pfaffli P, et al. 1980. The combined toxicity of technical grade xylene and ethanol in the rat.
Xenobiotica 10:435-445.
Elovaara E, Zitting A, Nickels J, et al. 1987. m-Xylene inhalation destroys cytochrome P-450 in rat lung at low
exposure. Arch Toxicol 61:21-26.
Engstrom K, Riihimaki V, Laine A. 1984. Urinary disposition of ethylbenzene and m-xylene in man following separate
and combined exposure. Int Arch Occup Environ Health 54:355-363.
Engstrom, J; Bjurstrom, R. 1978 Exposure to xylene and ethylbenzene. II. Concentration in subcutaneous adipose
tissue. Scand J Work Environ Health. 4:195–203.
EPA 2003. Air Toxics Website. Available in the Internet at http://www.epa.gov/ttnatw01/hlthef/xylenes.html#N_1_
EPA/California 2003, Chronic Reference Exposure Levels Adopted Office of Environmental Health Hazard
Assessment OEHHA as of October 2003. Available in the Internet
http://www.oehha.ca.gov/air/chronic_rels/pdf/xylensREL.pdf.
EXPOLIS 2002. Combined International Database CIDB, KTL- National Public Health Institute, Kuopio Finland.
Fang, Z ; Sonner, J; Laster, MJ; et al. 1996 Anesthetic and convulsant properties of aromatic compounds and
cycloalkanes: Implications for mechanisms of narcosis. Anesth Analg. 83:1097–1104.
Fishbein L 1988 Xylenes: Uses, occurrence and exposure. In: Fishbein L & O'Neill IK ed. Environmental carcinogens:
Methods of analysis and exposure measurement. Volume 10: Benzene and alkylated benzenes. Lyon, International
Agency for Research on Cancer, pp 109-120 IARC Scientific Publications No. 85.
Fishbein, L. 1988 Xylenes: uses, occurrence and exposure. IARC Sci Publ. 85:109-120.
Florin, I; Rutberg, L; Curvall, M; et al. 1980 Screening of tobacco smoke constituents for mutagenicity using the Ames’
test. Toxicology. 15:219–232.
Franchini I, Cavatorta A, Falzoi M, Lucertini S, Mutti A. 1983. Early indicators of renal damage in workers exposed to
organic solvents. Int. Arch. Occup. Environ. Health 52:1-9.
Freundt KJ, Romer KG, Federsel RJ. 1989. Decrease of inhaled toluene, ethyl benzene, m-xylene, or mesitylene in rat
blood after combined exposure to ethyl acetate. Bull Environ Contam Toxicol 42: 495-498.
Gagnaire, F; Marignac B; Langlais C; et al. 2001 Ototoxicity in rats exposed to ortho-, meta-, and para-xylene vapours
for 13 weeks. Pharm Toxicol. 89:6–14.
Gamberale F, Annwall G, and Hultengren M. 1978. Exposure to xylene and ethylbenzene: III. Effects on central
nervous functions. Scand. J. Work Environ. Health 4:204-211.
Gérin, M; Siemiatycki, J; Désy, M; et al. 1998 Associations between several cites of cancer and occupational exposure
to benzene, toluene, xylene, and styrene: results of a case-control study in Montreal. Am J Ind Med 34:144-156.
Gerner-Smidt, P; Friedrich, U. 1978 The mutagenic effect of benzene, toluene, and xylene studied by the SCE
technique. Mutat Res. 58:313–316.
Goldie I. 1960. Can xylene xylol provoke convulsive seizures? Ind. Med. Surg. 29:33-35.
Gralewicz, S; Wiaderna, D. 2001 Behavioral effects following subacute inhalation exposure to m-xylene or
trimethylbenzene in the rat: a comparative study. Neurotoxicology. 22:79–89.
Gralewicz, S; Wiaderna, D; Tomas, T. 1995 Development of spontaneous, age-related nonconvulsive seizure
electrocortical activity and radial-maze learning after exposure to mxylene in rats. Int J Occup Med Environ Health.
8:347–360.
The INDEX project Final report 311
Haglund, U; Lundberg, I; Zech, L. 1980 Chromosome aberrations and sister chromatid exchanges in Swedish paint
industry workers. Scand J Work Environ Health. 6:291–298.
Hake CLR, Stewart RD, Wu A, et al. 1981. p-Xylene: Development of a biological standard for the industrial worker.
Report to the National Institute for Occupational Safety and Health, Cincinnati, OH, by the Medical College of
Wisconsin, Inc, Milwaukee, WI. PB82-152844.
Hass U, Lund SP, and Simonsen L. 1997. Long-lasting neurobehavioral effects of prenatal exposure to xylene in rats.
Neurotoxicol. 18 2:547-552.
Hass U, Lund SP, Simonsen L, and Fries AS. 1995. Effects of prenatal exposure to xylene on postnatal development
and behavior in rats. Neurotoxicol. Teratol. 17 3:341-349.
Hass, U; Jakobsen, BM. 1993 Prenatal toxicity of xylene inhalation in the rat: a teratogenicity and postnatal study.
Pharmacol Toxicol. 73:20–23.
Hastings L, Cooper GP, Burg W. Human sensory response to selected petroleum hydrocarbons. In: Advances in
Modern Environmental Toxicology. Volume VI. Applied Toxicology of Petroleum Hydrocarbons. MacFarland HN,
Holdsworth CE, MacGregor JA, Call RW, and Lane ML, eds. Princeton, New Jersey: Princeton Scientific Publishers,
Inc, 1984. p. 255-270.
HESIS. 1986. Hazard Evaluation System and Information Service, Fact Sheet #7, Xylene. State of California,
Department of Health Services, Department of Industrial Relations, CAL/OSHA. 2151 Berkeley Way, Berkeley CA
94704.
Hipolito, RN. 1980 Xylene poisoning in laboratory workers: case reports and discussion. Lab Med. 11:593–595.
Honma, T; Sudo, A; Miyagawa, M; et al. 1983 Significant changes in the amounts of neurotransmitter and related
substances in rat brain induced by subacute exposure to low levels of toluene and xylene. Indust Health. 21:143–151.
HSDB. 1995. Hazardous Substances Data Bank. On-line data base. Xylenes. Micromedex, Inc. Vol. 25.
HSDB 2003. Hazardous Substances Data Bank. Specialised Information Services. National Library of Medicine.
Available online at http://toxnet.nlm.nih.gov/.
IARC 1989 Xylene. In: Some organic solvents, resin monomers and related compounds, pigments and occupational
exposures in paint manufacture and painting. Lyon, International Agency for Research on Cancer, pp 125-156 IARC
Monographs on the Evaluation of Carcinogenic Risks to Humans, Volume 47.
IARC International Agency for Research on Cancer. 1989 IARC Monographs on the Evaluation of Carcinogenic Risks
to Humans. Vol. 47. Some Organic Solvents, Resin Monomers and Related Compounds, Pigments and Occupational
Exposures in Paint Manufacture and Painting. Lyon, France; pp. 125–156.
Jenkins LJ, Jones RA, and Siegel J. 1970. Long-term inhalation screening studies of benzene, toluene, oxylene, and
cumene on experimental animals. Toxicol. Appl. Pharmacol. 16:818-823.
Kaneko T, Wang P-Y, Sato A. 1993. Enzyme induction by ethanol consumption affects the pharmacokinetics of inhaled
m-xylene only at high levels of exposure. Arch Toxicol 67:473-477.
Kilburn KH, Seidman BC, and Warshaw R. 1985. Neurobehavioral and respiratory symptoms of formaldehyde and
xylene exposure in histology technicians. Arch. Environ. Health 40:229-233.
Klaucke DN, Johansen M, and Vogt RL. 1982. An outbreak of xylene intoxication in a hospital. Am. J. Ind. Med.
3:173-178.
Korsak Z, Sokal JA, Dedyk A, et al. 1988. Toxic effects of combined exposure to toluene and xylene in animals: I.
Acute inhalation study. Pol J Occup Med 1:45-50.
Korsak, Z; Sokal, JA; Górny, R. 1992 Toxic effects of combined exposure to toluene and mxylene in animals. III.
Subchronic inhalation study. Polish J Occup Med Environ Health.5:27–33.
Korsak, Z; Sokal, JA; Wasiela, T; et al. 1990 Toxic effects of acute exposure to particular xylene isomers in animals.
Pol J Occup Med. 3:221–226.
Korsak, Z; Wisniewska-Knypl, J; Swiercz, R. 1994 Toxic effects of subchronic combined exposure to n-butyl alcohol
and m-xylene in rats. Int J Occup Med Environ Health. 7:155–166.
Kumarathasan, P; Otson, R; Chu, I. 1998 Application of an automated HS-GC method in partition coefficient
determination for xylenes and ethylbenzene in rat tissues. Chemosphere. 37:159–178.
Laine A, Savolainen K, Riihimaki V, Matikainen E, Salmi T, Juntunen J. Acute effects of mxylene inhalation on body
sway, reaction times, and sleep in man. Int Arch Occup Environ Health 1993;65:179-188.
Lee SC, Wang B. 2004. Characteristics of emissions of air pollutants from burning of incense in a large environmental
chamber. Atmospheric Environment 38:941-951.
Liira J, Elovaara E, Raunio H, et al. 1991. Metabolic interaction and disposition of methylethylketone and m-xylene in
rats at single and repeated inhalation exposures. Xenobiotica 21:53-64.
The INDEX project Final report 312
Litton Bionetics. 1978a Teratology study in rats - xylene. Final report EPA/OTS Public Files. Litton Bionetics,
Kensington, MD. Document 878210350.
Martinez JS, Sala JJG, Vea AM, et al. 1989. Renal tubular acidosis with an elevated anion gap in a ‘glue sniffer’: Letter
to editor. Human Toxicol. 8:139-140.
McCarroll, NE; Piper, CE; Keech, BH. 1981 An E. coli microsuspension assay for the detection of DNA damage
induced by direct-acting and promutagens. Environ Mutagen. 3:429–444.
Mergler D, Beauvais B. Olfactory threshold shift following controlled 7-hour exposure to toluene and/or xylene.
Neurotoxicology 1992;13 1:211-215.
Mirkova, E, C. Zaikov, G. Antov, A. Mikhailova, L. Khinkova and I. Benchev. 1983. Prenatal toxicity of xylene. J.
Hyg. Epidemiol. Microbiol. Immun. 27: 337-343.
Mohtashamipur, E; Norpoth, K; Woelke, U; et al. 1985 Effects of ethylbenzene, toluene, and xylene on the induction of
micronuclei in bone marrow polychromatic erythrocytes of mice. Arch Toxicol. 58:106–109.
Molnár, J; Paksy, KÁ.; Náray, M. 1986 Changes in the rat’s motor behavior during 4-hr inhalation exposure to
prenarcotic concentrations of benzene and its derivatives. Acta Physiol Hung. 67:349–354.
Morley R, Eccleston DW, Douglas CP, Greville WE, Scott DJ, Anderson J. 1970. Xylene poisoning: A report on one
fatal case and two cases of recovery after prolonged unconsciousness. Br. Med. J. 3:442-443.
Moser, VC; Coggeshall, EM; Balster, RL. 1985 Effects of xylene isomers on operant responding and motor
performance in mice. Toxicol Appl Pharmacol. 80:293–298.
Moszczynski P, and Lisiewicz J. 1983. Occupational exposure to benzene, toluene and xylene and the T lymphocyte
functions. J. Clin. Hematol. Oncol. 13:37-41.
Moszczynski P, and Lisiewicz J. 1984. Occupational exposure to benzene, toluene and xylene and the T lymphocyte
functions. Haematologia 17:449-453.
Nakajima T, Sato A. 1979b. [Metabolic antagonism among benzene, toluene and m-xylene in vitro.] Jpn J Ind Health
21:546-547. Japanese
Nelson KW, Ege JF, Ross M, Woodman LE, Silverman L. Sensory response to certain industrial solvent vapors. J Ind
Hyg Toxicol 1943;25 7:282-285.
Nersesian W, Booth H, Hoxie D, Hinckley W, Shehata T. 1985. Illness in office attributed to xylene [Letter]. Occup.
Health Saf. 54:88.
NTP National Toxicology Program. 1986 Technical Report on the Toxicology and Carcinogenesis of Xylenes mixed
60% m-xylene, 13.6% p-xylene, 17.0% ethylbenzene, and 9.1% o-xylene in F344/N Rats and B6C3F1 mice gavage
studies. Research Triangle Park, NC. NTP TR 327, NIH Publ. No. 86-2583.
Nylén, P; Hagman, M. 1994 Function of the auditory and visual systems, and of peripheral nerve, in rats after long-term
combined exposure to n-hexane and methylated benzene derivatives. II. Xylene. Pharmacol Toxicol. 74:124–129.
Ogata, M; Tomokuni, K; Takatsuka, Y. 1970 Urinary excretion of hippuric acid and m- or pmethylhippuric acid in the
urine of persons exposed to vapours of toluene and m- or p-xylene as a test of exposure. Brit J Indust Med. 27:43–50.
Ogata, M; Yamazaki, Y; Sugihara, R; et al. 1980 Quantitation of urinary o-xylene metabolites of rats and human beings
by high-performance liquid chromatography. Int Arch Occup Environ Health. 46:127–139.
Pap, M; Varga, C. 1987 Sister-chromatid exchanges in peripheral lymphocytes of workers occupationally exposed to
xylene. Mutat Res. 187:223–225.
Pate1 JM, Harper C, Drew RT. 1978. The biotransformation of p-xylene to a toxic aldehyde. Drug Metab Dispos 6:368-
374.
Pryor, GT; Rebert, CS; Howd, RA. 1987 Hearing loss in rats caused by inhalation of mixed xylene and styrene. J Appl
Toxicol. 7:55–61.
Richer, CL; Chakrabarti, S; Senécal-Quevillon, M; et al. 1993 Cytogenic effects of low-level exposure to toluene,
xylene, and their mixture on human blood lymphocytes. Arch Occup Environ Health. 64:581–585.
Riihimaki V, Laine A, Savolainen K, et al. 1982a. Acute solvent-ethanol interactions with special references to xylene.
Stand J Work Environ Health 8:77-79.
Riihimaki V, Savolainen K, Pfaffli P, et al. 1982b. Metabolic interaction between m-xylene and ethanol. Arch Toxicol
49:253-263.
Riihimäki, V. 1979 Conjugation and urinary excretion of toluene and m-xylene metabolites in a man. Scand J Work
Environ Health. 5:135–142.
Riihimäki, V; Savolainen, K. 1980 Human exposure to m-xylene. Kinetics and acute effects on the central nervous
system. Ann Occup Hyg. 23:411–422.
The INDEX project Final report 313
Roberts FP, Lucas EG, Marsden CD, and Trauer T. 1988. Near-pure xylene causing reversible neuropsychiatric
disturbance [Letter]. Lancet ii:273.
Romer KG, Federsel RJ, Freundt KJ. 1986. Rise of inhaled toluene, ethyl benzene, m-xylene, or mesitylene in rat blood
after treatment with ethanol. Bull Environ Contam Toxicol 37:874-876.
Rosen MB, Crofton KM, Chernoff, N. 1986. Postnatal evaluation of prenatal exposure to p-xylene in the rat. Toxicol.
Lett. 34:223-229.
Saarela K, Tirkkonen T, Laine-Ylijoki J, Jurvelin J, Nieuwenhuijsen MJ, Jantunen M 2003. Exposure of population and
microenvironmental distributions of volatile organic compound concentrations in the EXPOLIS study. Atmospheric
Environment 37, 5563-5575.
Sato, A; Nakajima, T. 1979 Partition coefficients of some aromatic hydrocarbons and ketones in water, blood and oil.
Br J Ind Med. 36:231–234.
Savolainen H, Vainio H, Helojoki M, et al. 1978. Biochemical and toxicological effects of short-term, intermittent
xylene inhalation exposure and combined ethanol intake. Arch Toxicol 41:195-205.
Savolainen H, Riihimaki V, and Linnoila M. 1979. Effects of short-term xylene exposure on psychophysiological
functions in man. Int. Arch. Occup. Environ. Health 44:201-211.
Savolainen K, and Linnavuo M. 1979. Effects of m-xylene on human equilibrium measured with a quantitative method.
Acta Pharmacol. Toxicol. 44:315-318.
Savolainen K, and Riihimaki V. 1981. An early sign of xylene effect on human equilibrium. Acta Pharmacol. Toxicol.
48:279-283.
Savolainen K, Kekoni J, Riihimaki V, and Laine A. 1984. Immediate effects of m-xylene on the human central nervous
system. Arch. Toxicol. Suppl 7:412-417.
Savolainen K, Riihimaki V, Laine A, et al. 1982b. Short-term exposure of human subjects to m-xylene and l,l,l-
trichloroethane. Arch Toxicol Suppl 5:96-99.
Savolainen K, Riihimaki V, Laine A. 1982a. Biphasic effects of inhaled solvents on human equilibrium. Acta
Pharmacol Toxicol 51:237-242.
Savolainen K, Riihimaki V, Linnoila M. 1979b. Effects of short-term xylene exposure on psychophysiological
functions in man. Int Arch Occup Environ Health 44:201-211.
Savolainen K, Riihimaki V, Luukkonen R, and Muona O. 1985b. Changes in the sense of balance correlate with
concentrations of m-xylene. Pr. J. Ind. Med. 42:765-769.
Savolainen K, Riihimaki V, Muona O, Kekoni J, Luukkonen R, Laine A. 1985a Conversely exposure-related effects
between atmospheric m-xylene concentrations and human body sense of balance. Acta Pharmacol Toxicol Copenh;57
2:67-71.
Savolainen K, Riihimaki V, Seppalainen AM, Linnoila M 1980a Effects of short-term m-xylene exposure and physical
exercise on the central nervous system. Int Arch Occup Environ Health;45 2:105-21
Savolainen K, Riihimaki V, Vaheri E, et al. 1980b Effects of xylene and alcohol on vestibular and visual functions in
man. Stand J Work Environ Health 6:94-103.
Savolainen K. 1980. Combined effects of xylene and alcohol on the central nervous system. Acta Pharmacol Toxicol
46:366-372.
Savolainen, K; Pfaffli, P. 1980 Dose-dependent neurochemical changes during short-term inhalation exposure to m-
xylene. Arch. Toxicol. 45:117-122.
Sedivec, V; Flek, J. 1976a The absorption, metabolism, and excretion of xylenes in man. Int Arch Occup Environ
Health. 37:205–217.
Senczuk, W; Orlowski, J. 1978 Absorption of m-xylene vapours through the respiratory tract and excretion. Br J Ind
Med. 35:50–55.
Seppalainen AM, Laine A, Salmi T, Verkkala E, Riihimaki V, Luukkonen R. Electroencephalographic findings during
experimental human exposure to m-xylene. Arch Environ Health 1991;46 1:16-24.
Seppalainen AM, Salmi T, Savolainen K, et al. 1983. Visual evoked potentials in short-term exposure of human
subjects to m- xylene and 1,l,l-trichloroethane. Appl Behav Pharmacol Toxicol 1983:349-352.
Silverman DM, Schatz RA. 1991. Pulmonary microsomal alterations following short-term low-level inhalation of para-
xylene in rats. Toxicology 65:27 l-281.
Spirtas, R; Stewart, PA; Lee, JS; et al. 1991 Retrospective cohort mortality study of workers at an aircraft maintenance
facility. I. Epidemiological results. Br J Ind Med 48:515-530.
Tahti, H. 1992 The neurotoxicity of organic solvents, studied with in vitro models. Altern Lab Anim. 20:290–296.
The INDEX project Final report 314
Tardif R, Lapare S, Krishnan K, et al. 1993a. Physiological based modeling of the toxicokinetic interaction between
toluene and m-xylene in the rat. Toxicol Appl Pharmacol 120:266-273.
Tardif R, Lapare S, Krishnan K, et al. 1993b. A descriptive and mechanistic study of the interaction between toluene
and xylene in humans. Int Arch Occup Environ Health, 65:S135-S137.
Tardif R, Lapare S, Plaa GL, et al. 1991. Effect of simultaneous exposure to toluene and xylene on their respective
biological exposure indices in humans. Int Arch Occup Environ Health 63:279-284.
Tardif R, Plaa GL, Brodeur J. 1992. Influence of various mixtures of inhaled toluene and xylene on the biological
monitoring of exposure to these solvents in rats. Can J Physiol Pharmacol 70:385-393.
Taskinen H, Kyyronen P, Hemminki K, Hoikkala M, Lajunen K, Lindbohm ML. 1994. Laboratory work and pregnancy
outcome. J. Occup. Med. 36 3:311-319.
Tatrai E, Ungvary G, Cseh IR, et al. 1981. The effects of long-term inhalation of ortho-xylene on the liver. Ind.
Environ. Xenobiotica. Proceedings of International Conference, Prague, Czechoslovakia, May 27-30, 1980. New York,
NY: Springer-Verlag, pp. 293-300.
Toftgard R, Nilsen OG. 1982. Effects of xylene and xylene isomers on cytochrome P-450 and in vitro enzymatic
activities in rat liver, kidney, and lung. Toxicology 23:197-212.
Uchida Y, Nakatsuka H, Ukai H, Watanabe T, Liu YT, Huang MY et al. 1993. Symptoms and signs in workers exposed
predominantly to xylenes. Int. Arch. Occup. Environ. Health 64:597-605.
Ungváry G. 1985. The possible contribution of industrial chemicals organic solvents to the incidence of congenital
defects caused by teratogenic drugs and consumer goods--an experimental study. Prog Clin Biol Res 160:295-300.
Ungváry, G; Tátrai, E. 1985 On the embryotoxic effects of benzene and its alkyl derivatives in mice, rats and rabbits.
Arch Toxicol, Suppl. 8:425–430.
Ungváry, G; Tátrai, E; Hudák, A; et al. 1980 Studies on the embryotoxic effects of ortho-, meta- and para-xylene.
Toxicology. 18:61–74.
Verschueren K 2001. Handbook of Environmental Data on Organic Chemicals. Fourth edition. John Wiley & Sons, Inc,
New York. Vol. 1, 24.
Wallace L, Nelson W, Ziegenfus R, Pellizzari ED, Michael L, Whitmore R, Zelon H, Hartwell T, Perritt R, Westerdahl
D 1991. The Los Angeles TEAM Study: Personal exposures, indoor-outdoor air concentrations of 25 volatile organic
compounds. Journal of Exposure Analysis and Environmental Epidemiology. 1, 157-192.
Wallace L, Pellizzari ED, Hartwell TD, Sparacino CM, Sheldon L, Zelon H 1985. Personal exposures, indoor-outdoor
relationships, and breath levels of toxic air pollutants measured for 355 persons in New Jersey. Atmospheric
Environment 19, 1651-1661.
Washington, WJ; Murthy, RC; Doye, A; et al. 1983 Induction of morphologically abnormal sperm in rats exposed to o-
xylene. Arch Andrology. 11:233–237.
WHO 1997. Environmental Health Criteria For Xylenes, 190. WHO, Geneva, 1997. Available in the Internet
http://www.inchem.org/documents/ehc/ehc/ehc190.htm.
Wilcosky, TC; Checkoway, H; Marshall, EG; et al. 1984 Cancer mortality and solvent exposures in the rubber industry.
Am Ind Hyg Assoc J 45:809-811.
Wisniewska-Knyp 1 JM, Wronska-Nofer T, Jajte J, et al. 1989. The effect of combined exposures to ethanol and xylene
on rat hepatic microsomal monooxygenase activities. Alcohol 6:347-352.
Wolfe GW. 1988. Subchronic toxicity study in rats with m-xylene. Report by Hazleton Laboratories America, Inc,
Rockville MD. Sponsored by Dynamac Corporation, Rockville, MD.
The INDEX project Final report 315
Styrene
Main sources :
Organization Shortcut Document link
World Health Organization - Air Quality
Guidelines for Europe 2000
[WHO] Not available on web
Environmental Protection Agency - Integrated
Risk Information System
[IRIS] http://www.epa.gov/iris/subst/0104.htm
EPA - Technology Transfer Network – Air
Toxics Website
[EPA] http://www.epa.gov/ttn/atw/hlthef/styrene.html
Office of Environmental Health Hazard
Assessment of the Californian Environmental
Protection Agency
[OEHHA1] http://www.oehha.ca.gov/air/chronic_rels/index.html
“ [OEHHA2] http://www.oehha.ca.gov/air/acute_rels/allAcRELs.html
Agency for Toxic Substances and Disease
Registry
[ATSDR] http://www.atsdr.cdc.gov/phshome.html
AIHA - American Industrial Hygiene Association. Odor thresholds for chemicals with established occupational health
standards. Akron OH: AIHA; 1989. p. 29.
Alarie Y. 1973. Sensory irritation of the upper airways by airborne chemicals. Toxicol Appl Pharmacol 24:279-297.
Andersson, H.C. et al. Chromosomal aberrations and sister-chromatid exchanges in lymphocytes of men occupationally
exposed to styrene in a plastic-boat factory. Mutation research, 73: 387–401 1980.
Anwar, W.A. & Shamy, M.Y. Chromosomal aberrations and micronuclei in reinforced plastics workers exposed to
styrene. Mutation research, 327: 41–47 1995.
Artuso, M. et al. Cytogenetic biomonitoring of styrene-exposed plastic boat builders. Archives of environmental
contamination and toxicology, 29: 270–274 1995.
ATSDR. 1992. Agency for Toxic Substances and Disease Registry. Toxicological Profile for Styrene. Atlanta, GA:
U.S. Department of Health and Human Services, Public Health Service, ATSDR.
Barale, R. The genetic toxicology of styrene and styrene oxide. Mutation research, 257: 107–126 1991.
Behari M, Choudhary C, Roy S, and Maheshwari MC. 1986. Styrene-induced peripheral neuropathy. A case report.
Eur. Neurol. 25:424-427.
Belvedere, G. et al. Styrene oxidation to styrene oxide coupled with arachidonic acid oxidation by soybean
lipoxygenase. Toxicology letters, 18: 39–44 1983.
Bergamaschi E, Smargiassi A, Mutti A, Franchini I, and Lucchini R. 1995. Immunological changes among workers
occupationally exposed to styrene. Int. Arch. Occup. Environ. Health. 67:165-171.
Bigbee, W.L. et al. Glycophorin A GPA somatic cell mutation frequencies in Finnish reinforced plastics workers
exposed to styrene. Cancer epidemiology, biomarkers and prevention, 5: 801–810 1996.
Bonassi, S. et al. Is human exposure to styrene a cause of cytogenetic damage? A reanalysis of the available evidence.
Biomarkers, 1: 217–225 1996.
Bond, G.G. et al. Mortality among workers engaged in the development or manufacture of styrene-based products - an
update. Scandinavian journal of work, environment & health, 18: 145–154 1992.
Brenner, D.D. et al. Biomarkers in styrene-exposed boatbuilders. Mutation research, 261: 225–236 1991.
Brown NA. 1991. Reproductive and developmental toxicity of styrene. Reprod. Toxicol. 5:3-29.
Campagna D, Mergler D, Huel G, Belanger S, Truchon G, Ostiguy C, and Drolet D. 1995. Visual dysfunction among
styrene-exposed workers. Scand. J. Work Environ. Health. 21: 382-390.
Camurri, L. et al. Chromosomal aberrations and sister chromatid exchanges in workers exposed to styrene. Mutation
research, 119: 361–369 1983.
Camurri, L. et al. Sister chromatid exchanges in workers exposed to low doses of styrene. Basic life sciences, 29B:
957–963 1984.
Canadian Environmental Protection Act. Priority Substances List assessment report: Styrene. Ottawa, Canada
Communication Group, 1993.
The INDEX project Final report 316
Carpenter CP, Shaffer CB, Weil CS, Smyth HF Jr. Studies on the inhalation of 1,3 butadiene; with a comparison of its
narcotic effect with benzol, toluol, and styrene, and a note on the elimination of styrene by the human. J Ind Hyg
Toxicol 1944;26:69-77.
Cerny S, Mraz J, Flek J, Tichy M. Effect of ethanol on the urinary excretion of mandelic and phenylglyoxylic acids
after human exposure to styrene. Int Arch Occup Environ Health 1990;62:243-247.
Checkoway H, Echeverria D, Moon J-D, Heyer N, and Costa LG. 1994. Platelet monoamine oxidase B activity in
workers exposed to styrene. Int. Arch. Occup. Health. 66:359-362.
Cherry N, and Gautrin D. 1990. Neurotoxic effects of styrene: further evidence. Br. J. Ind. Med. 47:29-37.
Chia S-E, Jeyaratnam J, Ong C-N, Ng T-P, and Lee H-S. 1994. Impairment of color vision among workers exposed to
low concentrations of styrene. Am. J. Ind. Med. 26:481-488.
Christakopoulos, A. et al. Monitoring of occupational exposure to styrene from hemoglobin adducts and metabolites in
blood. Scandinavian journal of work, environment & health, 19: 255–263 1993.
Coggon, D. et al. Mortality of workers exposed to styrene in the manufacture of glass-reinforced plastics. Scandinavian
journal of work, environment & health, 13: 94–99 1987.
Compton-Quintana, P.J.E. et al. Use of the glycophorin A human mutation assay to study workers exposed to styrene.
Environmental health perspectives, 99: 297–301 1993.
Csanady, G.A. et al. 1994. A physiologic pharmacokinetic model for styrene and styrene-7,8-oxide in mouse, rat and
man. Archives of toxicology, 68: 143–157 1994.
de Jong, G. et al. Cytogenetic monitoring of industrial populations potentially exposed to genotoxic chemicals and of
control populations. Mutation research, 204: 451–464 1988.
DeCeaurriz J, Desiles JP, Bonnet P, et al. 1983. Concentration-dependent behavioral changes in mice following short-
term inhalation exposure to various industrial solvents. Toxicol Appl Pharmacol 67:383-389.
Dolara P, Lodovici M, Salvadori M, Santoni G, Caderni G. Enzyme induction in humans exposed to styrene. Ann
Occup Hyg 1983;27 2:183-188.
Edling C, Ekberg K. No acute behavioral effects of exposure to styrene: a safe level of exposure? Br J Ind Med
1985;42:301-304.
Elovaara E, Vainio H, Aitio A. Pulmonary toxicity of inhaled styrene in acetone-, phenobarbitaland 3-
methylcholanthrene-treated rats. Arch Toxicol 1990;64:365-369.
EPA/California 2003, Chronic Reference Exposure Levels Adopted Office of Environmental Health Hazard
Assessment OEHHA as of August 2003. Available in the Internet
http://www.oehha.ca.gov/air/chronic_rels/AllChrels.html.
EXPOLIS 2002. Combined International Database CIDB, KTL- National Public Health Institute, Kuopio Finland.
Fallas C, Fallas J, Maslard P, and Dally S. 1992. Subclinical impairment of colour vision among workers exposed to
styrene. Br. J. Ind. Med. 49:679-682.
Fleig, I. & Thiess, A.M. Mutagenicity study of workers employed in the styrene and polystyrene processing and
manufacturing industry. Scandinavian journal of work, environment & health, 4: 254–258 1978.
Flodin U, Ekberg K, and Andersson L. 1989. Neuropsychiatric effects of low exposure to styrene. Br. J. Ind. Med.
46:805-808.
Forni, A. et al. Cytogenetic findings in styrene workers in relation to exposure. In: Seemayer, N.H. & Hadnagy, W, ed.
Environmental hygiene, Berlin, Springer, 1988, pp. 159–162.
Foureman GL. 1994. Rationale and derivation of the U.S. Environmental Protection Agency’s inhalation reference
concentration for styrene. Toxic Substances J. 13:283-302.
Fujita H, Koizumi A, Furusawa T, Ikeda M. Decreased erythrocyte d-aminolevulinate dehydratase activity after styrene
exposure. Biochem Pharmacol 1987;36 5:711-716.
Gamberale F, Hultengren M. Exposure to styrene II. Psychological functions. Work Environ Health 1974;11:86-93.
Gamberale F, Lisper HO, and Anshelm-Olson B. 1975. Effect of styrene gases on reaction time among workers in
plastic boat industry. Arbete och Halsa 8:23 Swedish; cited in World Health Organization, 1983.
Gobba F, and Cavelleri A. 1993. Kinetics of urinary excretion and effects on colour vision after exposure to styrene.
IARC Scientific Publ. 127:79-88.
Götell P, Axelson O, Lindelof B. 1972. Field studies on human styrene exposure. Work Environ. Health 9 2: 76-83
Swedish; cited in World Health Organization, 1983.
Governa M, Valentino M, and Visona I. 1994. Chemotactic activity of human polymorphonuclear leukocytes and
industrial xenobiotics: a brief review. Toxicology 91:165-177.
The INDEX project Final report 317
Guillemin M, Bauer D, Martin B, and Marazzi A. 1982. Human exposuire to styrene. IV. Industrial hygiene
investigations and biological monitoring in the polyester industry. Int. Arch. Occup. Environ. Health. 51:139-150.
Guillemin MP, and Berode M. 1988. Biological monitoring of styrene: a review. Am. Ind. Hyg. Assoc. J. 49:497-505.
Hagmar, L. et al. Cytogenetic and hematological effects in plastics workers exposed to styrene. Scandinavian journal of
work, environment & health, 15: 136–141 1989.
Hallier, E. et al. Intervention study on the influence of reduction of occupational exposure to styrene on sister chromatid
exchanges in lymphocytes. International archives of occupational and environmental health, 66: 167–172 1994.
Hansteen, I.L. et al. Low human exposure to styrene in relation to chromosome breaks, gaps and sister chromatid
exchanges. Hereditas, 100: 87–91 1983.
Harkonen H, Lindstrom K, Seppalainen AM, Sisko A Hernberg S. Exposure-response relationship between styrene
exposure and central nervous functions. Scand J Work Environ Health 1978;4:53-59.
Hemminki K, Hesso A. Reaction products of styrene oxide with guanosine in aqueous media. Carcinogenesis 1984;5
5:601-607.
Hoffmann K, Krause C, Seifert B, Ullrich D. 2000. The German Environmental Survey 1990/1992 GerES II: Sources of
Personal Exposure to Volatile Organic Compounds. J. Exp. And Environ. Epid. 10: 115-125.
Hogdson, J.T. & Jones, R.D. Mortality of styrene production, polymerization and processing workers at a site in
Northwest England. Scandinavian journal of work environment & health, 11: 347–352 1985.
Högstedt, B. et al. Increased frequency of chromosome aberrations in workers exposed to styrene. Scandinavian journal
of work environment & health, 5: 333–335 1979.
Högstedt, B. et al. Increased frequency of lymphocyte micronuclei in workers producing reinforced polyester resin with
low exposure to styrene. Scandinavian journal of work environment & health, 9: 241–246 1983.
Horvath, E. et al. 32P-post-labeling detection of DNA adducts in mononuclear cells of workers occupationally exposed
to styrene. Carcinogenesis, 15: 1309–1315 1994.
Hoshika Y, Imamura T, Muto G, Van Gemert LJ, Don JA, Walpot JI. 1993. International comparison of odor threshold
values of several odorants in Japan and in The Netherlands. Environ Res. 1993 Apr;61 1:78-83.
IARC 1979. International Agency for Research on Cancer. Monographs on the evaluation of the carcinogenic risk of
chemicals to man. Some monomers, plastics and synthetic elastomers, and acrolein. Vol. 19. Geneva: World Health
Organization; 1979. p. 231-274
IARC 1994. Some industrial chemicals. Lyon, International Agency for Research on Cancer, 1994, pp. 233–320 IARC
Monographs on the Evaluation of the Carcinogenic Risk of Chemicals to Humans, Vol. 60.
IARC 2002. Monographs on the Evaluation of Carcinogenic Risks to Humans. Some Traditional Herbal Medicines,
Some Mycotoxins, Naphthalene and Styrene. WHO, IARC, 82:437.
Ikeda M, Hirayama T. 1978. Possible metabolic interaction of styrene with organic solvents. Stand J Work Environ
Health 4:41-46.
Ikeda M, Ohtsuji H, Imamura T. 1972. In vivo suppression of benzene and styrene oxidation by co-administered toluene
in rats and effects of phenobarbital. Xenobiotica 2:101-106.
Jablonicka, A. et al. Analysis of chromosomes in peripheral blood lymphocytes of styrene-exposed workers. Mutation
research, 206: 167–169 1988.
Jantunen MJ, Katsouyanni K, Knöppel H, Künzli N, Lebret E, Maroni M, Saarela K, Srám R, Zmirou D. Final Report:
Air Pollution Exposure in European Cities: the EXPOLIS Study. Kuopio, Finland: National Public Health Institute B16,
1999 Annex II: Table 18.
Kankaanpaa JT, Eovaara E, Hemminki K, et al. 1980. The-effect of maternally inhaled styrene on embryonal and foetal
development in mice and Chinese hamsters. Acta Pharmacol Toxicol 47:127-129.
Kelsey, K.T. et al. Sister-chromatid exchanges in lymphocytes from styrene-exposed boat builders. Mutation research,
241: 215–221 1990.
Kishi, R, Y. Katakura, T. Ikeda, B.Q. Chen and H. Miyake. 1992a. Neurochemical effects in rats following gestational
exposure to styrene. Toxicol. Lett. 63: 141-146.
Kishi, R, Y. Katakura, T. Okui, B.Q. Chen, T. Nasu, R.S. Wang, H. Ogawa, T. Ikeda and H. Miyake. 1992b.
Distribution and effects of styrene on the fetus in pregnancy. Jpn. J. Toxicol. Environ. Health. 38: P-2. [Abstract]
Knudsen, L.E. & Sorsa, M. Human biological monitoring of occupational genotoxic exposures. Pharmacology and
toxicology, 72: 86–92 1993.
Kogevinas, M. et al. Cancer mortality in a historical cohort study of workers exposed to styrene. Scandinavian journal
of work environment & health, 20: 249–259 1994.
The INDEX project Final report 318
Kolstad, H.A. et al. Incidence of lymphohematopoietic malignancies among styrene-exposed workers of the reinforced
plastics industry. Scandinavian journal of work environment & health, 20: 272–278 1994.
Kolstad HA, Juel K, Olsen J, and Lynge E. 1995. Exposure to styrene and chronic health effects: mortality and
incidence of solid cancers in the Danish reinforced plastics industry. Occup. Environ. Med. 52:320-327.
Kolstad HA, Bonde JPE, Spano M, Giwercman A, Zschiesche W, Kaae D, and Roeleveld D. 1999a. Sperm chromatin
structure and semen quality following occupational styrene exposure. Scand. J. Work Environ. Health 25 suppl. 1:70-
73.
Kolstad HA, Bisanti L, Roeleveld N, Bonde JPE, and Joffe M. 1999b. Time to pregnancy for men occupationally
exposed to styrene in several European reinforced plastics companies. Scand. J. Work Environ. Health 25 suppl. 1: 66-
69.
Korn, M. et al. Styrene-7,8-oxide in blood of workers exposed to styrene. Archives of toxicology, 68: 524–527 1994.
Lee SC, Wang B. 2004. Characteristics of emissions of air pollutants from burning of incense in a large environmental
chamber. Atmospheric Environment 38:941-951.
Lindbohm, M.-L. et al. Effects of paternal occupational exposure on spontaneous abortions. American journal of public
health, 81: 1029–1033 1991.
Lindbohm M-L. 1993. Effects of styrene on the reproductive health of women: a review. IARC Scientific Publ.
127:163-169.
Lindstrom K, Harkonen H, and Hernberg S. 1976. Disturbances in psychological functions of workers occupationally
exposed to styrene. Scand. J. Work Environ. Health 3:129-139.
Liu, S.F. et al. Detection of styrene oxide-DNA adducts in lymphocytes of a worker exposed to styrene. In: Bartsch, H.
et al, ed. Methods for detecting DNA damaging agents in humans: applications in cancer epidemiology and prevention.
Lyon, International Agency for Research on Cancer, 1988, pp. 217–222 IARC Scientific Publications, No. 89.
Löf, A. & Johanson, G. 1993. Dose-dependent kinetics of inhaled styrene in man. In: Sorsa, M. et al, ed. Butadiene and
styrene: assessment of health hazards. Lyon, International Agency for Research on Cancer, 1993, pp. 89–99 IARC
Scientific Publications, No. 127.
Lorimer WV, Lilis R, Nicholson WJ, Anderson H, Fischbein A, Daum S, et al. Clinical studies of styrene workers:
initial findings. Environ Health Perspect 1976;17:171-181.
Mackay CJ, and Kelman GR. 1986. Choice reaction time in workers exposed to styrene vapour. Human Toxicol. 5:85-
89.
Mäki-Paakkanen, J. Chromosome aberrations, micronuclei and sister-chromatid exchanges in blood lymphocytes after
occupational exposure to low levels of styrene. Mutation research, 189: 399–406 1987.
Mäki-Paakkanen, J. et al. Single-strand breaks, chromosome aberrations, sister chromatid exchanges, and micronuclei
in blood lymphocytes of workers exposed to styrene during the production of reinforced plastics. Environmental and
molecular mutagenesis, 17: 27–31 1991.
Matanoski, G. et al. Cancer epidemiology of styrene-butadiene rubber workers. In: Sorsa, M. et al, ed. Butadiene and
styrene: assessment of health hazards. Lyon, International Agency for Research on Cancer, 1993, pp. 363–374 IARC
Scientific Publications, No. 127.
Matanoski, G.M. et al. Mortality of a cohort of workers in the styrene-butadiene polymer manufacturing industry 1943-
1982. Environmental health perspectives, 86: 107–117 1990.
Matikainen E, Forsman-Gronholm L, Pfaffli P, and Juntunen J. Neurotoxicity in workers exposed to styrene. 1993.
IARC Scientific Publ. 127:153-161.
McDonald, et al. Spontaneous abortion in women employed in plastics manufacture. American journal of industrial
medicine, 14: 9–14 1988.
Meretoja, T. et al. Chromosome aberrations in lymphocytes of workers exposed to styrene. Scandinavian journal of
work, environment & health, 4: 259–264 1978.
Meretoja, T. et al. Occupational styrene exposure and chromosomal aberrations. Mutation research, 56: 193–197 1977.
Moller C, Odkvist L, Larsby B, et al. 1990. Otoneurological findings in workers exposed to styrene. Scand. J. Work
Environ. Health 16 3:189-194.
Moscato G, Biscaldi G, Cottica D, Pugliese F, Candura S, Candura F. Occupational asthma due to styrene: Two case
reports. J Occup Med 1987;29 12:957-960.
Murata K, Araki S, and Yokoyama K. 1991. Assessment of the peripheral, central, and autonomic nervous system
function in styrene workers. Am. J. Ind. Med. 20:775-784.
Murray FJ, John JA, Balmer MF, Schwetz BA. Teratologic evaluation of styrene given to rats and rabbits by inhalation
or by gavage. Toxicology 1978;11:335-343.
The INDEX project Final report 319
Mutti A, Vescovi PP, Falzoi M, Arfini G, Valenti G, and Franchini I. 1984a. Neuroendocrine effects of styrene on
occupationally exposed workers. Scand. J. Work Environ. Health 10:225-228.
Mutti A, Mazzucchi A, Rustichelli P, Frigeri G, Arfini G, and Franchini I. 1984b. Exposure-effect and exposure-
response relationships between occupational exposure to styrene and neurophychological functions. Am. J. Ind. Med.
5:275-286.
Mutti A, Falzoi M, Romanelli MC, Bocchi MC, Ferroni C, and Frandhini I. 1988. Brain dopamine as a target for
solvent toxicity: effects of some monocyclic aromatic hydrocarbons. Toxicology 49:77-82.
Mutti A. 1993. Mechanisms and biomarkers of solvent-induced behavioral and neuroendocrine effects. In: Use of
Biomarkers in Assessing Health and Environmental Impacts of Chemical Pollutants. Travis CC, ed, New York: Plenum
Press. pp 183-199.
Nakajima, T. et al. Styrene metabolisms by cDNA-expressed human hepatic and pulmonary cytochromes P450.
Chemical research and toxicology, 7: 891–896 1994.
New Jersey Department of Health and Senior Services – Hazardous Substances Fact Sheets available on web at:
http://www.state.nj.us/health/eoh/rtkweb
.
Nordenson, I. & Beckman, L. Chromosomal aberrations in lymphocytes of workers exposed to low levels of styrene.
Human heredity, 34: 178–182 1984.
Norppa, H. & Sorsa, M. Genetic toxicity of 1,3-butadiene and styrene. In: Sorsa, M. et al, ed. Butadiene and styrene:
assessment of health hazards. Lyon, International Agency for Research on Cancer, 1993, pp. 185–193 IARC Scientific
Publications, No. 127.
Norppa, H. et al. Chromosome aberrations in lymphocytes of workers exposed to styrene. American journal of
industrial medicine, 2: 299–304 1981.
Okun, A.H. et al. Mortality patterns among styrene-exposed boatbuilders. American journal of industrial medicine, 8:
193–205 1985.
Ortiz de Montellano, P.R. & Catalano, C.E. epoxidation of styrene by hemoglobin and myoglobin. Journal of biology
and chemistry, 260: 9265–9271 1985.
Ott, M.G. et al. A mortality survey of employees in the development or manufacture of styrene-based products. Journal
of occupational medicine, 22: 445–460 1980.
Pekari, K. et al. Biological monitoring of exposure to styrene-assessment of different approaches. Journal of
occupational medicine and toxicology, 2: 115–126 1993.
Perera, F. et al. DNA adducts and related biomarkers in populations exposed to environmental carcinogens.
Environmental health perspectives, 98: 133–137 1992.
Phillips, D.H. & Farmer, P.B. Evidence for DNA and protein binding by styrene and styrene oxide. Critical reviews of
toxicology, 24: S35–S46 1994.
Pohlova, H. & Sram, R.J. Cytogenetic analysis of peripheral blood lymphocytes of workers occupationally exposed to
styrene. Journal of hygiene, epidemiology, microbiology and immunology, 28: 155–161 1985 .
Rappaport SM, Ting D, Jin Z, Yeowell-O’Connell K, Waidyanatha S, McDonald T. Application of Raney Nickel to
measure adducts of styrene oxide with hemoglobin and albumin. Chem Res Toxicol 1993;6:238-244.
Rebert CS, and Hall TA. 1994. The neuroepidemiology of styrene: a critical review of representative literature. Crit.
Rev. Toxicol. 24 Suppl 1:S57-S106.
Santos-Burgoa, C. et al. Lymphohematopoietic cancer in styrene-butadiene polymerization workers. American journal
of epidemiology, 136: 843–854 1992.
Savolainen H. 1977. Some aspects of the mechanisms by which industrial solvents produce neurotoxic effects. Chem.-
Biol. Interact. 18:1-10.
Severi, M. et al. Urinary mandelic acid and hemoglobin adducts in fiberglass-reinforced plastics workers exposed to
styrene. Scandinavian journal of work, environment & health, 20: 451–458 1994.
Sjoborg S, Fregert S, Trulsson L. Contact allergy to styrene and related chemicals. Contact Dermatitis 1984;10:94-96.
Sorsa, M. et al. Styrene revisited – exposure assessment and risk estimation in reinforced plastics industry. In: Gledhill,
B.L. & Mauro, F, ed. New horizons in biological dosimetry. New York, Wiley-Liss, 1991, pp. 187–195.
Spencer HC, Irish DD, Adams EM, et al. 1942. The response of laboratory animals to monomeric styrene. J Ind Hyg
Toxicol 24:295-301.
Stewart RD, Dodd HC, Baretta ED, Schaffer AW. Human exposure to styrene vapor. Arch Environ Health
1968;16:656-662.
Taskinen, H. et al. Spontaneous abortions and congenital malformations among the wives of men occupationally
exposed to organic solvents. Scandinavian journal of work, environment & health, 15: 345–352 1989.
The INDEX project Final report 320
Tates, A.D. et al. Measurement of HPRT mutants, chromosomal aberrations, micronuclei, sister-chromatid exchanges
and cells with high frequencies of SCEs in styrene/dichloromethane-exposed workers. Mutation research, 313: 249–262
1994.
Thiess, A.M. & Fleig, I. Chromosome investigations on workers exposed to styrene/polystyrene. Journal of
occupational medicine, 20: 747–749 1978.
Thiess, A.M. et al. Chromosome investigations in lymphocytes of workers employed in areas in which styrene-
containing unsaturated polyester resins are manufactured. American journal of industrial medicine, 1: 205–210 1980.
Tomanin, R. et al. Chromosome aberrations and micronuclei in lymphocytes of workers exposed to low and medium
levels of styrene. International archives of occupational and environmental health, 64: 209–215 1992.
Treibig G, Schaller K-H, and Valentin H. 1985. Investigations on neurotoxicity of chemical substances at the
workplace. VII. Longitudinal study with determination of nerve conduction velocities in persons occupationally
exposed to styrene. Int. Arch. Occup. Environ. Health. 56:239-247.
Tursi, F. et al. Styrene oxidation to styrene oxide in human erythrocytes is catalyzed by oxyhemoglobin. Experientia,
39: 593–594 1983.
U.S. EPA. 1996. Integrated Risk Information System IRIS. US Environmental Protection Agency, Washington, D.C.
styrene summary last revised 07/01/93,CD-ROM Version. Denver, CO: Microdex, Inc. Edition expires 07/31/96.
Van Hummelen et al. Cytogenetic analysis of lymphocytes from fiberglass-reinforced plastics workers occupationally
exposed to styrene. Mutation research, 310: 157–165 1994.
Verschueren K 2001. Handbook of Environmental Data on Organic Chemicals. Fourth edition. John Wiley & Sons, Inc,
New York. Vol. 1, 24.
Vodicka, P. & Hemminki, K. 32P-Postlabelling of DNA adducts in styrene oxide-modified DNA and in workers
exposed to styrene. In: Sorsa, M. et al, ed. Butadiene and styrene: assessment of health hazards. Lyon, International
Agency for Research on Cancer, 1993, pp. 109–118 IARC Scientific Publications, No. 127.
Vodicka, P. et al. Biomarkers of styrene exposure in lamination workers: levels of O6 guanine DNA adducts, DNA
strand breaks and mutant frequencies in the hypoxanthine guanine phosphoribosyltransferase gene in T-lymphocytes.
Carcinogenesis, 16: 1473–1481 1995.
Vodicka, P. et al. Persistence of O6-guanine DNA adducts in styrene-exposed lamination workers determined by 32P-
postlabelling. Carcinogenesis, 15: 1949–1953 1994.
Walles, S.A.S. et al. Exposure-dependent increase in DNA single-strand breaks in leukocytes from workers exposed to
low levels of styrene. British journal of industrial medicine, 50: 570–574 1993.
Watanabe, T. et al. Chromosome aberrations and sister chromatid exchanges in styrene-exposed workers with reference
to their smoking habits. Environmental mutagenesis, 5: 299–309 1983.
Watanabe, T. et al. Mutagenic potential of styrene in man. Industrial health, 19: 37–45 1981.
WHO 1983. Environmental Health Criteria, Styrene 26. WHO, Geneva, 1983. Available in the Internet
http://www.inchem.org/documents/ehc/ehc/ehc26.htm.
WHO 1987. Air quality guidelines for Europe. Copenhagen, World Health Organization Regional Office for Europe,
1987, pp. 118–126 WHO Regional Publications, European Series, No. 23.
WHO 2001. World Health Organization, Air quality guidelines for Europe 2000. WHO Regional Office for Europe,
WHO Regional Publications, European Series, No. 91, Copenhagen,2001.
WHO. Guidelines for Air Quality 2000a. WHO Geneva, Switzerland. Available in the Internet at
http://www.who.int/peh/air/Airqualitygd.htm.
Wilson, H.K. et al. Effect of alcohol on the kinetics of mandelic acid excretion in volunteers exposed to styrene vapour.
British journal of industrial medicine, 40: 75–80 1983.
Wong, O. A cohort mortality study and a case-control study of workers potentially exposed to styrene in the reinforced
plastics and composites industry. British journal of industrial medicine, 47: 753–762 1990.
Wong, O. et al. An updated cohort mortality study of workers exposed to styrene in the reinforced plastics and
composites industry. Occupational and environmental medicine, 51: 386–396 1994.
Yager, J.W. et al. Sister chromatid exchanges induced in peripheral lymphocytes of workers exposed to low
concentrations of styrene. Progress in clinical and biological research, 340C: 347–356 1990.
Yager, J.W. et al. Sister-chromatid exchanges in lymphocytes are increased in relation to longitudinally measured
occupational exposure to low concentrations of styrene. Mutation research, 319: 155-165 1993.
The INDEX project Final report 321
Ammonia
Main sources:
Authority Shortcut Link to original document
Office of
Environmental Health
Hazard Assessment
of the California
Environmental
Protection Agency
[OEHHA] http://www.oehha.ca.gov/air/chronic_rels/pdf/7664417.pdf
“ [OEHHA2] http://www.oehha.ca.gov/air/acute_rels/allAcRELs.html
EPA Integrated Risk
Information System
[IRIS] http://www.epa.gov/iris/subst/0422.htm
Agency for Toxic
Substances and
Disease Registry
[ATSDR] http://www.atsdr.cdc.gov/toxprofiles/tp126.html
International Program
on Chemical Safety
[IPCS] http://www.inchem.org/documents/hsg/hsg/hsg037.htm
Environmental Health
Criteria - World
Health Orgnization
Geneva, 1986
[EHC54] http://www.inchem.org/documents/ehc/ehc/ehc54.htm#SubSectionNumber:1.5.1
AIHA - American Industrial Hygiene Association. Odor thresholds for chemicals with established occupational health
standards. Akron OH: AIHA; 1989. p. 29.
Ammonia. September 2002. Available in the Internet at http://www.atsdr.cdc.gov/toxprofiles/tp126.html.
Amoore JE & Hautala E 1983 Odor as an aid to chemical safety: odor thresholds compared with threshold limit values
and volatilities for 214 industrial chemicals in air and water dilution. J Appl Toxicol, 3 6: 272-290.
Anderson DP, Beard CW, and Hanson RP. 1964. The adverse effects of ammonia on chickens including resistance to
infection with Newcastle disease virus. Avian. Dis. 8:369379.
ATSDR 2002. Agency for Toxic Substances and Disease Registry Toxicological Profile for Ammonia Draft for Public
Comment. Atlanta, GA: U.S. Department of Health and Human Services, Public Health Service.
Ballal SG, Ali BA, Albar AA, et al. 1998. Bronchial asthma in two chemical fertilizer producing factories in eastern
Saudi Arabia. Int J Tuberc Lung Dis 2 4:330-335.
Broderson JR, Lindsey JR, Crawford JE. 1976. The role of environmental ammonia in respiratory mycoplasmosis of
rats. Am J Pathol 85:115-130.
CCOHS. Canadian Centre for Occupational Health and Safety. Chemical Hazard Summary, Ammonia. CCOHS number
C88-1E. ISBN 1988;0-660-12738-5.
Choudat D, Goehen M, Korobaeff M, et al. 1994. Respiratory symptoms and bronchial reactivity among pig and dairy
farmers. Scand J Work Environ Health 20 1:48-54.
Coon RA, Jones RA, Jenkins LJ Jr, and Siegel J. 1970. Animal inhalation studies on ammonia, ethylene glycol,
formaldehyde, dimethylamine, and ethanol. Toxicol. Appl. Pharmacol. 16 3:646-655.
Cormier Y, Israel-Assayag E, Racine G, et al. 2000. Farming practices and the respiratory health risks of swine
confinement buildings. Eur Resp J 15 3:560-565.
Crump K. A new method for determining allowable daily intakes. Fundam Appl Toxicol 1984;4:860-866.
Dahlman, T. 1956. Mucus flow and ciliary activity in the trachea of healthy rats exposed to respiratory irritant gases
SO2, NH3 and HCHO - a functional and morphologic light microscopic and electron microscopic study, with special
reference to technique: VIII. The reaction of the tracheal ciliary activity to single exposure to respiratory irritant gases
and studies of the pH. Acta Physiol. Scand. 36 Suppl 123: 93-97.
The INDEX project Final report 322
Devos M, Patte F, Rouault J, Laffort P, Van Genert LJ, 1990. Standardized Human Olfactory Thresholds. IRL Press at
Oxforf University Press.
Donham KJ, Cumro D, Reynolds SJ, et al. 2000. Dose-response relationships between occupational aerosol exposures
and cross-shift declines of lung function in poultry workers: recommendations for exposure limits. J Occup Environ
Med 42 3:260-269.
Donham KJ, Reynolds SJ, Whitten P, et al. 1995. Respiratory dysfunction in swine production facility workers: dose-
response relationships of environmental exposures and pulmonary functions. Am J Ind Med 27 3:405-418.
Donham KJ. 1991. Association of environmental air contaminants with disease and productivity in swine. Am J Vet
Res 52 10:1723-1730.
Douglas RB, Coe JE. The relative sensitivity of the human eye and lung to irritant gases. Ann Occup Hyg 1987; 31
2:265-267.
EPA/California 2003, Chronic Reference Exposure Levels Adopted Office of Environmental Health Hazard
Assessment OEHHA as of August 2003. Available in the Internet
http://www.oehha.ca.gov/air/chronic_rels/AllChrels.html
.
Ferguson WS, Koch WC, Webster LB, et al. 1977. Human physiological response and adaption to ammonia. J Occup
Med 19:319-326.
Fisher ML, Littlejohn D, Lunden MM, Brown NJ 2003. Automated Measurements of ammonia and nitric acid in indoor
and outdoor air. Environ Sci Tech 37, 2114-2119.
Flury, K.E, D.E. Dines, J.R. Rodarte and R. Rodgers. 1983. Airway obstruction due to inhalation of ammonia. Mayo
Clin. Proc. 58: 389-393.
Fürst P, Josephson B, Maschio G, et al. 1969. Nitrogen balance after intravenous and oral administration of ammonium
salts to man. J Appl Phys 26:13-22.
Gamble, M.R. and G. Clough. 1976. Ammonia build-up in animal boxes and its effect on rat tracheal epithelium. Lab.
Anim. 10: 93-104.
Gay WMB, Crane CW, Stone WD. 1969. The metabolism of ammonia in liver disease: A comparison of urinary data
following oral and intravenous loading of [15N]ammonium lactate. Clin Sci 37:815-823.
Gomzi M 1999. Indoor air and respiratory health in preadolescent children. Atmospheric Environment 33, 4081-4086
Guyton AC. 1981. The body fluids and kidneys. Textbook of medical physiology. Philadelphia: WB Saunders
Company.
Heederik D, van Zwieten R, Brouwer R. 1990. Across-shift lung function changes among pig farmers. Am J Ind Med
17 1:57-58.
Heederik D, Brouwer R, Biersteker K, et al. 1991. Relationship of airborne endotoxin and bacteria levels in pig farms
with the lung function and respiratory symptoms of farmers. Int Arch Occup Environ Health 62 8:595-601.
Henderson Y, Haggard HW. Noxious gases. 2nd ed. New York: Reinhold Publishing Corporation; 1943.
Herrera L, Kazemi H. 1980. Interaction between ammonia and CO2 as ventilatory stimulants. Bull Eur Physiopathol
Respir 16:459-468.
Holness DL, Purdham JT, Nethercott JR. 1989. Acute and chronic respiratory effects of occupational exposure to
ammonia. Am Ind Hyg Assoc J 50 12:646-650.
HSDB 2003. Hazardous Substances Data Bank. Specialised Information Services. National Library of Medicine.
Available online at http://toxnet.nlm.nih.gov/.
Hyang-Suk K, Byung-Kee C 1993. Ammonia odor concentrations in toilets by ion chromatography. Proceedings of the
6
th
International Conference on Indoor Air Quality and Climate, vol 1, 71-75.
Industrial Bio-Test Laboratories, Inc. 1973. Irritation threshold evaluation study with ammonia. Report to international
institute of ammonia refrigeration IBT No. 663-03160. Northbrook, Ill: March 23, 1973. Industrial Bio-Test
Laboratories, Inc. [unpublished study to be peer reviewed]
Landahl HD, Herrmann RG. 1950. Retention of vapors and gases in the human nose and lung. Arch Ind Hyg Occup
Med 1:36-45.
The INDEX project Final report 323
Larson T, Frank R, Covert D, et al. 1980. The chemical neutralization of inhaled sulfuric acid aerosol. Am J Ind Med
1:449-452.
Leaderer BP, Stowe M, Li R, Sullivan J, Koutrakis P, Wolfson JM, Wilson W 1993. Residential levels of particle and
vapor phase acids associated with combustion sources. In: Proceedings of the 6
th
International Conference on Indoor Air
Quality and Climate, vol 3, 147 – 152.
Leonardos G, Kendall D, Barnard N. 1969. Odor threshold determinations of 53 odorant chemicals. J Air Pollut Control
Assoc 19:91-95.
MacEwen JD, Theodore J, Vernot EH. 1970. In: Proceedings 1st annual conference on environmental toxicology.
Human exposure to EEL concentrations of monomethylhydrazine AMRL-TR-70-102. Wright-Patterson Air Force Base,
Ohio: Wright-Patterson Air Force Base, 355-363.
Melbostad E, Eduard W. 2001. Organic dust-related respiratory and eye irritation in Norwegian farmers. Am J Ind Med
39 2:209-217.
OEHHA 1999. Office of Environmental Health Hazard Assessment of the Californian Environmental Protection
Agency.Chronic Reference Exposure Levels Adopted by OEHHA available on the Internet at
http://www.arb.ca.gov/toxics/tac/factshts/acetalde.pdf
Crump KS, Howe R. Probit-A computer program to extrapolate quantile animal toxicological data to low doses. Ruston
LA: KS Crump & Company, Inc.; 1983.
Pitts RF. 1971. The role of ammonia production and excretion in regulation of acid-base balance. N Engl J Med 284:32-
38.
Puhakka E, Joutsiniemi J, Karkkainen J 2000. Remedial measures against high concentrations of ammonia in buildings.
In Proceedings of Healthy Buildings Conference, Espoo, Finland, 6-10 August 2000, vol 3, 451- 456.
Reynolds SJ, Donham KJ, Whitten P, et al. 1996. Longitudinal evaluation of dose-response relationships for
environmental exposures and pulmonary function in swine production workers. Am J Ind Med 29 1:33-40.
Riala R, Korhonen K, Hoikkala M, Gustafsson D 1993 Air quality in the storage of archival records. In: Proceedings of
the 6
th
International Conference on Indoor Air Quality and Climate, vol 3, 247 – 250.
Richard D, Bouley G, Boudene C. 1978. [Effects of continuous inhalation of ammonia in the rat and mouse.] Bull Eur
Physiopathol Respir 14:573-582. French
Richards P, Brown CL, Houghton BJ, et al. 1975. The incorporation of ammonia nitrogen into albimum in man: the
effects of diet, uremia and growth hormone. Clin Nephrol 3 5:172-179.
Ruth JH. Odor thresholds and irritation levels of several chemical substances: a review. Am Ind Hyg Assoc J 1986;47
3:A142-A151.
Saarela K 2003. Personal communication.
Saarinen A, Vartiala T, Viinikka M 2002. Influence of the inhabitants on indoor air quality. Proceedings of the 9
th
International Conference on Indoor Air Quality and Climate, vol 2, 174-178.
Schoeb TR, Davidson MK, and Lindsey JR. 1982. Intracage ammonia promotes growth of Mycoplasma pulmonis in the
respiratory tract of rats. Infect. Immun. 38:212-217.
Sekizawa SI, Tsubone H. 1994. Nasal receptors responding to noxious chemical irritants. Respir Physiol 96 1:37-48.
Shim C, Williams MH. Effect of odors in asthma. Am J Med 1986;80:18-22.
Silverman L, Whittenberger JL, Muller J. 1949. Physiological response of man to ammonia in low concentrations. J Ind
Hyg Toxicol 31:74-78.
Summerskill WHJ, Wolpert E. 1978. Ammonia metabolism in the gut. Am J Clin Nutr 23:633-639.
Takagaki G, Berl S, Clarke DD, et al. 1961. Glutamic acid metabolism in brain and liver during infusion with ammonia
labelled with nitrogen-15. Nature 189:326.
Tuomainen M, Pirinen J 2002. TVOC, formaldehyde, and ammonia levels in two new block of flats. Proceedings of the
9
th
International Conference on Indoor Air Quality and Climate, vol 1, 244-248.
The INDEX project Final report 324
U.S.EPA 1979. Effects of endogenous ammonia on neutralization of inhaled sulfuric acid aerosols. Cininnatti, OH: U.S.
Environmental Protection Agency, Health Effects Research Lab. EPA-600/1-79-045PB 80 147 978.
U.S.EPA 1991. Integrated Risk Information System IRIS. Reference concentration RfC for ammonia. Online. National
Center for Environmental Assessment, Cincinnati, OH. Available at: http://www.epa.gov/iris/subst/0422.htm
Utell MJ, Mariglio JA, Morrow PE, et al. 1989. Effects of inhaled acid aerosols on respiratory function: The role of
endogenous ammonia. J Aerosol Med 2:141-147.
Verberk MM. 1977. Effects of ammonia in volunteers. Int Arch Occup Environ Health 39:73-81.
Verschueren K 2001. Handbook of Environmental Data on Organic Chemicals. Fourth edition. John Wiley & Sons, Inc,
New York. Vol. 1, 24.
Vogelzang PFJ, van der Gulden JWJ, Folgering H, et al. 2000. Longitudinal changes in bronchial responsiveness
associated with swine confinement dust exposure. Chest 117 5:1488-1495.
Vogelzang PFJ, van der Gulden JWJ, Preller L, et al. 1997. Bronchial hyperresponsiveness and exposure in pig farmers.
Int Arch Occup Environ Health 70 5:327-333.
VTT 2003. Indoor air database. VTT – Technical Research Centre of Finland, Espoo, Finland.
Wands RC. 1981. Alkaline materials. In: Clayton GD, Clayton FE, eds. Patty's industrial hygiene and toxicology, Vol.
II, 4th ed. New York, NY: John Wiley & Sons, Inc, 3045-3069.
Warren KS, Schenker S. 1964. Effect of inhibitor glutamine synthesis methionine sulfoximine on ammonia toxicity and
metabolism. J Lab Clin Med 64 3:442-449.
Way J, Baxter L, McGuinn D, Zitzer A, Petrikovics I. Ch. 9. Occupational hazards and ocular toxicity. In: Chiou GCY,
editor. Ophthalmic toxicology. Raven Press; 1992.
WHO 1997. Environmental Health Criteria. Nitrogen Oxides, 2nd ed. 188. WHO, Geneva, 1997. Available in the
Internet http://www.inchem.org/documents/ehc/ehc/ehc188.htm.
WHO. 1986. Ammonia. Environmental Health Criteria. Vol. 54. Geneva, Switzerland: World Health Organization.
The INDEX project Final report 325
Limonene
Main sources:
Authority Shortcut Link to original document
WHO – International
Programme on Chemical Safety
[WHO] http://www.inchem.org/documents/cicads/cicads/cicad05.htm
EPA Integrated Risk Information
System
[IRIS] http://www.epa.gov/iris/subst/0682.htm#refinhal
National Industrial Chemicals
Notification and Assessment
Scheme NICNAS - Australia
[NICNAS] http://www.nicnas.gov.au/publications/CAR/PEC/PEC22/PEC22_whole.pdf
AIHA 1993. American Industrial Hygiene Association. Workplace environmental exposure level guide: d-limonene
Ariyoshi T, Arakaki M, Ideguchi Y, Noda K and Ide H. 1975. Studie on the metabolism of d-limonene. III. Effects of d-
limonene on the lipids and drug metabolising enzymes in rat livers. Xenobiotica, 5: 33-38.
Brown VM, Coward SKD, Crump DR, Llewellyn JW, Mann HS, Raw GJ 2002. Indoor air quality in English homes –
VOCs. In: Proceedings of the 9
th
International Conference on Indoor Air Quality and Climate, vol 4, 477-482.
Clausen PA, Wilkins CK, Wolkoff P, Nielsen GD. 2001. Chemical and biological evaluation of a reaction mixture of R-
+-limonene/ozone: formation of strong airway irritants. Environ Int. 2001 Jun;26 7-8:511-22.
Crowell PL, Elegbede JA, Elson CE, Lin S, Vedejs E, Cunningham D, Bailey HH, Gould MN 1992. Human
metabolism of orally administered d-limonene. Proceedings of the American Association for Cancer Research, 33:524.
EPA 1994 Prevention, Pesticides And Toxic Substances. Re-registration Eligibility Decision RED, Limonene, 738-R-
94-034. Available at http://www.epa.gov/REDs/3083.pdf.
EXPOLIS 2002. Combined International Database CIDB, KTL- National Public Health Institute, Kuopio Finland.
Falk A, Gullstrand E, Löf A, Wigaeus Hjelm E 1990. Liquid/air partition coefficients of four terpenes. British journal of
industrial medicine, 47:62-64.
Falk A, Fischer T, Hagberg M 1991. Purpuric rash caused by dermal exposure to d-limonene. Contact dermatitis,
25:198-199.
Falk Filipsson A, Löf A, Hagberg M, Wigaeus Hjelm E, Wang Z 1993. d-Limonene exposure to humans by inhalation:
Uptake, distribution, elimination, and effects on the pulmonary function. Journal of toxicology and environmental
health, 38:77-88.
Finnish Institute of Occupational Health 2001.Finnish Occupational Exposure limits 2000. Unofficial translation,
http://www.occuphealth.fi/ttl/projekti/htp/english
Flavor and Extract Manufacturers Association 1991 d-Limonene monograph. Washington, DC, pp. 1-4.
Hoffmann K, Krause C, Seifert B, Ullrich D. The German Environmental Survey 1990/1992 GerES II: Sources of
Personal Exposure to Volatile Organic Compounds. J. Exp. And Environ. Epid. 2000; 10: 115-125.
HSDB 2003. Hazardous Substances Data Bank. Specialised Information Services. National Library of Medicine.
Available online at http://toxnet.nlm.nih.gov/.
IARC 1993. Monographs on the evaluation of carcinogenic risks to umans. Vol 56. International Agency for Research
on Cancer, Lyon, France, 1993, pp 135-162
IARC 1999. Some chemicals that cause tumours of the kidney or urinary bladder in rodents and some other substances.
Lyon, International Agency for Research on Cancer IARC Monographs on the evaluation of carcinogenic risks to
humans, Vol.73
Igimi H, Nishimura M, Kodama R, Ide H 1974. Studies on the metabolism of d-limonene p-mentha-1,8-diene. I. The
absorption, distribution and excretion of d-limonene in rats. Xenobiotica, 4:77-84.
Igimi H, Hisatsugu T, Nishimura M 1976. The use of d-limonene preparation as a dissolving agent of gallstones.
Digestive diseases and science, 21:926-939.
Igimi H, Tamura R, Toraishi K, Yamamoto F, Kataoka A, Ikejiri Y, Hisatsugu T, Shimura H 1991. Medical dissolution
of gallstones. Clinical experience of d-limonene as a simple, safe and effective solvent. Digestive diseases and science,
36:200-208.
IPCS 1994. Assessing health risks of chemicals: Derivation of guidance values for health-based exposure limits.
Geneva, World Health Organization, International Programme on Chemical Safety Environmental Health Criteria 170.
Jantunen MJ, Katsouyanni K, Knöppel H, Künzli N, Lebret E, Maroni M, Saarela K, Srám R, Zmirou D. Final Report:
Air Pollution Exposure in European Cities: the EXPOLIS Study. Kuopio, Finland: National Public Health Institute B16,
1999 Annex II: Table 18.
The INDEX project Final report 326
Karlberg A.T, Magnusson K, Nilsson U 1992. Air oxidation of d-limonene the citrus solvent creates potent allergens.
Contact dermatitis, 26:332-340.
Karlberg A.T. and Lindell B. 1993. Limonene. In: Beije B, Lundberg P, eds. Criteria documents from Nordic Expert
Group 1993. Solna, National Institute of Occupational Health, Nordic Council of Ministers.
Kleno J, Wolkoff P. 2004. Changes in eye blink frequency as a measure of trigeminal stimulation by exposure to
limonene oxidation products, isoprene oxidation products and nitrate radicals. Int Arch Occup Environ Health. 2004
Mar 6 [Epub ahead of print].
Kodama R, Yano T, Furukawa K, Noda K, Ide H 1976. Studies on the metabolism of d-limonene p-mentha-1,8-diene.
IV. Isolation and characterization of new metabolites and species differences in metabolism. Xenobiotica, 6:377-389.
Kostiainen R. Volatile Organic Compounds in the Indoor Air of Normal and Sick Houses. Atmos Environ 1995; 29:
693-702.
Larsen ST, Hougaard KS, Hammer M, Alarie Y, Wolkoff P, Clausen PA, Wilkins CK, Nielsen GD. 2000. Effects of R-
+- and S- --limonene on the respiratory tract in mice. Hum Exp Toxicol. 2000 Aug;19 8:457-66.
Leffingwell & Associates. Services and Software for the Perfume, Flavor, Food and Beverage Industries. Available in
the Internet at: http://www.leffingwell.com/index.htm
Maroni M, Seifert B, Lindvall T eds 1995. A Comprehensive Reference Book. Air Quality Monographs Vol. - 3.
Elsevier Science B.V, Amsterdam, The Netherlands. p 49, 56.
Matthys H, de Mey C, Carls C, Rys A, Geib A, Wittig T. 2000. Efficacy and tolerability of myrtol standardized in acute
bronchitis. A multi-centre, randomised, double-blind, placebo-controlled parallel group clinical trial vs. cefuroxime and
ambroxol. Arzneimittelforschung. 2000 Aug;50 8:700-11.
Mølhave L, Kjaergaard SK, Hempel-Jorgensen A, Juto JE, Andersson K, Stridh G, Falk J. 2000 The eye irritation and
odor potencies of four terpenes which are major constituents of the emissions of VOCs from Nordic soft woods. Indoor
Air. 2000 Dec;10 4:315-8
NICNAS 2002. National Industrial Chemicals Notification and Assessment Scheme. Priority Existing Chemicals
Assessment Report No.22 – Limonene. ISBN 0 642 50190 4. Commonwealth of Australia 2002. Also available in the
Internet at: http://www.nicnas.gov.au/publications/CAR/PEC/PEC22/PEC22_whole.pdf
Pellizzari ED, Hartwell TD, Harris BSH III, Wadell RD, Whitaker DA, Erickson MD 1982. Purgeable organic
compounds in mother's milk. Bulletin of environmental contamination and toxicology, 28:322-328.
Rohr AC, Wilkins CK, Clausen PA, Hammer M, Nielsen GD, Wolkoff P and Spengler JD. 2002. Upper airway and
pulmonary effects of oxidation products of +-alpha-pinene, d-limonene, and isoprene in BALB/c mice. Inhal Toxicol.
2002 Jul;14 7:663-84.
RTECS 2001. Registry of toxic effects of chemical substances. National Institute for Occupational Safety and Health,
Cincinnati, Ohio
Smith OW, Wade AP, Dean FM 1969. Uroterpenol, a pettenkofer chromogen of dietary origin and a common
constituent of human urine. Journal of endocrinology, 45:17-28.
U.S. EPA. 1991. Alpha2u-globulin: Association with chemically-induced renal toxicity and neoplasia in the male rat.
Washington, DC. EPA/625/3-91/019F.
U.S. EPA 1993. Integrated Risk Information System IRIS. d-Limonene. Online. National Center for Environmental
Assessment, Cincinnati, OH. Available at: http://www.epa.gov/iris/subst/0682.htm#refinhal
Verschueren K 2001. Handbook of Environmental Data on Organic Chemicals. Fourth edition. John Wiley & Sons, Inc,
New York. Vol. 1, 24.
Wallace L, Nelson W, Ziegenfus R, Pellizzari ED, Michael L, Whitmore R, Zelon H, Hartwell T, Perritt R, Westerdahl
D 1991. The Los Angeles TEAM Study: Personal exposures, indoor-outdoor air concentrations of 25 volatile organic
compounds. Journal of Exposure Analysis and Environmental Epidemiology. 1, 157-192.
Watabe T, Hiratsuka A, Osawa N, Isobe M 1981. A comparative study on the metabolism of d-limonene and 4-
vinylcyclohex-1-ene by hepatic microsomes. Xenobiotica, 11:333-344.
Webb DR, Ridder GM, Alden CL 1989 Acute and subchronic nephrotoxicity of d-limonene in Fischer 344 rats. Food
and chemical toxicology, 27:639-649.
Weschler C.J. and Shields H.C. 1997. Potential reactions among indoor pollutants. Atmospheric Environment, 31:
3487-3495.
WHO 1989. Indoor Air Quality: Organic Pollutants. European Reports and Studies No. 111. 1989. Copenhagen,
Denmark.
WHO 1998. International Programme on Chemical Safety IPCS Concise International Chemical Assessment Document
CICAD No.5 Limonene. Available in the Internet at: http://www.inchem.org/documents/cicads/ cicads/cicad05.htm
The INDEX project Final report 327
Wilkins CK, Wolkoff P, Clausen PA, Hammer M, Nielsen GD. 2003. Upper airway irritation of terpene/ozone
oxidation products TOPS. Dependence on reaction time, relative humidity and initial ozone concentration. Toxicol
Lett.2003;143 2:109-14.
Wolkoff P, Clausen P.A, Jensen B. and Nielsen G.D. 1997. Are we measuring the relevant indoor pollutants ? Indoor
Air, 7, 92-106.
Wolkoff P, Clausen PA, Wilkins CK, Nielsen GD. 2000. Formation of strong airway irritants in terpene/ozone
mixtures. Indoor Air. 2000 Jun;10 2:82-91
The INDEX project Final report 328
alpha-Pinene
Baxter, C. S. 2001. Alicyclic Hydrocarbons. In: Patty’s Toxicology, vol. 8: Chapter 50. 5th ed. Bingham, E, B.
Cohrssen, and C.H. Powell Eds.. John Wiley and Sons, Inc, New York, NY; pp. 710-1080.
Bingham, E, Cohrssen, B, Powell, C.H. 2001. Patty's Toxicology Volumes 1-9 5th ed. John Wiley & Sons. New York,
N.Y. 2001.V4 P212 peer reviewed
Brun R. 1975. Epidemiology of contact dermatitis in Geneva 1000 cases. Contact Dermatitis 1975;1:214-7.
Budavari, S. ed.. The Merck Index - Encyclopedia of Chemicals, Drugs and Biologicals. Rahway, NJ: Merck and Co,
Inc, 1989. 1545
Budavari, S. 1996. The Merck Index, 12th ed. Merck & Co, Inc, Whitehouse Station, NJ.
Bystrom, L. 2000. Pine: A nutritional supplement/medicinal, toxicant and effective repellent. Available on the Internet
at: http://www.ansci.cornell.edu/courses/as625/2000term/pine/pine.htm. Last accessed August 9
, 2001.
Chinn, H. 1989. Turpentine-United States CEH Data Summary. In: Chemical Economics Handbook, SRI International,
pages 596.5000A-596.5000O.
Clausen PA, Wilkins CK, Wolkoff P, Nielsen GD. 2001. Chemical and biological evaluation of a reaction mixture of R-
+-limonene/ozone: formation of strong airway irritants. Environ Int. 2001 Jun;26 7-8:511-22.
Connor T. H, Theiss, J.C, Hanns HA; Monteith DK. and Matney TS. 1985. Genotoxicity of organic chemicals
frequently found in the air of mobile homes. Toxicology Letters, 25 33-40 1985.
Dooms-Goossens H, Degreef H, Holvoet C, Maertens M. 1977. Turpentine-induced hypersensitivity to peppermint oil.
Contact Dermatitis 1977;3:304-8.
Edwards R, Jurvelin J, Saarela K, Jantunen M. 2001. VOC concentrations measured in personal samples and residential
indoor, outdoor and workplace microenvironments in EXPOLIS-Helsinki, Finland. Atmos Environ 35:4531-4543.
Ellenhorn MJ & Barceloux DG ed 1988 Medical toxicology diagnosis and treatment of human poisoning. New York,
Elsenin, pp 820-829.
Eriksson K, Levin J-O. 1990. Identification of cis and trans verbenol in human urine after occupational exposure to
terpenes. Int Arch Occup Environ Health 1990;62:379-83.
Eriksson KA, Levin JO, Sandstrom T, Lindstrom-Espeling K, Linden G, Stjernberg NL. 1997. Terpene exposure and
respiratory effects among workers in Swedish joinery shops. Scand J Work Environ Health. 1997 Apr;23 2:114-20.
Eriksson KA, Stjernberg NL, Levin JO, Hammarstrom U, Ledin MC. 1996. Terpene exposure and respiratory effects
among sawmill workers. Scand J Work Environ Health. 1996 Jun;22 3:182-90.
EXPOLIS 2002. Combined International Database CIDB, KTL- National Public Health Institute, Kuopio Finland.
Falk A, Hagberg M, Lof A, Wigaeus-Hjelm E, Wang Z. 1990a. Uptake, distribution and elimination of alpha-pinene in
man after exposure by inhalation. Scand J Work Environ Health 1990;16:372-8.
Falk A, Gullstrand E, Lof A, Wigeaus-Hjelm E. 1990b. Liquid/air partition coefficients of four terpenes. Br J Ind Med
1990;47:62-4.
Falk Filipsson Agneta 1996. Short term inhalation exposure to turpentine: toxicokinetics and acute effects in men.
Occupational & Environmental Medicine, 53 2:100-105.
Florin I, Rutberg L, Curvall M. and Enzell C. 1980. Screening of tobacco smoke constituents for mutagenicity using the
Ames test. Toxicology, 18 219-232 1980.
Hedenstierna G, Alexandersson R, Wimander K, Rosen G. 1983. Exposure to terpenes: effects on pulmonary function.
Int Arch Occup Environ Health. 1983;51 3:191-8.
Hoffmann K, Krause C, Seifert B, Ullrich D. The German Environmental Survey 1990/1992 GerES II: Sources of
Personal Exposure to Volatile Organic Compounds. J. Exp. And Environ. Epid. 2000; 10: 115-125.
HSDB, 1985. Hazardous Substance Fact Sheet. Turpentine. Trenton, NJ: New Jersey Department of Health.
HSDB 2003. Hazardous Substances Data Bank. Specialised Information Services. National Library of Medicine.
Available online at http://toxnet.nlm.nih.gov/.
Kasanen, J. -P, A. -L. Pasanen, P. Pasanen, J. Liesivuori, V. -M. Kosma, and Y. Alarie. 1998. Stereospecificity of the
sensory irritation receptor for nonreactive chemicals illustrated by pinene enantiomers. Arch. Toxicol. 72:514-523.
Cited by Kasanen et al, 1999.
Kasanen, J. -P, A. -L. Pasanen, P. Pasanen, J. Liesivuori, V. -M. Kosma, and Y. Alarie. 1999. Evaluation of sensory
irritation of 3-carene and turpentine, and acceptable levels of monoterpenes in occupational and indoor environment.
J. Toxicol. Environ. Health 57:89-114.
The INDEX project Final report 329
Key, M. M, et al. 1977. In: Occupational Diseases: A Guide to their Recognition, rev. ed, U.S. Department of Health,
Education, and Welfare, Washington, D.C. Cited by Baxter, 2001.
Koppel C, Tenczer J, Tonnesmann U, Schirop T, Ibe K. Acute poisoning with pine oil-metabolism of monoterpenes.
Arch Toxicol 1981;49:73-8.
Kostiainen R. 1995. Volatile Organic Compounds in the Indoor Air of Normal and Sick Houses. Atmos Environ 29:
693-702.
Leffingwell & Associates - Services and Software for the Perfume, Flavor, Food and Beverage Industries – in the
Internet: http://www.leffingwell.com/index.htm
Levin J-O, Eriksson K, Falk A, Lof A. Renal elimination of verbenols in man following experimental alpha-pinene
inhalation exposure. Int Arch Occup Environ Health 1992;63:571-3.
Maroni M, Seifert B, Lindvall T eds 1995. A Comprehensive Reference Book. Air Quality Monographs Vol. - 3.
Elsevier Science B.V, Amsterdam, The Netherlands.
Mølhave L, Kjaergaard SK, Hempel-Jorgensen A, Juto JE, Andersson K, Stridh G, Falk J. 2000. The eye irritation and
odor potencies of four terpenes which are major constituents of the emissions of VOCs from Nordic soft woods. Indoor
Air. 2000 Dec;10 4:315-8.
NIEHS, 2002. National Institute of Environmental Health Sciences. Turpentine Turpentine Oil, Wood Turpentine,
Sulfate Turpentine, Sulfite Turpentine: Review of Toxicological Literature. Research Triangle Park, North Carolina
27709. Available in the Internet: http://ntp-server.niehs.nih.gov/htdocs/Chem_Background/ExSumPdf/Turpentine.pdf
Rehwagen M, Schlink U, Herbarth O 2003. Seasonal cycle of VOCs in apartments. Indoor Air 13:283-291.
Rockwell P, and Raw I. 1979. A mutagenic screening of various herbs, spices, and food additives. Nature and Cancer, 1
4 10-15 1979.
Rohr AC, Wilkins CK, Clausen PA, Hammer M, Nielsen GD, Wolkoff P and Spengler JD. 2002. Upper airway and
pulmonary effects of oxidation products of +-alpha-pinene, d-limonene, and isoprene in BALB/c mice. Inhal Toxicol.
2002 Jul;14 7:663-84.
Santodonato, J, S. Bosch, W. Meylan, J. Becker, and M. Neal. 1985. Final Report. Monograph on the Potential
Carcinogenic Risk to Humans: Turpentine. National Cancer Institute, Bethesda, MD.
Savolainen H, Pfaffli P 1978. Effects on longterm turpentine inhalation on rat brain protein metabolism. Chem Biol
Interact 1978;21:271-6.
Sperling F, Marcus WL, Collins C 1967. Acute toxic effects of turpentine vapor on rats and mice. Toxicol Appl
Pharmacol 1967;10:8-20.
Verschueren K 2001. Handbook of Environmental Data on Organic Chemicals. Fourth edition. John Wiley & Sons, Inc,
New York. Vol. 1, 24.
Wallace L, Buckley T, Pellizzari E, Gordon S 1996. Breath Measurements as Volatile Organic Compound Biomarkers.
Environ Health Perspect 104, Suppl 5: 861-869.
Weschler C.J. and Shields H.C. 1997. Potential reactions among indoor pollutants. Atmospheric Environment, 31:
3487-3495.
Wilkins CK, Wolkoff P, Clausen PA, Hammer M, Nielsen GD. 2003. Upper airway irritation of terpene/ozone
oxidation products TOPS. Dependence on reaction time, relative humidity and initial ozone concentration. Toxicol
Lett.2003;143 2:109-14.
Winter PM and Miller JN 1976 Carbon monoxide poisoning. J Am Med Assoc, 236: 1503.
Wolkoff P, Clausen P.A, Jensen B. and Nielsen G.D. 1997. Are we measuring the relevant indoor pollutants ? Indoor
Air, 7, 92-106.
Wolkoff P, Clausen PA, Wilkins CK, Nielsen GD 2000. Formation of strong airway irritants in terpene/ozone mixtures.
Indoor Air 10:82-91.
The INDEX project Final report 330
References in Annex 1, 2 and 3
1. Jantunen MJ, Katsouyanni K, Knöppel H, Künzli N, Lebret E, Maroni M, Saarela K, Srám R, Zmirou D. Final
Report: Air Pollution Exposure in European Cities: the EXPOLIS Study. Kuopio, Finland: National Public Health
Institute B16, 1999 Annex II: Table 18.
2. WHO (2000): Air Quality Guidelines for Europe. WHO Regional Publications, European Series, No. 91, Regional
Office for Europe, Copenhagen.
3. Directive 2000/69/EC of the European Parliament and of the Council of 16 November 2000 relating to limit values
for benzene and carbon monoxide in ambient air.
4. National Air Quality Standard in Finland. VNa ilmanlaadusta 711/2001 in (Finnish).
5. Clayton CA, Pellizzari ED, Whitmore RW, Perritt RL, Quackenboss JJ (1999). National Human Exposure
Assessment Survey (NHEXAS): distributions and associations of lead, arsenic, and volatile organic compounds in
EPA Region 5. J Expo Anal Environ Epidemiol 9:381-392.
6. COMEAP Committee on the medical effects of air pollutants (1997): Handbook on air pollution and health.
Department of health, committee on the medical effects of air pollutants.
7. Cocheo V, Sacco P, Boaretto C, de Saeger E, Ballesta P, Skov H, Goelen E, Gonzalez N, Caracena A (2000).
Urban benzene and population exposure. Nature 404:141.
8. Hoffmann K, Krause C, Seifert B, Ullrich D. The German Environmental Survey 1990/1992 (GerES II): Sources of
Personal Exposure to Volatile Organic Compounds. J. Exp. And Environ. Epid. 2000; 10: 115-125.
9. NIOSH Pocket guide to chemical hazards (http://www.cdc.gov/niosh/npg/npgdname.html). Accessed on Feb. 24,
2003.
10. EPA California, USA. OEHHA Office of Environmental Health Hazard Assessment.,
(http://www.oehha.ca.gov/air/chronic_rels/)
. Accessed on Feb. 24, 2003.
11. WHO. Guidelines for Air Quality (1999). WHO Geneva, Switzerland.
(http://www.who.int/peh/air/Airqualitygd.htm)
.
12. NKB. Testing and discussion of the proposed criteria for the ability of chemical substances to couse allergy and
hypersensitivy in the skin and the lower airways. Summary Report. Nordic Committee on Building Regulations,
NKB Committee and Work Reports 1994:03 E. Painatuskeskus helsinki, Finland.
13. Georgoulis LB, Hänninen O, Samoli E, Katsouyanni K, Kuenzli N, Polanska L, Bruinen de Bruin Y, Alm S,
Jantunen M (2002). Personal carbon monoxide exposure in five European cities and its determinants. Atmos
Environ 36:963-974.
14. IEH Indoor air quality in the home (2): carbon monoxide. Assessment A5. Leicester (UK): Institute of of
Environment and Health 1998.
15. Alm S. Personal exposures of pre-school children to carbon monoxide and nitrogen dioxide in urban air. Academic
dissertation. Publications of the National Public Health Institute A19/99. National Public Health Institute, Kuopio,
Finland; 1999.
16. Kousa A., Monn C., Rotko T., Alm S., Oglesby L. and Jantunen M. (2001): Personal exposures to NO2 in the
Expolis-study: Relation to residential indoor, outdoor and workplace concentrations in Basel, Helsinki and Prague.
Atmos Environ 35(20):3405-3412.
17. COMEAP Committee on the medical effects of air pollutants (1997): Handbook on air pollution and health.
Department of health, committee on the medical effects of air pollutants.
The INDEX project Final report 331
18. Tuomainen M, Pasanen A-L, Tuomainen A, Liesivuori J, Juvonen P (1999). Quality of indoor climate in two new
blocks of flats. In Proceedings of the 8
th
International Conference on Indoor Air Quality and Climate held in
Edinburgh, Scotland, 8-13 August 1999, Vol 4.
19. HTP-arvot 2002. Sosiaali- ja terveysministeriö. Työsuojelusäädöksiä 3, 2002, Tampere, Finland (in Finnish).
(Reports of the Ministry of Social Affairs and Health ). (http://www.ketsu.net/htp/2002.pdf)
.
20. Jantunen M, Hänninen O, Katsouyanni K, Knöppel H, Künzli N, Lebret E, Maroni M, Saarela K, Srám R, Zmirou
D. Air pollution exposure in European cities: the EXPOLIS study. J Expo Anal Environ Epidemiol 1998;8:495-
518.
21. EXPOLIS Combined International Database (CIDB).
22. Wolff T and Stirn H. 2000. Richtwerte fur die Innenraumluft: Disisocyanate. Bundesgesundheitsbl –
Gesundheitsforsch – Gesundheitsschutz. 43:505-512 (in German).
23. Wolkoff P and Nielsen GD (2001). Organic compounds in indoor air - their relevance for perceived indoor air
quality? Atmos Environ 35:4407-4417.
24. Sagunski H, Rosskamp E (2002) Richtwerte fur die Innenraumluft: Tris(2-chloroethyl)phosphat.
Bundesgesundheitsbl – Gesundheitsforsch – Gesundheitsschutz. 43:300-306 (in German).
25. The German Environmental Survey 1991/1992 (GerES IIb). (http://www.umweltbundesamt.de/survey-
e/us9092/innen.htm)
26. Seifert B, Englert N, Sagunski H, Witten J (1999). Guideline values for indoor air pollutants in Raw G, Aizlewood
C and Warren P (eds): Proceedings of the 8th International Conference on Indoor Air Quality and Climate,
Edinburgh, Scotland, 8-13 August 1999, Vol 1:499-504.
European Commission
EUR 21590 EN – DG Joint Research Centre, Institute for Health and Consumer Protection
The INDEX Project - Critical Appraisal of the Setting and Implementation of Indoor Exposure Limits in the EU
Kotzias Dimitrios, Koistinen Kimmo, Kephalopoulos Stylianos, Carrer Paolo, Maroni Marco, Schlitt Christian,
Jantunen Matti, Cochet Christian, Kirchner Severine, Lindvall Thomas, Mclaughlin James, Molhave Lars
Luxembourg: Office for Official Publications of the European Communities
2005334 pp. – 21 x 29.7 cm
EUR - Scientific and Technical Research series; ISSN 1018-5593
The mission of the JRC is to provide customer-driven scientific and technical support for the
conception, development, implementation and monitoring of EU policies. As a service of the European
Commission, the JRC functions as a reference centre of science and technology for the Union. Close
to the policy-making process, it serves the common interest of the Member States, while being
independent of special interests, whether private or national.
AA- BB- XXXXX- LL- C