26.12.2012 Views

Toxicology of Industrial Compounds

Toxicology of Industrial Compounds

Toxicology of Industrial Compounds

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>Toxicology</strong> <strong>of</strong> <strong>Industrial</strong> <strong>Compounds</strong>


<strong>Toxicology</strong> <strong>of</strong> <strong>Industrial</strong><br />

<strong>Compounds</strong><br />

Edited by<br />

HELMUT THOMAS<br />

CIBA-GEIGY Ltd, Basel, Switzerland<br />

ROBERT HESS<br />

Dornach, Switzerland<br />

and<br />

FELIX WAECHTER<br />

CIBA-GEIGY Ltd, Basel, Switzerland


This edition published in the Taylor & Francis e-Library, 2005.<br />

“To purchase your own copy <strong>of</strong> this or any <strong>of</strong> Taylor & Francis or Routledge’s<br />

collection <strong>of</strong> thousands <strong>of</strong> eBooks please go to www.eBookstore.tandf.co.uk.”<br />

UK Taylor & Francis Ltd, 4 John Street, London WC1N 2ET<br />

USA Taylor & Francis Inc., 1900 Frost Road, Suite 101, Bristol, PA 19007<br />

Copyright © Taylor & Francis Ltd 1995<br />

All rights reserved. No part <strong>of</strong> this publication may be reproduced, stored in a<br />

retrieval system, or transmitted, in any form or by any means, electronic, electro<br />

static, magnetic tape, mechanical, photocopying, recording or otherwise, without<br />

the prior permission <strong>of</strong> the copyright owner.<br />

Library <strong>of</strong> Congress Cataloguing Publication data are available<br />

Cover design by Hybert Design & Type, Maidenhead, Berks.<br />

British Library Cataloguing in Publication Data<br />

A catalogue record for this book is available from the British Library.<br />

ISBN 0-203-97962-1 Master e-book ISBN<br />

ISBN 0-7484-0239-X (Print Edition) (cloth)


Contents<br />

Preface vii<br />

List <strong>of</strong> Contributors ix<br />

PART ONE Bioavailability and metabolic aspects <strong>of</strong> industrial<br />

chemicals<br />

1. Biomonitoring and Absorption <strong>of</strong> <strong>Industrial</strong><br />

Chemicals: the Challenge <strong>of</strong> Organic Solvents<br />

F.A.de Wolff S.Kezic J.G.M.van Engelen<br />

A.C.Monster<br />

2. Toxicokinetics and Biodisposition <strong>of</strong> <strong>Industrial</strong><br />

Chemicals<br />

N.P.E.Vermeulen R.T.H.van Welie B.M.de<br />

Rooij J.N.M.Commandeur<br />

3. Metabolic Activation <strong>of</strong> <strong>Industrial</strong> Chemicals<br />

and Implications for Toxicity<br />

G.J.Mulder<br />

4. Sizing Up the Problem <strong>of</strong> Exposure<br />

Extrapolation: New Directions in Allometric<br />

Scaling<br />

D.B.Campbell<br />

PART TWO Reactive industrial chemicals 59<br />

5. Metabolism <strong>of</strong> Reactive Chemicals<br />

P.J.van Bladeren B.van Ommen<br />

60<br />

6. Methods for the Determination <strong>of</strong> Reactive<br />

<strong>Compounds</strong><br />

P.Sagelsdorff<br />

72<br />

PART THREE Pulmonary toxicology <strong>of</strong> industrial chemicals 90<br />

7. Studies to Assess the Carcinogenic Potential <strong>of</strong><br />

Man-Made Vitreous Fibers<br />

T.W.Hesterberg G.R.Chase R.A.Versen<br />

R.Anderson<br />

91<br />

1<br />

2<br />

12<br />

36<br />

44


8. Pulmonary Toxicity Studies with Man-Made<br />

Organic Fibres: Preparation and Comparisons<br />

<strong>of</strong> Size-separated Para-aramid with Chrysotile<br />

Asbestos Fibres<br />

D.B.Warheit M.A.Hartsky C.J.Butterick<br />

S.R.Frame<br />

9. Pulmonary Hyperreactivity to <strong>Industrial</strong><br />

Pollutants<br />

J.Pauluhn<br />

10. Mechanisms <strong>of</strong> Pulmonary Sensitization<br />

I.Kimber<br />

11. Occupational Asthma Induced by Chemical<br />

Agents<br />

C.A.C.Pickering<br />

PART FOUR Biomarkers and risk assessment <strong>of</strong> industrial<br />

chemicals<br />

12. Biomarkers and Risk Assessment<br />

K.Hemminki<br />

13. Extrapolation <strong>of</strong> Toxicity Data and Assessment<br />

<strong>of</strong> Risk<br />

N.Fedtke<br />

14. Molecular Approaches to Assess Cancer Risks<br />

A.S.Wright J.P.Aston N.J.van Sittert<br />

W.P.Watson<br />

15. Evaluation <strong>of</strong> Toxicity to the Immune System<br />

H.-W.Vohr<br />

16. New Strategies: the Use <strong>of</strong> Long-term Cultures<br />

<strong>of</strong> Hepatocytes in Toxicity Testing and<br />

Metabolism Studies <strong>of</strong> Chemical Products<br />

Other than Pharmaceuticals<br />

V.Rogiers M.Akrawi S.Coecke<br />

Y.Vandenberghe E.Shephard I.Phillips<br />

A.Vercruysse<br />

117<br />

129<br />

138<br />

149<br />

157<br />

158<br />

167<br />

180<br />

197<br />

207<br />

PART FIVE Mechanisms <strong>of</strong> toxicity <strong>of</strong> industrial chemicals 222<br />

17. Peroxisome Proliferation<br />

B.G.Lake R.J.Price<br />

223<br />

v


vi<br />

18. Neurotoxicity Testing <strong>of</strong> <strong>Industrial</strong><br />

<strong>Compounds</strong>: in vivo Markers and Mechanisms<br />

<strong>of</strong> Action<br />

K.J.van den Berg J.-B.P.Gramsbergen<br />

E.M.G.Hoogendijk J.H.C.M.Lammers<br />

W.S.Sloot B.M.Kulig<br />

19. Endocrine <strong>Toxicology</strong> <strong>of</strong> the Thyroid for<br />

<strong>Industrial</strong> <strong>Compounds</strong><br />

C.K.Atterwill S.P.Aylward<br />

20. Testing and Evaluation for Reproductive<br />

Toxicity<br />

A.K.Palmer<br />

238<br />

255<br />

280<br />

PART SIX Toxicity <strong>of</strong> selected classes <strong>of</strong> industrial chemicals 300<br />

21. Special Points in the Toxicity Assessment <strong>of</strong><br />

Colorants (Dyes and Pigments)<br />

H.M.Bolt<br />

301<br />

22. <strong>Toxicology</strong> <strong>of</strong> Textile Chemicals<br />

D.Sedlak<br />

309<br />

23. Antioxidants and Light Stabilisers: Toxic<br />

Effects <strong>of</strong> 3,5-Dialkyl-hydroxyphenyl Propionic<br />

Acid Derivatives in the Rat and their Relevance<br />

for Human Safety Evaluation<br />

H.Thomas P.Dollenmeier E.Persohn H.Weideli<br />

F.Waechter<br />

317<br />

24. <strong>Toxicology</strong> <strong>of</strong> Surfactants: Molecular,<br />

Mechanistic and Regulatory Aspects<br />

W.Sterzel<br />

339<br />

PART SEVEN Controversial mechanistic and regulatory issues in<br />

the safety assessment <strong>of</strong> industrial chemicals<br />

25. Low Dose <strong>of</strong> a Genotoxic Carcinogen does not<br />

‘Cause’ Cancer; it Accelerates Spontaneous<br />

Carcinogenesis<br />

W.K.Lutz<br />

26. Controversial Mechanistic and Regulatory<br />

Issues in Safety Assessment <strong>of</strong> <strong>Industrial</strong><br />

Chemicals—an Industry Point <strong>of</strong> View<br />

H.-P.Gelbke<br />

355<br />

356<br />

362<br />

Index 373


Preface<br />

A large number <strong>of</strong> chemical compounds are being constantly introduced<br />

and produced to ease and comfort modern human life. Among those, the<br />

industrial compounds represent that particular fraction <strong>of</strong> chemicals which<br />

are not intended for use in biological systems, but to which humans may be<br />

non-intentionally exposed; at the workplace, by product application or<br />

through the environment.<br />

The International Society for the Study <strong>of</strong> Xenobiotics (ISSX) committed<br />

itself to address, for the first time in the long history <strong>of</strong> industrial<br />

chemicals, the toxicology <strong>of</strong> this class <strong>of</strong> compounds in an intensive<br />

scientific workshop held June 12 through 15, 1994 in Schluchsee,<br />

Germany. This workshop was not only the first such event hosted by ISSX<br />

since its foundation in 1981, but also an extension <strong>of</strong> the society’s scope<br />

beyond its traditionally covered objective to promote studies on xenobiotic<br />

metabolism, disposition and kinetics mainly <strong>of</strong> drugs and agrochemicals.<br />

The large classes <strong>of</strong> pharmaceuticals and agrochemicals had been<br />

deliberately excluded from the scope <strong>of</strong> this workshop, since their terms <strong>of</strong><br />

use generally demand ample registrational toxicity testing that inevitably<br />

leads to a wealth <strong>of</strong> information on, and pr<strong>of</strong>ound toxicological<br />

characterisation <strong>of</strong>, these compounds.<br />

<strong>Industrial</strong> chemicals, instead, which are frequently produced in large<br />

quantities such as pigments, dye-stuffs, plastic materials and additives,<br />

detergents, solvents, etc., to name but a few, are in many cases subjected to<br />

the examination <strong>of</strong> a very basic handling safety only, and may lack any<br />

further toxicity testing. This implies that essentially nothing is known<br />

about their bioavailability, metabolism, excretion and toxicological<br />

properties—unless problems arise. And once toxicity problems come up,<br />

the question arises with them <strong>of</strong> whether or not the available and<br />

traditionally employed methodology is appropriate to approach and solve<br />

them. This, because different from the largely low molecular weight<br />

structures developed for use in biological systems, industrial chemicals are<br />

<strong>of</strong>ten characterised by rather high molecular weight and the incorporation<br />

<strong>of</strong> peculiar structural entities.


viii<br />

Therefore, it was the aim <strong>of</strong> this workshop to contribute to the<br />

investigation <strong>of</strong> industrial chemicals by focussing on the individual<br />

structure, its biological fate, its potential toxicity to mammals and the<br />

molecular mechanisms possibly underlying such adverse effects by<br />

highlighting the use and significance <strong>of</strong> experimental toxicology, with<br />

special emphasis on mechanistic aspects, in the safety assessment <strong>of</strong><br />

industrial compounds as well as to current regulatory and legal<br />

considerations. Topics had been selected to review generally approved facts<br />

and mechanisms, and to particularly address and explore areas <strong>of</strong><br />

investigative and regulatory uncertainty, thereby intending to bring<br />

together the broadly diverse expertise and interests <strong>of</strong> academic<br />

researchers, corporate scientists, experts in safety assessment and<br />

representatives from regulatory authorities.<br />

The following contributions reflect a substantial selection <strong>of</strong> the 27<br />

lectures and six short communications presented during the workshop.<br />

May they succeed in setting a landmark for the due change from the current<br />

era <strong>of</strong> black-box toxicology and largely undifferentiated regulatory<br />

treatment <strong>of</strong> industrial chemicals to the desirable toxicology and safety<br />

assessment by structure in the future.<br />

We gratefully acknowledge the substantial financial support by CIBA-<br />

GEIGY and the RCC Group as well as the financial contributions <strong>of</strong><br />

ADME Bioanalysis, BASF, Henkel, Hüls, Lonza, Schering and Union<br />

Carbide.<br />

Our gratitude is also extended to Mrs Ch.Zehnder for secretarial<br />

assistance and to Taylor & Francis for continuous support, patience and<br />

encouragement to make this publication possible.<br />

H.Thomas<br />

R.Hess<br />

F.Waechter


Contributors<br />

May Akrawi<br />

Department <strong>of</strong> Biochemistry and Molecular Biology, University College<br />

London, Gower Street, London WC1E 6BT, UK<br />

Robert Anderson<br />

Schuller MTC, Health, Safety and Environmental Department,<br />

<strong>Toxicology</strong> Group, PO Box 625005, Littleton, CO 80162–5005, USA<br />

J.Paul Aston<br />

Sittingbourne Research Centre, Sittingbourne, Kent ME9 8AG, UK<br />

Christopher K.Atterwill<br />

CellTox Centre, University <strong>of</strong> Hertfordshire, Hatfield Campus, College<br />

Lane, Hatfield AL10 9AB, UK<br />

Samuel P.Aylward<br />

CellTox Centre, University <strong>of</strong> Hertfordshire, Hatfield Campus, College<br />

Lane, Hatfield AL10 9AB, UK<br />

Peter J.van Bladeren<br />

TNO Nutrition and Food Research, PO Box 360, Utrechtseweg 48,<br />

NL-3700 AJ Zeist, The Netherlands<br />

Hermann M.Bolt<br />

Institut für Arbeitsphysiologie, Universität Dortmund, Ardeystrasse 67,<br />

D-44139 Dortmund, Germany<br />

Charles J.Butterick<br />

Texas Technical Health Sciences Centre, Lubbock, TX, USA<br />

D.Bruce Campbell<br />

Servier Research and Development, Fulmer Hall, Windmill Road,<br />

Fulmer, Slough SL3 6HH, UK<br />

Gerald R.Chase<br />

Schuller MTC, Health, Safety and Environmental Department,<br />

<strong>Toxicology</strong> Group, PO Box 625005, Littleton, CO 80162–5005, USA


x<br />

Sandra Coecke<br />

Vrije Universiteit Brussel, Department <strong>of</strong> <strong>Toxicology</strong>, Laarbeeklaan 103<br />

B-1090, Brussels, Belgium<br />

Jan N.M.Commandeur<br />

Division <strong>of</strong> Molecular <strong>Toxicology</strong>, Department <strong>of</strong> Pharmacochemistry,<br />

Vrije Universiteit van Amsterdam, De Boelelaan 1083, H1081 NL-V<br />

Amsterdam, The Netherlands<br />

Peter Dollenmeier<br />

CIBA-GEIGY Ltd., R-1002.2.62, PO Box CH-4002 Basel, Switzerland<br />

Jacqueline G.M.van Engelen<br />

Coronel Laboratory, University <strong>of</strong> Amsterdam, Academic Medical<br />

Centre, Meibergdreef 15, NL-1105 Amsterdam, The Netherlands<br />

Norbert Fedtke<br />

Hüls AG, Bau 2328/PB 12, D-45764 Marl, Germany<br />

Steven R.Frame<br />

DuPont Central Research and Development, Haskell Laboratory, PO<br />

Box 50, Elkton Road, Newark, DE 19714–0050, USA<br />

Heinz-Peter Gelbke<br />

BASF AG, Abt. Toxikologie, D-67056 Ludwigshafen, Germany<br />

Jan-Bert P.Gramsbergen<br />

Department <strong>of</strong> Public Health, Erasmus University, Rotterdam, The<br />

Netherlands<br />

Mark A.Hartsky<br />

DuPont Central Research and Development, Haskell Laboratory, PO<br />

Box 50, Elkton Road, Newark, DE 19714–0050, USA<br />

Kari Hemminki<br />

CNT, Karolinska Institute, Novum, S-141 57 Huddinge, Sweden<br />

Thomas W.Hesterberg<br />

Schuller MTC, Health, Safety and Environmental Department,<br />

<strong>Toxicology</strong> Group, PO Box 625005, Littleton, CO 80162–5005, USA<br />

Elisabeth M.G.Hoogendijk<br />

TNO <strong>Toxicology</strong>, Department <strong>of</strong> Neurotoxicology, PO Box 5815,<br />

NL-2280 HV Rijswijk, The Netherlands<br />

Sanja Keži<br />

Coronel Laboratory, University <strong>of</strong> Amsterdam, Academic Medical<br />

Centre, Meibergdreef 15, NL-1105 Amsterdam, The Netherlands<br />

Ian Kimber<br />

Zeneca Central <strong>Toxicology</strong> Laboratory, Alderley Park, Macclesfield,<br />

Cheshire SK10 4TJ, UK


Beverly M.Kulig<br />

TNO <strong>Toxicology</strong>, Department <strong>of</strong> Neurotoxicology, PO Box 5815,<br />

NL-2280 HV Rijswijk, The Netherlands<br />

Brian G.Lake<br />

BIBRA International, Woodmansterne Road, Carshalton, Surrey, SM5<br />

4DS, UK<br />

Jan H.C.M.Lammers<br />

TNO <strong>Toxicology</strong>, Department <strong>of</strong> Neurotoxicology, PO Box 5815,<br />

NL-2280 HV Rijswijk, The Netherlands<br />

Werner K.Lutz<br />

Universität Würzburg, Institut für Toxikologie, Versbacher Strasse 9,<br />

D-97078 Würzburg, Germany<br />

Aart C.Monster<br />

Coronel Laboratory, University <strong>of</strong> Amsterdam, Academic Medical<br />

Centre, Meibergdreef 15, NL-1105 Amsterdam, The Netherlands<br />

Gerard J.Mulder<br />

Center for Bio-Pharmaceutical Sciences, Sylvius Laboratories, Leiden<br />

University, PO Box 9503, NL-2300 RA Leiden, The Netherlands<br />

Ben van Ommen<br />

TNO Nutrition and Food Research, PO Box 360, Utrechtseweg 48,<br />

NL-3700 AJ Zeist, The Netherlands<br />

Anthony K.Palmer<br />

Huntingdon Research Centre Ltd., PO Box 2, Huntingdon, Cambs,<br />

PE18 6ES UK<br />

Jürgen Pauluhn<br />

BAYER AG, Department <strong>of</strong> <strong>Toxicology</strong>, Institute <strong>of</strong> <strong>Industrial</strong><br />

<strong>Toxicology</strong>, Bldg. 514, D-42096 Wuppertal, Germany<br />

Elke Persohn<br />

CIBA-GEIGY Ltd., Cell Biology Unit, R-1058.2.64, PO Box, CH-4002<br />

Basel, Switzerland<br />

Ian Phillips<br />

Department <strong>of</strong> Biochemistry, Queen Mary and Westfield College,<br />

University <strong>of</strong> London, Mile End Road, London, E1 4NS, UK<br />

C.A.C.Pickering<br />

North West Lung Centre, Wythenshawe Hospital, Southmoor Road,<br />

Manchester M23 9LT, UK<br />

Roger J.Price<br />

BIBRA International, Woodmansterne Road, Carshalton, Surrey SM5<br />

4DS, UK<br />

xi


xii<br />

Vera Rogiers<br />

Vrije Universiteit Brussel, Department <strong>of</strong> <strong>Toxicology</strong>, Laarbeeklaan<br />

103, B-1090 Brussels, Belgium<br />

Ben M.de Rooij<br />

Division <strong>of</strong> Molecular <strong>Toxicology</strong>, Department <strong>of</strong> Pharmacochemistry,<br />

Vrije Universiteit van Amsterdam, De Boelelaan 1083, H1081 NL-V<br />

Amsterdam, The Netherlands<br />

Peter Sagelsdorff<br />

CIBA-GEIGY Ltd., Cell Biology Unit, R-1058.2.52, PO Box, CH-4002<br />

Basel, Switzerland<br />

Dieter Sedlak<br />

Enviro Tex GmbH, Provinostrasse 52, D-86153 Augsburg, Germany<br />

Elizabeth Shephard<br />

Department <strong>of</strong> Biochemistry and Molecular Biology, University College<br />

London, Gower Street, London WC1E 6BT, UK<br />

Nico J.van Sittert<br />

Sittingbourne Research Centre, Sittingbourne, Kent ME9 8AG, UK<br />

Willem S.Sloot<br />

TNO <strong>Toxicology</strong>, Department <strong>of</strong> Neurotoxicology, PO Box 5815,<br />

NL-2280 HV Rijswijk, The Netherlands<br />

Walter Sterzel<br />

Henkel KGaA, TTB-Toxikologie, Geb. Z33, D-40191 Düsseldorf,<br />

Germany<br />

Helmut Thomas<br />

CIBA-GEIGY Ltd., Cell Biology Unit, R-1058.2.46, PO Box, CH-4002<br />

Basel, Switzerland. Current address: Ciba-Pharmaceuticals, Stamford<br />

Lodge, Wilmslow, Cheshire SK9 4LY, UK<br />

Kornelis J.van den Berg<br />

TNO <strong>Toxicology</strong>, Department <strong>of</strong> Neurotoxicology, PO Box 5815,<br />

NL-2280 HV Rijswijk, The Netherlands<br />

Yves Vandenberghe<br />

Vrije Universiteit Brussel, Department <strong>of</strong> <strong>Toxicology</strong>, Laarbeeklaan 103<br />

B-1090 Brussels, Belgium<br />

Antoine Vercruysse<br />

Vrije Universiteit Brussel, Department <strong>of</strong> <strong>Toxicology</strong>, Laarbeeklaan 103<br />

B-1090 Brussels, Belgium<br />

Nico P.E.Vermeulen<br />

Division <strong>of</strong> Molecular <strong>Toxicology</strong>, Department <strong>of</strong> Pharmacochemistry,<br />

Vrije Universiteit Amsterdam, De Boelelaan 1083, H1081 NL-V<br />

Amsterdam, The Netherlands


Richard A.Versen<br />

Schuller MTC, Health, Safety and Environmental Department,<br />

<strong>Toxicology</strong> Group, P.O. Box 625005, Littleton, CO 80162–5005, USA<br />

Hans-Werner Vohr<br />

Bayer AG, Fachbereich Toxikologie, Institut für Toxikologie<br />

Landwirtschaft, Friedrich-Ebert-Strasse 217, D-42096 Wuppertal,<br />

Germany<br />

Felix Waechter<br />

CIBA-GEIGY Ltd, Cell Biology Unit, R-1058.2.68, PO Box, CH-4002<br />

Basel, Switzerland<br />

David B.Wahrheit<br />

DuPont Central Research and Development, Haskell Laboratory, PO<br />

Box 50, Elkton Road, Newark, Delaware 19714–0050, USA<br />

William P.Watson<br />

Sittingbourne Research Centre, Sittingbourne, Kent ME9 8AG, UK<br />

Hansjörg Weideli<br />

CIBA-GEIGY Ltd, R-1002.2.59, PO Box, CH-4002 Basel, Switzerland<br />

Ronald T.H.van Welie<br />

Division <strong>of</strong> Molecular <strong>Toxicology</strong>, Department <strong>of</strong> Pharmacochemistry,<br />

Vrije Universiteit van Amsterdam, De Boelelaan 1083, H1081 NL-V<br />

Amsterdam, The Netherlands<br />

Frederik A.de Wolff<br />

Coronel Laboratory, University <strong>of</strong> Amsterdam, Academic Medical<br />

Centre, Meibergdreef 15, 1105 Amsterdam, The Netherlands<br />

Alan S.Wright<br />

Sittingbourne Research Centre, Sittingbourne, Kent ME9 8AG, UK<br />

xiii


PART ONE<br />

Bioavailability and metabolic aspects <strong>of</strong><br />

industrial chemicals


1<br />

Biomonitoring and Absorption <strong>of</strong> <strong>Industrial</strong><br />

Chemicals: the Challenge <strong>of</strong> Organic Solvents<br />

FREDERIK A.DE WOLFF*, SANJA KEŽI , JACQUELINE<br />

G.M.van ENGELEN and AART C.MONSTER<br />

University <strong>of</strong> Amsterdam, Academic Medical Center,<br />

Amsterdam<br />

Introduction<br />

Organic solvents form a very important group <strong>of</strong> industrial chemicals.<br />

They are widely used in a range <strong>of</strong> occupational settings and may exert a<br />

number <strong>of</strong> deleterious effects when subjects are acutely or chronically<br />

exposed. Among the acute effects are skin and mucosal irritation and<br />

general anaesthesia produced by most solvents at high air concentrations.<br />

Examples <strong>of</strong> chronic effects are peripheral neuropathy after long-term<br />

exposure to n-hexane or carbon disulphide, and the organo-psychosyndrome<br />

or ‘solvent dementia’ which may occur after chronic<br />

occupational exposure to a variety <strong>of</strong> volatile organic compounds.<br />

In order to prevent workers from developing solvent-induced<br />

occupational disease, it is essential to set standards for the duration and the<br />

level <strong>of</strong> external exposure. For a scientifically based standard, a clear<br />

understanding is required <strong>of</strong> the relationship between external exposure,<br />

the uptake by the body, the metabolic fate and the internal dose <strong>of</strong> the<br />

substance. The purpose <strong>of</strong> this contribution is to demonstrate the value <strong>of</strong><br />

biokinetic studies in humans to provide a sound scientific basis for<br />

regulatory decisions on occupational standards.<br />

Biological monitoring<br />

In occupational health practice, monitoring is a tool to protect workers<br />

from developing chemically-induced disease. Monitoring in preventive<br />

health care is described as ‘a systemic continuous or repetitive healthrelated<br />

activity, designed to lead if necessary to corrective action’. In<br />

occupational health, a complete monitoring programme consists <strong>of</strong> four<br />

parts: environmental, biological and biological effect monitoring, and<br />

* Also: University Hospital <strong>of</strong> Leiden, Leiden. The Netherlands


F.A.DE WOLFF ET AL. 3<br />

health surveillance. The latter is a major task for the occupational health<br />

physician, but biological monitoring and biological effect monitoring are<br />

fields <strong>of</strong> interest to the occupational toxicologist. In this contribution, only<br />

biological monitoring will be expounded upon.<br />

Biological monitoring (BM) is defined as the ‘measurement and<br />

assessment <strong>of</strong> workplace agents or their metabolites either in tissues,<br />

secreta, excreta or any combination <strong>of</strong> these to evaluate exposure and<br />

health risk compared to an appropriate reference’ (Zielhuis & Henderson,<br />

1986). This means that a biological monitoring programme is not limited<br />

to the assay <strong>of</strong> xenobiotics in biological samples. As in clinical laboratory<br />

medicine, the pre-analytical phase <strong>of</strong> the process is very important, and<br />

even more so the post-analytical phase <strong>of</strong> the laboratory analysis, which<br />

means the interpretation <strong>of</strong> the analytical data in biomedical terms. The<br />

ultimate goal <strong>of</strong> biological monitoring is the evaluation <strong>of</strong> the health risk <strong>of</strong><br />

workers by estimation <strong>of</strong> the internal dose <strong>of</strong> a chemical. This is not limited<br />

to measurement <strong>of</strong> the quantity <strong>of</strong> the substance absorbed by the body, but<br />

may also include the assay <strong>of</strong> metabolites <strong>of</strong> toxicological interest, if<br />

possible in or near a critical organ (Monster & van Hemmen, 1988).<br />

This implies that the absorption, metabolism and elimination <strong>of</strong> a<br />

substance in man should be known before a biological monitoring<br />

programme can be performed in practice. Animal experiments are <strong>of</strong><br />

limited value; volunteer studies in order to determine pulmonary and<br />

dermal uptake <strong>of</strong> organic solvents provide more relevant data for this<br />

purpose.<br />

Owing to the existence <strong>of</strong> very sensitive analytical methods it is possible<br />

to study the kinetics and metabolism <strong>of</strong> solvents in volunteers who are<br />

experimentally exposed to levels at or far below the <strong>of</strong>ficial threshold limit<br />

values, so that any health risk for the volunteers can almost totally be<br />

excluded.<br />

As with biological monitoring <strong>of</strong> most other substances, in the case <strong>of</strong><br />

organic solvents the compound itself and/or its metabolite in blood or urine<br />

can be measured. Studies with volatile, rather lipophilic, substances have an<br />

additional advantage, namely that the solvent can also be measured in<br />

expired air. Analytically this has the advantage <strong>of</strong> an extremely clean<br />

matrix in comparison with body fluids, whereas biologically, air samples<br />

provide us with information on the blood concentration <strong>of</strong> a volatile<br />

compound. Moreover, collection <strong>of</strong> expired air is non-invasive and large<br />

volumes are readily available (Droz & Guillemin, 1986).<br />

An example <strong>of</strong> a study on solvents in volunteers is the one carried out in<br />

our laboratory on the biokinetics <strong>of</strong> n-hexane and its neurotoxic metabolite<br />

2,5-hexanedione (Van Engelen et al., in preparation). Volunteers are<br />

exposed during 15 min to 60 ppm hexane by inhalation. The minute volume<br />

and the respiratory rate are measured and blood and exhaled air sampled<br />

frequently for determination <strong>of</strong> 2,5-hexanedione and n-hexane,


4 BIOMONITORING AND ABSORPTION OF INDUSTRIAL CHEMICALS<br />

respectively. Each volunteer is exposed twice in succession on one test day<br />

in order to get an impression <strong>of</strong> the within-day intra-individual variation.<br />

Venous blood is sampled through a catheter, and alveolar air is collected<br />

after holding breath for 30 s (to achieve equilibrium between pulmonary<br />

blood and air) by exhaling through a glass tube which is stoppered<br />

immediately. These tubes contain 70 ml alveolar air and the total volume is<br />

analyzed for n-hexane by using a purge-and-trap system. 2,5-Hexanedione<br />

in serum is measured by using electron capture detection after<br />

derivatization, with a detection limit <strong>of</strong> 30 micro-mol l −1 (Keži and<br />

Monster, 1991). During exposure the concentration <strong>of</strong> n-hexane in<br />

alveolar air increases very rapidly and decreases after discontinuation <strong>of</strong><br />

exposure. The half-life time <strong>of</strong> exhalatory elimination after the distribution<br />

phase is in the order <strong>of</strong> 30 min.<br />

2,5-Hexanedione becomes detectable in blood as fast as 2–3 min after<br />

commencement <strong>of</strong> n-hexane exposure. After discontinuation <strong>of</strong> dosing the<br />

metabolite concentration continues to increase for another 3 min, to<br />

disappear from the plasma with a half-life <strong>of</strong> approximately 1.5 h. The<br />

second exposure period on the same day shows very reproducible n-hexane<br />

and 2,5-hexanedione curves in the same individual. Between individuals<br />

there is considerable variation in kinetics and metabolism, and this issue is<br />

being studied in detail at present.<br />

Before a biological monitoring programme can be designed, a detailed<br />

biokinetic study like this one, <strong>of</strong> every solvent being used in industry, has to<br />

be performed. Without kinetic data it is impossible to choose for instance<br />

the correct matrix, the compound to be measured, or the sampling<br />

frequency and time. In addition, these data are necessary to establish a<br />

relationship between ambient air concentrations <strong>of</strong> a chemical (external<br />

exposure), and the biological parameters used to estimate a health risk.<br />

Absorption<br />

The primary association <strong>of</strong> the pharmacologist or general toxicologist,<br />

when reading or hearing the term ‘absorption’, is with ‘intestinal’. For<br />

drugs, gastrointestinal uptake is indeed the most common route to enter<br />

the body. In case <strong>of</strong> occupational exposure, however, intestinal absorption<br />

is <strong>of</strong> minor importance. The occupational toxicologist is, therefore, more<br />

inclined to pay attention to entry routes other than the intestine, the most<br />

important being pulmonary and dermal uptake.<br />

Pulmonary uptake<br />

There are a number <strong>of</strong> parameters which affect the pulmonary uptake <strong>of</strong><br />

organic solvents. In the first place, the physical chemistry <strong>of</strong> the compound<br />

is <strong>of</strong> importance. Both the blood-to-gas and the tissue-to-blood partition


F.A.DE WOLFF ET AL. 5<br />

Figure 1.1 The mean minute volume (1 min −1 ) and the percentage <strong>of</strong> the minute<br />

volume cleared from solvent (shaded area) during exposure to styrene (left) and 1,1,<br />

1-trichloroethane (right) at increasing degree <strong>of</strong> workload.<br />

coefficients determine the absorption through the alveolar membrane and<br />

the distribution over the body. Furthermore, exercise is an important<br />

physiological determinant. With increasing exercise, ventilation increases<br />

and, therefore, also the availability <strong>of</strong> the vapour to the lung per unit <strong>of</strong><br />

time. In addition, cardiac output increases during exercise, and this may<br />

affect absorption, distribution and metabolism through enhanced blood<br />

flow.<br />

Finally, the elimination <strong>of</strong> a solvent which occurs during exposure may<br />

significantly affect the uptake rate. The percentage <strong>of</strong> the vapour not<br />

retained by the body but exhaled again is dependent on, again,<br />

physicochemical factors such as solubility, but also on the rate <strong>of</strong><br />

metabolism (Fiserova-Bergerova, 1985).<br />

In order to demonstrate the different factors which may affect<br />

pulmonary absorption <strong>of</strong> vapours we have constructed Figure 1.1, based<br />

on earlier work <strong>of</strong> Astrand et al. (Astrand, 1975). In their studies,<br />

volunteers were exposed to different vapours such as styrene or 1,1,1trichloroethane<br />

at increasing degrees <strong>of</strong> workload during 2 h.<br />

The first 30 min they were exposed at rest, and then the workload was<br />

increased every 30 min with 50 W. The minute volume, here referred to as<br />

‘supply’, was measured and expressed in 1 min −1 , and the exhaled solvent<br />

concentration was also measured at regular intervals. The shaded area <strong>of</strong><br />

the vertical bars in Figure 1.1 indicate the percentage <strong>of</strong> minute volume<br />

cleared from the solvent, averaged over the observation period. This is<br />

considered to be a measure for pulmonary uptake.


6 BIOMONITORING AND ABSORPTION OF INDUSTRIAL CHEMICALS<br />

During continuous exposure to a constant concentration and at<br />

increasing exercise the uptake <strong>of</strong> styrene remains constant, expressed in<br />

terms <strong>of</strong> percentage <strong>of</strong> the minute volume cleared. Apparently, the body is<br />

not easily saturated with styrene. The picture for 1,1,1-trichloroethane is<br />

completely different. Although the minute volume at each level <strong>of</strong><br />

workload is comparable with that <strong>of</strong> the styrene experiment, it is clear that<br />

the retention <strong>of</strong> 1,1,1-trichloro-ethane is much lower. Apparently, the body<br />

becomes rapidly saturated with 1,1,1-trichloroethane. The reasons for the<br />

difference in pulmonary uptake between these two solvents are evident.<br />

Styrene is highly soluble in blood and it is extensively metabolized to<br />

mandelic acid and phenyl glyoxylic acid. The retention in the body remains<br />

the same, and therefore the uptake increases proportionally with the<br />

minute volume.<br />

In contrast to styrene, 1,1,1-trichloroethane has only a limited solubility<br />

in blood, and it is hardly metabolized. This means that during exposure the<br />

body becomes rapidly saturated with the substance, and that an increase in<br />

minute volume by increasing workload results in a lower retention, and<br />

hardly in higher uptake. Differences in kinetic behaviour, as demonstrated<br />

for styrene and 1,1,1-trichloroethane, are important for the design <strong>of</strong> a<br />

biological monitoring programme.<br />

Dermal uptake<br />

Absorption <strong>of</strong> solvents through the skin may be affected by a number <strong>of</strong><br />

factors. Many organic solvents are able to penetrate the skin and thus enter<br />

the body. This is a rather well-known fact which can be prevented in<br />

industrial practice by use <strong>of</strong> protective clothing. It is, however, less<br />

common knowledge that solvents in the vapour phase may also penetrate<br />

the skin. In case <strong>of</strong> skin exposure to liquids usually a small surface is<br />

exposed, whereas in case <strong>of</strong> vapour the whole body surface <strong>of</strong> about 2 m 2<br />

may be exposed. This means that under certain conditions skin absorption<br />

<strong>of</strong> vapour may significantly contribute to the amount absorbed by<br />

inhalation.<br />

Other parameters which may affect skin absorption are the temperature,<br />

and the ability <strong>of</strong> some solvents to increase their own absorption by<br />

causing skin hyperaemia through irritation. To demonstrate these factors,<br />

some preliminary results are shown <strong>of</strong> a volunteer study on skin<br />

penetration <strong>of</strong> solvents in the liquid and vapour phases (Keži et al., in<br />

preparation).<br />

The experimental conditions are as follows. The volunteer is seated in a<br />

clear-air cabin in order to avoid additional inhalatory exposure to vapour<br />

in the experimental room. The arm is the only part <strong>of</strong> the body outside the<br />

cabin. In case <strong>of</strong> exposure to liquid on the skin, the solvent is put in a<br />

chamber which is pressed on to the skin during the exposure period, which


F.A.DE WOLFF ET AL. 7<br />

is usually no longer than a few minutes. The exposed area is usually in the<br />

order <strong>of</strong> 20 cm 2 .<br />

In the case <strong>of</strong> dermal exposure to vapour, the volunteer places the lower<br />

arm into a piece <strong>of</strong> drainage pipe through which the vapour is led with<br />

controlled flow and concentration in air. Uptake <strong>of</strong> liquid or vapour is<br />

measured in both cases by determination <strong>of</strong> the solvent in expired air, by<br />

the sampling method described earlier.<br />

Figure 1.2 shows the dermal uptake and elimination <strong>of</strong> two different<br />

liquids in one volunteer. A surface <strong>of</strong> 27 cm 2 was exposed during 3 min to<br />

pure 1,1.1-trichloroethane and to tetrachloroethene. It is clear that 1,1,1trichloroethane<br />

is absorbed through the skin much faster and to a much<br />

greater extent than tetrachloroethene, at least in exposure to the liquids.<br />

However, when the skin is exposed to the same solvents in the vapour<br />

phase the picture becomes totally different. Here the lower arm, which has<br />

a surface <strong>of</strong> about 500 cm 2 , was exposed during 15 min to solvent<br />

concentrations <strong>of</strong> approximately 500 µmol 1 −1 air (Figure 1.3).<br />

In the case <strong>of</strong> vapour exposure no difference in absorption kinetics is<br />

observed, and only a small difference in expired air concentration is seen.<br />

The reason for the discrepancy between vapour exposure is that 1,1,1trichloro-ethane<br />

causes skin irritation as the liquid, but not in the vapour<br />

phase. Irritation leads to hyperaemia and, hence, increased absorption.<br />

As it is known that dermal exposure to vapour may lead to detectable<br />

absorption, the contribution <strong>of</strong> vapour uptake <strong>of</strong> the skin in comparison to<br />

inhalatory absorption should be evaluated. This was done with<br />

trichloroethene as an example (Figure 1.4). Both curves were obtained in<br />

the same volunteer. The dermal exposure was performed first, followed by<br />

the inhalatory test after a wash out period <strong>of</strong> 2 weeks. The exposure period<br />

was 15 min, and the inhalatory concentration was 4.1 µmol l −1 . Dermal<br />

exposure <strong>of</strong> the lower arm took place at 1.4 mmol l −1 .<br />

It appears that uptake from the lungs occurs much faster than via the<br />

skin. This is conceivable because the stratum corneum is a stronger barrier<br />

than the alveolar epithelium, and causes a shift to the right <strong>of</strong> the t max. It<br />

can also be seen that inhalatory exposure leads to a much higher expired<br />

air concentration than dermal exposure. But in this respect we should<br />

realize that only a small part <strong>of</strong> the skin was exposed, namely about 500<br />

cm 2 . In fact the result should be extrapolated to the total surface <strong>of</strong> the<br />

human skin, which is about 2 m 2 . These results indicate that dermal<br />

exposure to solvent vapour should not be neglected when the safety <strong>of</strong> the<br />

industrial environment is evaluated. This is <strong>of</strong> special importance when<br />

ambient air concentrations are high, and workers are protected with<br />

protective masks but not with gloves. Another example in which skin<br />

absorption may be high in comparison with inhalation are those solvents<br />

which are readily absorbed by the skin, such as 2-butoxyethanol (Johanson<br />

and Boman, 1991).


8 BIOMONITORING AND ABSORPTION OF INDUSTRIAL CHEMICALS<br />

Figure 1.2 Elimination <strong>of</strong> 1,1,1-trichloroethane and tetrachloroethene by expired<br />

air after dermal exposure to the liquid <strong>of</strong> 27 cm 2 fore-arm skin during 3 min. 1,1,1trichloroethane<br />

liquid irritates the skin.<br />

Figure 1.3 Elimination <strong>of</strong> 1,1,1-trichloroethane and tetrachloroethene by expired<br />

air after dermal exposure to the vapour <strong>of</strong> 500 cm 2 lower-arm skin during 15 min<br />

to 500 µmol l −1 air.<br />

The temperature <strong>of</strong> the solvent is another factor that may have an<br />

influence on uptake through the skin. Figure 1.5 shows the results <strong>of</strong><br />

dermal exposure to liquid tetrachloroethene and n-hexane at two different<br />

temperatures in one volunteer. Exposure time here was only 1 min, and<br />

absorption and elimination were measured by analysis <strong>of</strong> the vapours in<br />

expired air.


At the low temperature <strong>of</strong> the liquid (15°C), the uptake <strong>of</strong><br />

tetrachloroethene is negligible when compared with a normal skin<br />

temperature <strong>of</strong> 33°C. In case <strong>of</strong> n-hexane, under comparable circumstances<br />

and in the same volunteer, the effect <strong>of</strong> temperature is much less<br />

pronounced. Apparently, the physicochemical properties <strong>of</strong> the solvent are<br />

an additional determining factor. The mechanism on which the difference<br />

between tetrachloroethene and n-hexane is based is the subject <strong>of</strong> further<br />

study.<br />

Conclusions<br />

F.A.DE WOLFF ET AL. 9<br />

Figure 1.4 Elimination <strong>of</strong> trichloroethene by expired air during and after inhalatory<br />

exposure to 4.1 µmol l −1 trichloroethene during 15 min, and after dermal vapour<br />

exposure during 15 min <strong>of</strong> the lower-arm skin (500 cm 2 to 1.4 mmol l −l air).<br />

In occupational health practice, the major absorption routes for organic<br />

solvents are not ingestion, but inhalation and skin penetration, the latter<br />

both as liquid and as vapour. The physical chemistry <strong>of</strong> the compound,<br />

exercise, and the elimination rate may affect pulmonary uptake. Factors<br />

affecting dermal uptake are the ability <strong>of</strong> the solvent to penetrate the skin as<br />

liquid or vapour, the temperature <strong>of</strong> the liquid, and the irritability <strong>of</strong> the<br />

chemical to the skin.<br />

Before a biological monitoring programme for solvent exposure can be<br />

set up, the kinetics and metabolism <strong>of</strong> the various solvents in man should<br />

be known. Owing to the availability <strong>of</strong> sensitive analytical methods it is<br />

usually possible to perform volunteer studies at safe exposure levels.<br />

Measurement <strong>of</strong> solvents in expired air and <strong>of</strong> their metabolites in body<br />

fluids is <strong>of</strong> the utmost importance to estimate the internal dose <strong>of</strong> the<br />

solvents and health risk to which man can be exposed in the work and<br />

general environment.


10 BIOMONITORING AND ABSORPTION OF INDUSTRIAL CHEMICALS<br />

Figure 1.5 Elimination <strong>of</strong> tetrachloroethene and n-hexane by expired air after<br />

dermal exposure during 1 min to liquid at 15°C and 33°C<br />

References<br />

ǺSTRAND, I., 1975, Uptake <strong>of</strong> solvents in the blood and tissues in man. A review,<br />

Scand J Work Environ Health, 1, 199–218.<br />

DROZ, P.O. and GUILLEMIN, M.P., 1986, Occupational exposure monitoring<br />

using breath analysis, J Occup Med, 28, 593–602.


F.A.DE WOLFF ET AL. 11<br />

FISEROVA-BERGEROVA, V., 1985, Toxicokinetics <strong>of</strong> organic solvents, Scand J<br />

Work Environ Health, 11, suppl. 1, 7–21.<br />

JOHANSON, G. and BOMAN, A., 1991, Percutaneous absorption <strong>of</strong> 2butoxyethanol<br />

vapour in human subjects, Br J Ind Med, 48, 788–92.<br />

KEŽI , S. and MONSTER, A.C., 1991, Determination <strong>of</strong> 2,5-hexanedione in urine<br />

and serum by gaschromatography after derivatization with O-<br />

(pentafluorobenzyl)-hydroxylamine and solid-phase extraction, J Chromatogr,<br />

563, 199–204.<br />

MONSTER, A.C. and VAN HEMMEN, J.J., 1988, Screening models in<br />

occupational health practice <strong>of</strong> assessment <strong>of</strong> individual exposure and health<br />

risk by means <strong>of</strong> biological monitoring in exposure to solvents, In Notten,<br />

W.R.F., Herber, R.F. M., Hunter, W.J. et al. (Eds) Health Surveillance <strong>of</strong><br />

lndividual Workers Exposed to Chemical Agents, pp. 47–53, Berlin: Springer.<br />

ZIELHUIS, R.L. and HENDERSON, P.Th., 1986, Definitions <strong>of</strong> monitoring<br />

activities and their relevance for the practice <strong>of</strong> occupational health, Int Arch<br />

Occup Environ Health, 57, 249–57.


2<br />

Toxicokinetics and Biodisposition <strong>of</strong> <strong>Industrial</strong><br />

Chemicals<br />

NICO P.E.VERMEULEN, RONALD T.H.van WELIE, BEN<br />

M.de ROOIJ and JAN N.M.COMMANDEUR<br />

Vrije Universiteit, Amsterdam<br />

Introduction<br />

In our industrialized world with increasing numbers <strong>of</strong> body foreign<br />

chemicals (xenobiotics) including drugs, food additives, pesticides,<br />

industrial chemicals and environmental pollutants, public concern about<br />

possible adverse (health) effects is growing. In 1989, for example, actual<br />

environmental topics in the Netherlands were photochemical summersmog<br />

and the presence <strong>of</strong> dioxines in milk <strong>of</strong> cows feeding in the neighbourhood<br />

<strong>of</strong> household refuse combustion furnaces and cable stills (CCRX, 1989). In<br />

this regard, most attention is paid to exposure to potentially mutagenic and<br />

carcinogenic xenobiotic chemicals. Apart from environmental exposure,<br />

especially at the workplace man may be exposed to elevated levels <strong>of</strong><br />

mixtures <strong>of</strong> known or unknown chemicals. Two centuries ago, cancer <strong>of</strong> the<br />

scrotum and testicles in chimney-sweepers was the first recognized<br />

occupational cancer (Pott, 1795). Since then numerous other hazardous<br />

occupational activities have been traced (Farmer et al., 1987).<br />

Nowadays, toxicologists are more and more focussed on the in vivo and<br />

in vitro bioactivation and bioinactivation mechanisms <strong>of</strong> chemicals. In the<br />

development <strong>of</strong> toxicity different stages are generally being distinguished:<br />

(1) toxicokinetics (absorption, distribution and elimination), (2)<br />

biotransformation, resulting in activation or inactivation <strong>of</strong> the chemicals,<br />

(3) reversible or irreversible interactions with cellular or tissue<br />

components, (4) protection and repair mechanisms and (5) nature and<br />

extent <strong>of</strong> the toxic effect for the organism (Vermeulen et al., 1990).<br />

Knowledge <strong>of</strong> for example species, dose, route <strong>of</strong> absorption, time <strong>of</strong><br />

exposure, tissue and organ selective interactions with (critical) cellular<br />

macro-molecules contributes to the understanding <strong>of</strong> molecular mechanisms<br />

<strong>of</strong> toxicity. Molecular mechanisms are useful in the prediction and<br />

prevention <strong>of</strong> chemically induced toxicities and they may play an<br />

important role in for example risk assessment and in the development <strong>of</strong><br />

safer chemicals (Vermeulen et al., 1990).


In this chapter, first the basic toxicokinetic concepts concerning the dis<br />

tribution, elimination and biotransformation <strong>of</strong> xenobiotics will be<br />

summarized. Subsequently, the relevance <strong>of</strong> these concepts will be<br />

illustrated and evaluated with the aid <strong>of</strong> a number <strong>of</strong> toxicokinetic studies<br />

in animals and humans concerning the nematocide 1,3-dichloropropene,<br />

the fungicide etridiazol, the chemical monomer 1,3-butadiene and the<br />

industrial solvent, 1,1,2-tri-chloroethylene. Apart from interspecies<br />

differences in the toxicokinetics, special attention will be given to<br />

interindividual differences in the toxicokinetics, among other things, as a<br />

result <strong>of</strong> genetically determined deficiencies in biotransformation enzymes<br />

as well as to its importance for the risk assessment <strong>of</strong> human exposure to<br />

industrial chemicals.<br />

Disposition <strong>of</strong> xenobiotics<br />

N.P.E.VERMEULEN ET AL. 13<br />

The overall fate <strong>of</strong> xenobiotics in an organism is determined by various<br />

toxicokinetic processes notably the route <strong>of</strong> administration, absorption,<br />

distribution and elimination. Chemicals may enter the body via various<br />

routes. Main routes are the lung, skin and gastrointestinal tract. The<br />

intraperitoneal, intramuscular, intravenous and subcutaneous routes are<br />

largely confined to experimental toxicological and therapeutic agents.<br />

Following absorption, xenobiotics enter the systemic or portal blood<br />

circulation. Distribution <strong>of</strong> chemicals in blood, organs and tissues usually<br />

occurs rapidly. The final plasma concentration depends on the ability <strong>of</strong><br />

the chemicals to pass cell membranes and on their affinity to various<br />

macromolecular proteins and tissues. Distribution to the kidney may result<br />

in direct excretion <strong>of</strong> the unchanged parent chemical. The physicochemical<br />

characteristics, such as lipophilicity and binding to plasma proteins, play an<br />

important role in the ultimate fate <strong>of</strong> a chemical in the body. The<br />

disposition <strong>of</strong> xenobiotics in the body is shown schematically in Figure 2.1.<br />

Its schematic relationship with biological/ toxicological effects is shown in<br />

Figure 2.2.<br />

Biotransformation plays an important role in the disposition <strong>of</strong><br />

xenobiotics in vivo. The liver is quantitatively the most important organ in<br />

the process <strong>of</strong> biotransformation. It receives a relative high bloodflow<br />

directly from the gastrointestinal tract via the portal vein, sometimes giving<br />

rise to the so-called hepatic ‘first-pass effect’ due to the presence <strong>of</strong> high<br />

concentrations <strong>of</strong> phase I and phase II metabolizing enzymes.<br />

Other important organs in biotransformation are the lungs, kidneys and<br />

the intestine. The primary object <strong>of</strong> biotransformation generally is to<br />

increase the hydrophilicity <strong>of</strong> chemicals, thus facilitating excretion by the<br />

kidneys in the urine or by the liver in the bile. Phase I reactions involve<br />

oxidation, reduction and hydrolysis reactions and phase II reactions<br />

conjugation or synthetic reactions. Phase I metabolic reactions generally


14 TOXICOKINETICS AND BIODISPOSITION OF INDUSTRIAL CHEMICALS<br />

Figure 2.1 Schematic representation <strong>of</strong> the fate <strong>of</strong> xenobiotics in the body according<br />

to their physico-chemical properties. Phase I and phase II represent the<br />

biotransformation processes. Adapted from Ariens and Simonis (1980).<br />

convert xenobiotic chemicals to more hydrophilic derivatives by<br />

introducing functional groups such as hydroxyl, sulphydryl and amino- or<br />

carboxylic acid groups. Phase II reactions are conjugation reactions in<br />

which the parent compounds or phase I derived metabolites are covalently<br />

bound to for example glucuronic acid, sulphate or glutathione.<br />

The group <strong>of</strong> cytochrome P-450 isoenzymes is the most important enzyme<br />

system in the catalysis <strong>of</strong> phase I reactions. The microsomal cytochrome<br />

P-450 system consists <strong>of</strong> various cytochrome P-450 isoenzymes and<br />

NADPH-cytochrome P450 reductase. It is involved in different metabolic<br />

reactions. At least three main types <strong>of</strong> activities can be distinguished,<br />

namely monooxygenase activity, oxidase activity and reductive activity<br />

(Guengerich 1994; Koymans et al., 1993). Glucuronic acid conjugation,<br />

catalyzed by UDP-glucuronyltransferases, represents one <strong>of</strong> the major<br />

phase II conjugation reactions in the conversion <strong>of</strong> exogenous and


endogenous chemicals. In mammals, another important conjugation<br />

reaction <strong>of</strong> hydroxyl groups is sulfatation, catalyzed by sulfotransferases<br />

(Sipes and Gandolfi, 1986). The group <strong>of</strong> glutathione S-transferase (GST)<br />

isoenzymes also represents an important phase II enzyme system. GST<br />

isoenzymes consist <strong>of</strong> two subunits on which the nomenclature is based<br />

(Warholm et al., 1986). The most important activity <strong>of</strong> GSTs is the<br />

catalysis <strong>of</strong> the conjugation <strong>of</strong> electrophilic, hydrophobic chemicals with<br />

the tripeptide glutathione (GSH). In general, GSH conjugation ultimately<br />

leads to the urinary excretion <strong>of</strong> mercapturic acids (N-acetyl-L-cysteine Sconjugates)<br />

(Vermeulen, 1989; Van Welie et al., 1992).<br />

Toxicokinetic principles<br />

General principles<br />

N.P.E.VERMEULEN ET AL. 15<br />

Figure 2.2 Disposition and biological effects <strong>of</strong> xenobiotics subdivided into three<br />

phases.<br />

The time course for the absorption, distribution, metabolism and<br />

elimination <strong>of</strong> a toxic substance is the subject <strong>of</strong> toxicokinetics. Implicit in<br />

any toxicokinetic description is the assumption that the response <strong>of</strong> target<br />

tissues or organs can be related to concentration pr<strong>of</strong>iles <strong>of</strong> the active form<br />

<strong>of</strong> the substance in that tissue or organ. Furthermore, it is <strong>of</strong>ten assumed<br />

that blood or plasma concentrations in one way or the other will reflect<br />

target tissue or organ concentrations and by inference the toxic effects.<br />

Under normal conditions one is generally dealing with first-order or linear<br />

kinetics, meaning that the amount <strong>of</strong> compound absorbed or eliminated<br />

(dQ) per unit <strong>of</strong> time (dt) is proportional to the total amount <strong>of</strong> compound<br />

present in the body. Zeroorder or non-linear kinetics may be valid as a<br />

consequence <strong>of</strong> various causes, e.g. saturation <strong>of</strong> binding <strong>of</strong> the toxic<br />

substance to plasma proteins or tissue components, or, more frequently


16 TOXICOKINETICS AND BIODISPOSITION OF INDUSTRIAL CHEMICALS<br />

Table 2.1 Frequently used toxicokinetic parameters and their formulas<br />

occurring, saturation <strong>of</strong> biotransformation enzyme systems. For the<br />

(mathematical) description <strong>of</strong> the toxicokinetics <strong>of</strong> substances, there exist<br />

at least two approaches at the moment: the traditional compartment<br />

pharmaco(toxico-)kinetic approach, in which the body is divided into one<br />

or more compartments, which do not necessarily correspond to<br />

physiological or anatomical units, and the physiologically-based pharmaco<br />

(toxico-)kinetic approach (PBPK or PBTK), in which organs, tissues and<br />

blood flow are taken into consideration. In Table 2.1 a summary <strong>of</strong> the most<br />

important and most frequently used traditional toxicokinetic parameters is<br />

shown. The value <strong>of</strong> some <strong>of</strong> these parameters is illustrated below, with the<br />

examples <strong>of</strong> 1,3-dichloropropene and etridiazol. The PBPK/PBTK approach<br />

is illustrated with the example <strong>of</strong> 1,3-butadiene.<br />

Principles <strong>of</strong> urinary excretion<br />

Of special interest in relation to this contribution also is the urinary<br />

excretion <strong>of</strong> xenobiotics and their metabolites by the kidneys. Two basic


N.P.E.VERMEULEN ET AL. 17<br />

processes, namely glomerular filtration and tubular secretion are used by<br />

the kidneys to remove chemicals from the bloodstream into the urine<br />

(Hook and Hewitt 1986). The kidneys are highly vulnerable to potential<br />

toxicants not only because they receive a high bloodflow (25% <strong>of</strong> the<br />

cardiac output), but also because they have the intrinsic ability to<br />

concentrate compounds. Recently, it has also become clear that xenobiotics<br />

may become nephrotoxic in the kidney itself due to bioactivation processes<br />

in combination with insufficient protection mechanisms (Commandeur and<br />

Vermeulen, 1991).<br />

The elimination <strong>of</strong> chemicals by the kidney is generally governed by firstorder<br />

processes. During first-order excretion kinetics the urinary<br />

elimination rate <strong>of</strong> a chemical is directly proportional to the plasma<br />

concentration. This means that the higher the plasma concentration the<br />

more <strong>of</strong> the chemical will be excreted in urine per unit <strong>of</strong> time. The urinary<br />

elimination rate (dQ/dt) can be calculated from a semi-logarithmic plot <strong>of</strong><br />

the urinary elimination rate versus the time <strong>of</strong> the intermittently collected<br />

urine samples (dQ/dt (mg h −l )=volume (1)×concentration (mg 1 −1 )/time (h))<br />

(Figure 2.3A).<br />

From the slope <strong>of</strong> the semi-logarithmic plasma concentration or urinary<br />

excretion rate versus time curve, the elimination rate constant (k el) and the<br />

urinary half-life <strong>of</strong> elimination (t 1/2) can be calculated. The half-life <strong>of</strong><br />

elimination is the time required to decrease the plasma concentration or the<br />

urinary elimination rate by one-half. The volume <strong>of</strong> distribution <strong>of</strong> the<br />

chemical normally can not be calculated from the urinary excretion data.<br />

Because the amount <strong>of</strong> chemical excreted in urine per unit <strong>of</strong> time (dQ/dt)<br />

is proportional to the plasma concentration (C p), the t 1/2 derived from the<br />

urinary elimination rate constant is identical to the t 1/2 <strong>of</strong> the chemical in<br />

plasma. It is evident that under these conditions the urinary excretion rate<br />

curve has the same shape as the plasma concentration curve (Figure 2.3B).<br />

In practice, the concentration <strong>of</strong> a chemical in urine (mg l −1 ) can be<br />

determined and multiplied by the volume (1) <strong>of</strong> the urine sample in order<br />

to calcu late the amount (mg) <strong>of</strong> chemical excreted over a period <strong>of</strong> time. In<br />

a semi-logarithmic plot the amount <strong>of</strong> chemical excreted is plotted against<br />

the midpoint <strong>of</strong> the interval <strong>of</strong> collection (Figure 2.3B). The accuracy <strong>of</strong> the<br />

method strongly depends on the way and the number <strong>of</strong> urine samples<br />

collected. As a rule <strong>of</strong> thumb, urine samples have to be collected during at<br />

least four half-lives <strong>of</strong> elimination. The complete cumulative urinary<br />

excretion <strong>of</strong> a chemical can be calculated as the area under the urinary<br />

excretion rate versus time curve including extrapolation time to infinity.<br />

Occupational exposure to chemicals frequently occurs 5 days a week, 8 h<br />

a day, with an exposure free period <strong>of</strong> 16 h. Intermittent exposure to a<br />

chemical may lead to different accumulation situations in the body<br />

depending on the periods between exposure in relation to t 1/2 (Table 2.1).<br />

No accumulation will occur when the intervals between the exposure


18 TOXICOKINETICS AND BIODISPOSITION OF INDUSTRIAL CHEMICALS<br />

Figure 2.3 Schematic representation <strong>of</strong> first order kinetics <strong>of</strong> (A) the plasma<br />

concentration (C p) <strong>of</strong> a chemical versus the urinary elimination rate (dQ/dt), (B) the<br />

relation between the elimination rate in plasma and urine and (C) the cumulative<br />

excretion ( (%)) versus time. In (B): slope=–k el/2.303 and t 1/2=0.693/k el.<br />

Figure 2.4 Urinary excretion <strong>of</strong> a hypothetical metabolite during 3 days <strong>of</strong><br />

intermittent exposure: t 1/2


urinary concentration at certain time points the net total cumulative<br />

excretion <strong>of</strong> day 2 can be calculated.<br />

Monitoring in occupational toxicology<br />

N.P.E.VERMEULEN ET AL. 19<br />

In occupational toxicology generally four monitoring approaches are<br />

distinguished, namely: environmental monitoring (EM), biological<br />

monitoring (BM), biological effect monitoring (BEM) and health<br />

surveillance (HS) (Figure 2.5). EM and BM are concerned with the<br />

measurement and assessment <strong>of</strong> ambient exposure and health risk<br />

compared to appropriate references. EM determines xenobiotics at the<br />

workplace, BM determines xenobiotics or their metabolites in tissues or<br />

secreta. BEM is concerned with the measurement and assessment <strong>of</strong> early,<br />

non-adverse, biological alterations in exposed workers to evaluate<br />

exposure and/or health risk compared to appropriate references. HS is<br />

concerned with periodic medico-physiological examination <strong>of</strong> exposed<br />

workers with the objective <strong>of</strong> protecting and preventing occupationally<br />

related diseases (Zielhuis and Henderson, 1986).<br />

EM was shown to be <strong>of</strong> limited value for assessing the internal dose <strong>of</strong> a<br />

chemical by not taking into account for example toxicokinetic and<br />

toxicodynamic processes determining the ultimate fate <strong>of</strong> xenobiotics in the<br />

body. To a certain extent, BM appeared to overcome the problems<br />

inherently related to EM. BM assesses the overall exposure to xenobiotics<br />

that are present at the workplace through measurement <strong>of</strong> the appropriate<br />

determinant(s) in biological specimens collected from the worker at specific<br />

timepoints (ACGIH, 1990).<br />

Ideally, not only the relation between exposure and effect is known, but<br />

also the toxicokinetic and toxicodynamic interactions linking these two. If<br />

these processes are elucidated, quantitative knowledge <strong>of</strong> a determinant <strong>of</strong><br />

one <strong>of</strong> the different monitoring methods allows an assessment either <strong>of</strong> the<br />

level <strong>of</strong> exposure or <strong>of</strong> the level <strong>of</strong> effect (Figure 2.5). For example, the<br />

level <strong>of</strong> urinary mercapturic acid excretion could assess the potential health<br />

hazard <strong>of</strong> an occupational exposure situation (Henderson et al., 1989).<br />

In practice, a complete view on the relation between toxicokinetics and<br />

toxicodynamics has not been elucidated for a single chemical up to now.<br />

Occupational monitoring methods all have their specific values based on<br />

their selectivity, sensitivity, validity and logistics and should therefore be<br />

used complementary to each other. All methods operate on the continuum<br />

from exposure to effect, the limits between which occupational toxicology<br />

studies operate.


20 TOXICOKINETICS AND BIODISPOSITION OF INDUSTRIAL CHEMICALS<br />

Figure 2.5 Occupational monitoring methods and their relation to exposure versus<br />

effect assessment and to toxicokinetic and toxicodynamic processes. (Adapted from<br />

Henderson et al., 1989).<br />

Glutathione conjugation products as biomarkers<br />

In principle, GSH-conjugation derived metabolites can be used as a<br />

biomarker <strong>of</strong> internal dose. Glutathione (GSH), a tripeptide consisting <strong>of</strong><br />

the amino acids glycine, cysteine and -glutamine, plays an important role<br />

in the detoxification <strong>of</strong> potentially electrophilic chemicals or metabolites. In<br />

contrast, toxification via GSH-conjugation, for example <strong>of</strong> 1,2dibromoethane,<br />

hexachlorobutadiene, benzyl- and allylisothiocyanate has<br />

also been reported. β-lyase dependent bioactivation <strong>of</strong> cysteine-conjugates,<br />

derived from the initially formed GSH-conjugates, sometimes resulted in<br />

the formation <strong>of</strong> new reactive intermediates which are responsible for<br />

carcinogenic, mutagenic and other toxicological effects (Vermeulen, 1989;<br />

Van Welie et al., 1992).<br />

The initial step in GSH-conjugation is reaction <strong>of</strong> the nucleophilic<br />

sulphhydryl with electrophilic centers <strong>of</strong> a chemical. GSH-conjugation is<br />

catalysed by a family <strong>of</strong> glutathione S-transferase (GST) enzymes. A wide<br />

range <strong>of</strong> chemicals can be handled by this enzyme system due to the


existence <strong>of</strong> a large number <strong>of</strong> isoenzymes with different, though<br />

overlapping, substrate selectivity. The final detoxification capacity through<br />

GSH and GST enzymes <strong>of</strong> an organism depends on endogenous factors<br />

such as tissue distribution, genetic deficiencies, aging and hormonal<br />

influences and on exogenous factors such as sensitivity to inhibition and<br />

induction <strong>of</strong> GSTs (Vermeulen, 1989; Van Welie et al., 1992).<br />

GSH-conjugates normally are not excreted unchanged in urine or faeces.<br />

Catabolism <strong>of</strong> the GSH-conjugates results in the formation and excretion<br />

<strong>of</strong> a variety <strong>of</strong> sulphur containing metabolites, among which thioethers and<br />

mercapturic acids (S-substituted N-acetyl-cysteine conjugates) belong to the<br />

most important. The mercapturic acid pathway is shown in Figure 2.6.<br />

Thioethers in human studies<br />

N.P.E.VERMEULEN ET AL. 21<br />

Figure 2.6 Schematic representation <strong>of</strong> the mercapturic acid pathway: GSHconjugation<br />

with an electrophilic chemical (RX) and the biosynthesis to a<br />

mercapturic acid. E1: glutathione S-transferase, E2: -glutamyltranspeptidase, E3:<br />

cysteinylglycinase and aminopeptidase, E4: cysteine conjugate N-acetyltransferase,<br />

E5: N-deacetylase.<br />

Several years ago, Seutter-Berlage et al. proposed the appearance <strong>of</strong><br />

thioethers such as mercapturic acids (R-S-R′), mercaptans (R-SH) and<br />

disulfides (R-S-S-R′) in urine as an indicator <strong>of</strong> exposure to potentially<br />

alkylating chemicals. The thioether assay is an aselective assay to detect<br />

metabolic end-products excreted in urine <strong>of</strong> (non)occupational exposure to<br />

various electrophilic chemicals. It includes three steps, namely: (i)<br />

extraction, (ii) alkaline hydrolysis and (iii) derivatization, subsequently<br />

followed by spectrophotometric analysis at 412 nm. The thioether assay


22 TOXICOKINETICS AND BIODISPOSITION OF INDUSTRIAL CHEMICALS<br />

Figure 2.7 Urinary excretion <strong>of</strong> thioethers (mmol SH/mol creatinine), <strong>of</strong> applicators<br />

exposed to 3.8 (− −), 9.8 (− −) and 18.9 (− −) mg m −3 8-h TWA (Z+E)-1,3dichloropropene<br />

in respiratory air, respectively. Darker shaded areas indicate<br />

exposure periods.<br />

was first applied to compare thioether excretions in urine <strong>of</strong> employees <strong>of</strong> a<br />

chemical plant. Highest thioether excretions were found in rubber workers<br />

and radial tyre builders when compared with clerks, plastic monomer<br />

mixers and footwear preparers. Recently, urinary thioether excretion was<br />

related to the occupational respiratory exposure <strong>of</strong> applicators in the Dutch<br />

flower-bulb culture to 1,3-dichloropropene (DCP) (Van Welie et al.,<br />

1991a). Instead <strong>of</strong> a discrete comparison <strong>of</strong> thioether excretion with<br />

exposed versus non-exposed groups, in this study thioether excretion was<br />

related to a continuous scale <strong>of</strong> airborne DCP concentrations.<br />

Significant linear relations between respiratory exposure to DCP and postminus<br />

preshift thioether concentration and cumulative thioether excretion<br />

were found. The urinary excretion <strong>of</strong> DCP-thioethers followed first-order<br />

elimination kinetics (Figure 2.7) with half-lives <strong>of</strong> elimination <strong>of</strong> 8.0±2.5 h<br />

(n=5) based on urinary excretion rates and 9.5±3.1 h (n=5) based on<br />

creatinine excretion. The elimination half-lives <strong>of</strong> the thioethers were<br />

almost two fold higher when compared to the half-lives <strong>of</strong> elimination <strong>of</strong><br />

the mercapturic acids <strong>of</strong> Z-and E-1,3 dichloropropene. This illustrates the<br />

main problem <strong>of</strong> urinary thioethers, viz. high background levels originating<br />

from endogenous or exogenous sources, such as smoking and diet (e.g.<br />

horse radish, onion and garlic).


Mercapturic acids in human studies<br />

Mercapturic acids, S-substituted N-acetyl-L-cysteine S-conjugates, in urine<br />

can be used as biomarkers <strong>of</strong> internal dose <strong>of</strong> electrophilic xenobiotics.<br />

Mercapturic acids are metabolic end products <strong>of</strong> GSH-conjugation <strong>of</strong><br />

various potentially electrophilic chemicals (Figure 2.6). The first<br />

mercapturic acids were identified in 1879 as sulphur containing<br />

metabolites after administration <strong>of</strong> bromobenzene to dogs (see references in<br />

Vermeulen, 1989). Since then mercapturic acids from many chemicals have<br />

been identified and these types <strong>of</strong> urinary metabolites have been used in<br />

biotransformation, biological monitoring and toxicological studies<br />

(Vermeulen, 1989; Van Welie et al., 1992).<br />

Commercial availability <strong>of</strong> reference compounds and the development <strong>of</strong><br />

a number <strong>of</strong> different analytical techniques attributed to the popularity <strong>of</strong><br />

mercapturic acids in biological monitoring studies during the last few<br />

years. Urinary excretion <strong>of</strong> the stereoisomeric mercapturic acids <strong>of</strong> Z- and<br />

E-1,3-dichloropropene, a soil fumigant frequently used in agriculture,<br />

proved to be a suitable biomarker for the exposure to both isomers in man.<br />

Strong correlations were observed between 8-h time weighted average<br />

exposure to Z- and E-DCP and complete cumulative excretion <strong>of</strong> N-acetyl-<br />

S-(Z- and E-3-chloropropenyl-2)-L-cysteine in urine. N-acetyl-S-<br />

(cyanoethyl)-L-cysteine was proposed as biomarker <strong>of</strong> exposure to<br />

acrylonitrile. The best correlation between uptake <strong>of</strong> acrylonitrile via the<br />

lungs and excretion <strong>of</strong> the cyanoethyl mercapturic acid in urine was<br />

obtained in samples collected between the sixth and the eighth hour after<br />

the beginning <strong>of</strong> exposure (Jakubowoski et al., 1987). The phenyl<br />

mercapturic acid <strong>of</strong> benzene was regarded as a useful biomarker <strong>of</strong><br />

exposure below 1 ppm <strong>of</strong> workers in a chemical production plant<br />

(Stommel et al., 1989). The use <strong>of</strong> certain foodstuffs and drugs may also<br />

give rise to the excretion <strong>of</strong> mercapturic acids. Consumption <strong>of</strong> cabbage<br />

and horse radish for example gave rise to increased thioether excretion.<br />

Consumption <strong>of</strong> garlic and onions resulted in the excretion <strong>of</strong> N-acetyl-S-<br />

(allyl- and 2-carboxypropyl)-L-cysteine in urine (Van Welie et al., 1992).<br />

The hypnotic drug ( α-bromo-isovalerylurea<br />

also gave rise to the excretion<br />

<strong>of</strong> two diastereomeric α-bromoisovalerylurea<br />

mercapturic acid conjugates<br />

in urine (Mulders et al., 1993). S-Phenyl mercapturic acid was present in<br />

urine <strong>of</strong> groups <strong>of</strong> smokers and non-smokers, not exposed to benzene, in<br />

concentrations <strong>of</strong> 4.0±4.0 µg g −1 creatinine (Stommel et al., 1989).<br />

Toxicokinetics<br />

N.P.E.VERMEULEN ET AL. 23<br />

Knowledge about the toxicokinetics <strong>of</strong> mercapturic acids is necessary to<br />

develop optimal sampling strategies in occupational studies. Urinary<br />

excretion rates <strong>of</strong> mercapturic acids theoretically may reflect the rates <strong>of</strong>


24 TOXICOKINETICS AND BIODISPOSITION OF INDUSTRIAL CHEMICALS<br />

Figure 2.8 Urinary excretion (– –=Z, − −=E) and cumulative excretion (– –=Z,<br />

− −=E) <strong>of</strong> Z- and E-DCP-MA <strong>of</strong> an applicator due to an 8-h TWA respiratory<br />

exposure to 2.32 mg m −3 Z-DCP and 1.73 mg m −3 E-DCP. In (A) the mercapturic acid<br />

excretion rate is depicted and in (B) the mercapturic acid excretion based on<br />

creatinine excretion.<br />

elimination <strong>of</strong> the parent compounds from blood and can be used to<br />

calculate the (complete) cumulative excretion <strong>of</strong> mercapturic acids related<br />

to exposure. By knowing an individual’s mercapturic acid excretion rate,<br />

the contribution to urinary mercapturic acid excretion <strong>of</strong> the day under<br />

study on succeeding day(s) can be calculated. The contributions <strong>of</strong> previous<br />

days <strong>of</strong> exposure can also be used to correct the mercapturic acid excretion<br />

<strong>of</strong> the exposure day under study. The urinary half-life <strong>of</strong> elimination is<br />

inversely proportional to the elimination rate constant. The urinary halflife<br />

<strong>of</strong> both mercapturic acids <strong>of</strong> Z- and E-DCP in man was ca. 5 h<br />

(Figure 2.8) and they were not significantly different, i.e. 5.0±1.2 h for Z-<br />

DCP-MA and 4.7±1.3 h for E-DCP-MA. Strong corre-lations (r≥0.93) were<br />

observed between respiratory 8 h time weighted average (TWA) exposure<br />

to Z- and E-DCP and complete cumulative urinary excretion <strong>of</strong> Z- and E-<br />

DCP-MA. There is still a lack <strong>of</strong> knowledge about the magnitude <strong>of</strong> the


N.P.E.VERMEULEN ET AL. 25<br />

intra- and inter-individual differences in GSH-conjugation and mercapturic<br />

acid excretion. Factors causing these differences are sex, stress, diet, age,<br />

enzyme induction and inhibition, pathology and genetic variability. Apart<br />

from these factors the presence or absence <strong>of</strong> glutathione S-transferases<br />

(GSTs) or GST activity in different persons is <strong>of</strong> special interest in relation<br />

to urinary mercapturic acid excretion. The most intriguing factor known in<br />

this context is the human genetic polymorphism <strong>of</strong> mu-class GSTs. The<br />

GST isoenzyme µ is expressed only in approximately 60% <strong>of</strong> the human<br />

population. Mu-class GST isoenzymes showed a high specific activity<br />

towards for example styrene-7,8-oxide and benzo(a)pyrene-4,5dihydrodiol-4,5-oxide<br />

and E- and Z-DCP. Genetic polymorphism <strong>of</strong> muclass<br />

GSTs was postulated as a determinant in the excretion <strong>of</strong> the<br />

mercapturic acids <strong>of</strong> Z- and E-DCP in occupationally exposed applicators.<br />

However, between mu-class positive (n=9) and mu-class<br />

Table 2.2 Urinary excretion levels, urinary ratios and half-lives <strong>of</strong> elimination <strong>of</strong> Zand<br />

E-DCP mercapturic acids <strong>of</strong> mu-class positive and mu-class negative<br />

individuals a<br />

a Urinary excretion level represents the cumulative excretion <strong>of</strong> Z- and E-DCP-MA<br />

in 0–36 h urine, corrected for the time weighted average 8-h exposure to Z- and E-<br />

DCP. Values are expressed as means±SD for the number <strong>of</strong> individuals indicated in<br />

parentheses.<br />

b (mmol mercapturic acid)/(mmol DCP m −3 ).<br />

c Z-DCP-MA/E-DCP-MA<br />

d Half-life <strong>of</strong> elimination<br />

negative (n=3) applicators, neither a difference in urinary half-lives <strong>of</strong><br />

elimination nor in cumulative excretion <strong>of</strong> both mercapturic acids <strong>of</strong> Zand<br />

E-DCP was seen (Vos et al., 1991) (Table 2.2).<br />

α-Bromoisovalerylurea,<br />

a sedative and hypnotic drug, is a racemic drug<br />

which is also metabolized by GSH-conjugation. It was proposed as a<br />

model substrate to study the pharmacokinetics and stereoselectivity <strong>of</strong> GSHconjugation<br />

in humans. Stereoselective mercapturic acid formation <strong>of</strong> Rand<br />

S-α-bromoisovalerylurea was seen in in vitro studies with purified GST<br />

isoenzymes and in vivo in rat and man. In humans, a pronounced<br />

stereoselectivity in urinary mercapturic acid excretion was observed. Of an<br />

oral dose <strong>of</strong> R- and S-α-bromoisovalerylurea, 22.5±4.3 and 5.7±1.6% was<br />

excreted as mercapturic acid in 24 h, respectively. The half-lives <strong>of</strong><br />

elimination <strong>of</strong> both diastereoisomeric mercapturic acids were 1.5±0.4 and<br />

3.1±1.3 h, respectively. Both the pharmacokinetics <strong>of</strong> α-bromoisovaleryl


26 TOXICOKINETICS AND BIODISPOSITION OF INDUSTRIAL CHEMICALS<br />

Figure 2.9 Proposed biotransformation pathway <strong>of</strong> etridiazol leading to 5-ethoxy-1,<br />

2,4-thiadiazole-3-carboxylic acid (ET-CA) and N-acetyl-S-(ethoxy-1,2,4thiadiazol-3-yl-methyl)-L-cysteine<br />

(ET-MA) in rat and humans. Unidentified<br />

intermediates are presented between brackets ([...]). GSH: glutathione, MAP:<br />

mercapturic acid pathway.<br />

ureas and their stereoselectivity, however, were not found to be different for<br />

subjects who were GSH S-transferase class mu deficient and subjects who<br />

were not (Mulders et al., 1993).<br />

Disposition <strong>of</strong> etridiazol<br />

Etridiazol (Aaterra; 5-ethoxy-3-trichloromethyl-l,2,4-thiadiazole<br />

(Figure 2.9)) is an agricultural fungicide used to control phycomycetous<br />

fungi in, for example, plants, tomatoes, cucumbers, cauliflowers and<br />

celery. Concerning external exposure <strong>of</strong> applicators (e.g. greenhouse<br />

handgunners and foggers) it has been concluded that exposure may occur<br />

through inhalation and dermal absorption. For the purpose <strong>of</strong> the<br />

development <strong>of</strong> a biomonitoring assay disposition studies were performed<br />

recently in rats and human volunteers (Van Welie et al., 1991c). Two<br />

metabolites, 5-ethoxy-l,2,4-thiadiazole-3-carboxylic acid (ET-CA) and a<br />

mercapturic acid, N-acetyl-S-(5-ethoxy-l,2,4-thiadiazol-3-yl-methyl)-Lcysteine<br />

(ET-MA) were identified as new metabolites. Based on a<br />

preliminary toxicokinetic study, the urinary excretion <strong>of</strong> the former<br />

metabolite amounted to 22±9% <strong>of</strong> an oral dose <strong>of</strong> etridiazol (while ET-MA<br />

and unchanged etridiazol were less than 1 % <strong>of</strong> the dose), ET-CA was<br />

proposed as a possible biomarker <strong>of</strong> exposure to this fungicide.


1,1,2-Trichloroethylene<br />

N.P.E.VERMEULEN ET AL. 27<br />

The solvent properties <strong>of</strong> 1,1,2-trichloroethylene (TRI) have resulted in its<br />

widespread use in metal degreasing and a wide variety <strong>of</strong> other industrial<br />

applications. TRI has now been in common use for more than 50 years.<br />

During this period <strong>of</strong> time, workers have been exposed to a wide range <strong>of</strong><br />

concentrations, in some cases for periods <strong>of</strong> 25 years or longer. This has<br />

allowed the compilation <strong>of</strong> a great data base about the effects <strong>of</strong> TRI on<br />

human health. Moreover, information has been supplemented by<br />

numerous studies in experimental animals.<br />

Epidemiological studies on more than 15000 individuals with a followup<br />

<strong>of</strong> more than 25 years have shown no evidence <strong>of</strong> an association<br />

between human exposure to TRI and increased incidence <strong>of</strong> cancer or<br />

cancer mortality. However, several <strong>of</strong> these studies had more or less serious<br />

shortcomings. A summary <strong>of</strong> effects related to TRI and/or TRI-related<br />

metabolism is given in Table 2.3. These and other data are taken from<br />

Goeptar et al., 1995a.<br />

An increased incidence <strong>of</strong> lung tumors has been reported in female<br />

B 6C 3F 1 and male Swiss mice exposed to TRI by inhalation. The effect was<br />

not observed in male B 6C 3F 1 nor in female Swiss mice nor in rats. This<br />

apparent strain-, sex- and lung-specific response fails to resolve the issue <strong>of</strong><br />

whether or not TRI is a carcinogenic hazard to man. Mechanistic studies<br />

on mouse lung tumor formation have explained the sex and species<br />

differences. In this context, chloral formation (Figure 2.10) in Clara cells,<br />

containing relatively high cytochrome P-450 concentrations, has been<br />

identified to be responsible for the development <strong>of</strong> mouse lung tumors.<br />

Importantly, lung tumors have not been found in humans after long-term<br />

occupational exposure in TRI.<br />

TRI causes an increase in the incidence <strong>of</strong> liver cancer in both sexes <strong>of</strong><br />

B 6C 3F 1 and Swiss mice following either gavage or inhalatory exposure, but<br />

not in NMRI and Ha: ICR mice nor in rats. A rodent specific link between<br />

peroxisome proliferation, DNA synthesis, inhibition <strong>of</strong> intercellular<br />

communication and cancer (Table 2.3) suggests that these responses are the<br />

basis <strong>of</strong> the hepatocarcinogenicity induced by TRI. The identification <strong>of</strong><br />

TCA in cancer bioassays as the responsible metabolite for these effects<br />

confirmed this hypothesis. However, when TCA was administered to both<br />

rats and mice, liver cancer was only observed in mice and not in rats. The<br />

reason for this species selectivity in liver effects is explained by the kinetic<br />

behavior <strong>of</strong> TRI and TCA in rodents. Both rats and mice have a considerable<br />

capacity to metabolize TRI to TCA and TCE, the maximal capacities being<br />

closely related to the relative surface areas rather than to their body<br />

weights. Oxidative metabolism <strong>of</strong> TRI in rats is linearly related to dose at<br />

lower dose levels, but it becomes saturated at higher dose levels. Thus, an<br />

important difference between rats and mice is the lower saturation


28 TOXICOKINETICS AND BIODISPOSITION OF INDUSTRIAL CHEMICALS<br />

Table 2.3 Reported toxic effects related to TRI and/or TRI-derived metabolites<br />

a References: see review Goeptar et al., 1995b.<br />

n.d.: not determined.


N.P.E.VERMEULEN ET AL. 29<br />

Figure 2.10 Oxidative metabolism <strong>of</strong> TRI in the rodent and mammalian liver and<br />

the formation <strong>of</strong> metabolites which are excreted in the urine.<br />

concentration in the former species. The relevance <strong>of</strong> the mechanisms <strong>of</strong><br />

liver tumor formation in B 6C 3F 1 and Swiss mice for humans exposed to<br />

TRI has been assessed in studies comparing metabolic rates in mice, rats<br />

and humans. In contrast to the rat, the oxidative metabolism <strong>of</strong> TRI to<br />

TCA in humans is not limited by saturation. In this respect, humans<br />

resemble the mouse and might be able to produce sufficient TCA to induce<br />

peroxisome proliferation and consequently liver cancer. However, there are<br />

significant differences between mice and humans. First, humans metabolize<br />

approximately 60 times less TRI on a body weight basis than mice at<br />

similar exposure levels. Second, TCA has been shown to induce<br />

peroxisome proliferation in mouse hepatocytes but not in human<br />

hepatocytes (Table 2.3). Consequently, the combination <strong>of</strong> extensive<br />

oxidative metabolism <strong>of</strong> TRI to TCA and the ability <strong>of</strong> TCA to induce<br />

peroxisome proliferation appear to be unique to B 6C 3F 1 and Swiss mice.<br />

TRI-induced renal toxicity and tumors were found in Sprague-Dawley,<br />

Fischer 344 and Osborne-Mendel rats. These nephrocarcinogenic effects <strong>of</strong><br />

TRI were specific to male rats and were not seen in female rats nor in mice<br />

<strong>of</strong> either sex. 1,2-DCV-Cys, formed from TRI via the mercapturic acid<br />

pathway, has been identified as a likely metabolite involved in the observed<br />

renal toxicity and probably also in renal carcinogenicity in rats. TRI is<br />

metabolized by a minor pathway involving initial hepatic GSH-conjugation<br />

<strong>of</strong> TRI. The resulting DCV-G is further metabolized (Figure 2.11) and<br />

excreted in urine as two regioisomeric mercapturic acids, namely vicinal 1,<br />

2-DCV-Nac and geminal 2,2-DCV-Nac (Figure 2.11). 1,2-DCV-Cys (the<br />

precursors <strong>of</strong> 1,2-DCV-Nac) is a substrate for the renal L-cysteine S-


30 TOXICOKINETICS AND BIODISPOSITION OF INDUSTRIAL CHEMICALS<br />

Figure 2.11 Possible routes <strong>of</strong> metabolism <strong>of</strong> S-(1,2-dichlorovinyl)glutathione (1,2-<br />

DCV-G). Steps are catalyzed by (a) -glutamyltransferase; (b) cysteinylglycine<br />

dipeptidase; (c) L-cysteine S-conjugate β-lyase; (d) L-cysteine S-conjugate Nacetyltransferase;<br />

(e) acylase.<br />

conjugate β-lyase and it is more mutagenic and cytotoxic than 2,2-DCV-<br />

Cys (the precursors <strong>of</strong> 2,2-DCV-Nac).<br />

The bioactivation <strong>of</strong> 1,2-DCV-Cys is without a doubt a crucial step in<br />

the onset <strong>of</strong> nephrotoxicity in the rat, although the precise biological<br />

mechanisms by which these metabolites exert their nephrocarcinogenic<br />

effects are not yet fully understood. A key aspect in the onset <strong>of</strong><br />

nephrocarcinogenicity in rats, however, is that it will not occur in the<br />

absence <strong>of</strong> nephrotoxicity. This suggests that the alkylating effects <strong>of</strong> the<br />

reactive metabolites (most likely thioketenes) derived from bioactivation <strong>of</strong><br />

1,2-DCV-Cys by β-lyase may not be sufficient to cause kidney tumors. The<br />

specific activity <strong>of</strong> β-lyase, the key enzyme involved in the bioactivation <strong>of</strong><br />

DCV-Cys isomers, is similar in humans to that in the mouse and only 10%<br />

<strong>of</strong> that in the rat. Moreover, human TRI metabolism via the mercapturic<br />

acid pathway resembles that <strong>of</strong> the mouse. It is, therefore, questionable<br />

whether humans are able to produce sufficient DCV-Cys isomers from TRI<br />

to cause first nephrotoxicity and then nephrocarcinogenicity. An important<br />

finding is also that the occurrence <strong>of</strong> nephrotoxicity and


N.P.E.VERMEULEN ET AL. 31<br />

nephrocarcinogenicity in the male rat is dose-dependent. More specifically,<br />

cytotoxic kidney damage is a feature <strong>of</strong> high continuous exposure to TRI<br />

over prolonged periods <strong>of</strong> time. This is unlikely to occur in humans during<br />

occupational exposure. In fact, TRI has been found not to be nephrotoxic<br />

in humans chronically exposed to low levels <strong>of</strong> TRI (50 mg m −3 ).<br />

Consequently, it is unlikely that the renal tumors which are seen in rats at<br />

nephrotoxic dose levels <strong>of</strong> TRI and which are related to β-lyase mediated<br />

bioactivation <strong>of</strong> 1,2-DCV-Cys, are relevant to human health hazards at<br />

reasonably foreseeable levels <strong>of</strong> exposure.<br />

Physiologically based toxicokinetic modeling <strong>of</strong> 1,3butadiene<br />

Physiologically based pharmaco(toxico)-kinetic models differ from the<br />

conventional compartmental models in that they are based to a large extent<br />

on the actual physiology <strong>of</strong> the organism. Instead <strong>of</strong> compartments defined<br />

largely by the experimental data themselves, actual organ and tissue groups<br />

are used with weights and blood flows from the literature (Bisch<strong>of</strong> and<br />

Brown, 1966). Instead <strong>of</strong> composite rate constants determined by fitting<br />

the actual experimental data, physical-chemical and biochemical constants<br />

<strong>of</strong> the compound are used. The result is a mode which predicts the<br />

qualitative behavior <strong>of</strong> the experimental time course without being based<br />

on it. Refinements <strong>of</strong> the model to incorporate additional insights gained<br />

from comparison with experimental data yields a model which can be used<br />

for quantitative extrapolations well beyond the range <strong>of</strong> experiments. In<br />

recent years several PBTK- and PBPK-models have been published: for<br />

methylene chloride, see Andersen et al., 1987; for a review see Leung et al.,<br />

1988; for 1,3-butadiene, see Evelo et al., 1993.<br />

The development <strong>of</strong> a PBTK/PBPK model can be divided into a number<br />

<strong>of</strong> steps: (a) inventory <strong>of</strong> physiological and toxicological behaviour <strong>of</strong> the<br />

compound, (b) mathematical description <strong>of</strong> the biochemical/(patho)<br />

physiological processes involved, (c) parameterization <strong>of</strong> the mathematical<br />

descriptions, (d) the construction <strong>of</strong> the model, (e) refinement and<br />

validation <strong>of</strong> the model and (f) use <strong>of</strong> the predictions and risk assessment.<br />

As an illustrative example <strong>of</strong> this approach the recently described PBTKmodeling<br />

<strong>of</strong> 1,3-butadiene disposition and toxicity might be used (Evelo et<br />

al., 1993). 1,3-Butadiene used for the production <strong>of</strong> styrene-butadiene<br />

rubber, is known amongst others to cause lung carcinogenicity. In the rat<br />

the carcinogenicity <strong>of</strong> 1,3-butadiene is less pronounced while the evidence<br />

for human carcinogenicity is inconclusive, Monoand di-epoxy-butadiene<br />

are reactive metabolites held responsible for this effect. Butadiene<br />

monoxide is formed by microsomal fractions <strong>of</strong> the lung and liver <strong>of</strong> several


32 TOXICOKINETICS AND BIODISPOSITION OF INDUSTRIAL CHEMICALS<br />

Figure 2.12 Physiologically based toxicokinetic model for description <strong>of</strong> butadiene<br />

distribution and metabolism in mice, rats and humans. Gas exchange occurs in the<br />

alveoli <strong>of</strong> the lung. Metabolism occurs in both the alveolar and bronchial areas <strong>of</strong><br />

the lung and in the liver. Metabolic activity in the three other compartments is<br />

ignored (Evelo et al., 1993).<br />

species. There are, however, large interspecies differences in the lung vs<br />

liver activities: mice>rats>humans/monkeys.<br />

The PBTK model used to describe butadiene distribution and metabolism<br />

in mice, rats and humans is shown in Figure 2.12. Gas exchange is<br />

supposed to occur in the alveoli <strong>of</strong> the lung and metabolism in both the<br />

alveolar and bronchial areas <strong>of</strong> the lung and in the liver. By using the<br />

experimentally determined or estimated species-selective parameters for


volumes, masses and blood flows <strong>of</strong> different organs, partition coefficients<br />

<strong>of</strong> 1,3-butadiene between blood and organs/tissues and for metabolic<br />

capacities in liver and lung (bronchial and alveolar areas), accurate dosedependent<br />

simulations were performed for the uptake <strong>of</strong> 1,3-butadiene in<br />

mice and rats in gas-closed chambers. Moreover, with the resulting model<br />

the relative importance <strong>of</strong> lung metabolism as compared to metabolism in<br />

the liver was predicted for the three different species. Lung metabolism<br />

appeared to be much more important than liver metabolism in mice, this in<br />

contrast to the situation in the rat and humans. Moreover, at low exposure<br />

concentrations the relative importance <strong>of</strong> lung metabolism was predicted to<br />

increase in mice as a result <strong>of</strong> diminished saturation <strong>of</strong> metabolism in this<br />

species. It was concluded that the observed species differences in lung vs<br />

liver metabolism <strong>of</strong> 1,3-butadiene (mice>rat>human) and the tendency<br />

towards increased lung metabolism at low doses might rationalize the<br />

observed species differences in the lung carcinogenicity <strong>of</strong> 1,3-butadiene<br />

and this knowledge should be useful in the in vivo extrapolation from high<br />

dose to low dose risk assessments within one species as well as in<br />

interspecies risk assessment extrapolations.<br />

Conclusions<br />

In conclusion, a pr<strong>of</strong>ound knowledge <strong>of</strong> the biodisposition and the<br />

toxicokinetics <strong>of</strong> a toxic or potentially toxic chemical is <strong>of</strong> utmost<br />

importance to the design and interpretation <strong>of</strong> laboratory assessments <strong>of</strong><br />

toxicity, to explain interspecies differences in toxicities and to extrapolate<br />

more reliably from animal experiments to man in the process <strong>of</strong> risk<br />

assessment. This also holds true for the design for proper biological<br />

monitoring procedures and for the interpretation <strong>of</strong> the results in terms <strong>of</strong><br />

potential health risks <strong>of</strong> exposure to chemicals. Apart from traditional<br />

compartment-based toxicokinetic approaches, more recent physiologicallybased<br />

toxicokinetics modeling approaches have distinct advantages for the<br />

above-mentioned purposes.<br />

References<br />

N.P.E.VERMEULEN ET AL. 33<br />

ACGIH, 1990, in 1990–1991 Threshold limit values for chemical substances and<br />

physical agents and biological exposure indices, American Conference <strong>of</strong><br />

Governmental <strong>Industrial</strong> Hygienists, No. 0205.<br />

ANDERSEN, M.E., CLEWELL, H.J., GARGAS, M.L., SMITH, F.A. and REITZ,<br />

R.H., 1987, Physiologically-based pharmacokinetics and the risk assessment for<br />

methylene chloride, Toxicol. Appl. Pharmacol., 87, 185–205.<br />

ARIENS, E.J. and SIMONIS, M.A., 1980, in BREIMER, D.D. (Ed.) Towards better<br />

Safety <strong>of</strong> Drugs and Pharmaceutical Products, Amsterdam: Elsevier<br />

Biomedical Press.


34 TOXICOKINETICS AND BIODISPOSITION OF INDUSTRIAL CHEMICALS<br />

BISCHOF, K.B. and BROWN, R.G., 1966, Drug distribution in mammals, Chem.<br />

Eng. Prog. Symp. Ser., 62(66), 33–45.<br />

CCRX 1989, in Metingen van radioactiviteit en xenobiotische st<strong>of</strong>fen in het<br />

biologische milieu in Nederland 1989 (in Dutch with English summary),<br />

Coördinatie-commissie voor de metingen van radioactiviteit en xenobiotische<br />

st<strong>of</strong>fen, Bilthoven: RIVM.<br />

COMMANDEUR, J.N.M. and VERMEULEN, N.P.E. 1991, Molecular and<br />

biochemical mechanism <strong>of</strong> chemically induced nephrotoxicity: a review, Chem.<br />

Res. Toxicol., 3, 171–94.<br />

EVELO, C.T.A., OOSTENDORP, J.G.M., TEN BERGE, W.F. and BORM, P.J. A.,<br />

1993, Physiologically based toxicokinetic modeling <strong>of</strong> 1,3-butadiene lung<br />

metabolism in mice becomes more important at low doses. Environ. Hlth<br />

Perspect., 101(6), 496–502 (no. 24).<br />

FARMER, P.B., NEUMANN, H.-G. and HENSCHLER, D., 1987, Estimation <strong>of</strong><br />

exposure <strong>of</strong> man to substances reacting covalently with macromolecules, Arch.<br />

Toxicol, 60, 251–60.<br />

GOEPTAR, A.R., COMMANDEUR, J.N.M., OMMEN, B.VAN, BLADEREN,<br />

P.J. VAN and VERMEULEN, N.P.E. 1995a, The metabolism and kinetics <strong>of</strong><br />

trichloroethylene in relation to toxicity and carcinogenicity. Relevance <strong>of</strong> the<br />

Mercapturic Acid Pathway, Chem. Res. Toxicol, 8, 3–21.<br />

GOEPTAR, A.R., SCHEERENS, H. and VERMEULEN, N.P.E., 1995b, Oxygen<br />

and xenobiotic reductase activities <strong>of</strong> cytochrome P450, Crit. Rev. Toxicol.,<br />

25, 25–65.<br />

GUENGERICH, F.P., 1994, Catalytic selectivity <strong>of</strong> human cytochrome P450<br />

enzymes: relevance to drug metabolism and toxicity, Toxicol. Lett., 70, 133–8.<br />

HENDERSON, R.F., BECHTOLD, W.E., BOND, J.A. and SUN, J.D., 1989, The use<br />

<strong>of</strong> biological markers in toxicology, Crit. Rev. Toxicol, 20, 65–82.<br />

HOOK, J.B. and HEWITT, W.R., 1986, Toxic responses <strong>of</strong> the kidney, in Klaassen,<br />

C.D., Doull, J. and Amdur, M.O. (Eds) Casarett and Doull’s <strong>Toxicology</strong>, pp.<br />

310–29, New York: Macmillan.<br />

JAKUBOWOSKI, M., LINHART, I., PIELAS, G. and KOPECKY, J., 1987, 2-<br />

Cyanoethylmercapturic acid (CEMA) in the urine as a possible indicator <strong>of</strong><br />

exposure to acrylonitrile, Brit. J. Ind. Med., 44, 834–40.<br />

KOYMANS, L., DONNÉ-OP DEN KELDER, G.M., TE KOPPELE, J.M. and<br />

VERMEULEN, N.P.E., 1993, Cytochromes P450: their active-site structure<br />

and mechanism <strong>of</strong> oxidation, Drug Metab. Rev., 25, 325–87.<br />

LEUNG, H.W., Ku, R.H., PAUSTENBACH, D.J. and ANDERSEN, M.E., 1988, A<br />

physiologically-based pharmacokinetic model for 2,3,7,8-tetrachlorodibenzo-pdioxin<br />

in C57BL/6J and DBA/2J mice, Toxicol. Lett., 42, 15–28.<br />

MULDERS, T.M.T., VENIZELOS, V., SCHOEMAKER, R., COHEN, A.F.,<br />

BREIMER, D.D. and MULDER, G.J., 1993, Characterization <strong>of</strong> glutathione<br />

conjugation in humans: stereoselectivity in plasma elimination<br />

pharmacokinetics and urinary excretion <strong>of</strong> (R)- and (S)-2-bromoisovalerylurea<br />

in healthy volunteers. Clin. Phar. Ther., 53, 49–58.<br />

POTT, P. 1795, Chirurgical observations relative to the cataract, the polypus <strong>of</strong> the<br />

nose, the cancer <strong>of</strong> the scrotum, the different kinds <strong>of</strong> ruptures and the<br />

mortification <strong>of</strong> the toes and feet, in Haes, Clarke and Collins (Eds) National<br />

Cancer Institute Monograph, 1962, Vol 10, pp. 7–13, London.


N.P.E.VERMEULEN ET AL. 35<br />

SEUTTER-BERLAGE, F., VAN DORP, H.L., KOSSE, H.G.J. and HENDERSON,<br />

P.T.H., 1979, Urinary mercapturic acid excretion as a biological parameter <strong>of</strong><br />

exposure to alkylating agents, Int. Arch. Occup. Environ. Hlth, 39, 45–51.<br />

SIPES, I.G. and GANDOLFI, A.J., 1986, Biotransformation <strong>of</strong> chemicals, in<br />

Klaassen, C.D., Doull, J. and Amour, M.O. (Eds) Casarett and Doull’s<br />

<strong>Toxicology</strong>, pp. 64–98, New York: Macmillan.<br />

STOMMEL, P., MÜLLER, G., STÜCKER, W., VERKOYEN, C., SCHÖBEL, S.<br />

and NORPOTH, K., 1989, Determination <strong>of</strong> S-phenylmercapturic acid in the<br />

urine an improvement in the biological monitoring <strong>of</strong> benzene exposure,<br />

Carcinogenesis, 10, 279–82.<br />

VAN WELIE, R.T.H., VAN MARREWIJK C.M., DE WOLFF, F.A. and<br />

VERMEULEN, N.P.E., 1991a, Thioether excretion in urine <strong>of</strong> applicators<br />

exposed to 1,3-dichloropropene: a comparison with urinary mercapturic acid<br />

excretion, Brit. J. Ind. Med., 48, 492–8.<br />

VAN WELIE, R.T.H., VAN DUYN, P., BROUWER, E.J., VAN HEMMEN, J.J.<br />

and VERMEULEN, N.P.E., 1991b, Inhalation exposure to 1.3dichloropropene<br />

in the Dutch flower-bulb culture. Part II. Biological<br />

monitoring by measurement <strong>of</strong> urinary excretion <strong>of</strong> two mercapturic acid<br />

metabolites, Arch. Environ. Contam. Toxicol, 20, 6–12.<br />

VAN WELIE R.T.H., MENSERT, R.,, VAN DUYN, P. and VERMEULEN, N.P.<br />

E. 1991c, Identification and quantitative determination <strong>of</strong> a carboxylic and a<br />

mercapturic acid metabolite <strong>of</strong> etridiazole in urine <strong>of</strong> rat and man. Potential<br />

tools for biological monitoring. Arch. Toxicol., 65, 625–32.<br />

VAN WELIE, R.T.H., VAN DIJCK, R.G.J.M., VERMEULEN, N.P.E. and VAN<br />

SITTERT, N.J., 1992, Mercapturic acids, protein adducts, and DNA adducts<br />

as biomarkers <strong>of</strong> electrophilic chemicals, Crit. Rev. Toxicol., 22, 271–306.<br />

VERMEULEN, N.P.E., 1989, Analysis <strong>of</strong> mercapturic acids as a tool in<br />

biotransformation, biomonitoring and toxicological studies. TiPS, 10, 177–81.<br />

VERMEULEN, N.P.E., VAN DER STRAAT, R., TE KOPPELE, J.M., BALDEW,<br />

G.S., COMMANDEUR. J.N.M., HAENEN, G.R.M.M., KOYMANS, L. and<br />

VAN WELIE, R.T.H., 1990, Molecular mechanisms in toxicology and drug<br />

design, in Claassen, V. (Ed.) Vol. 13, pp. 253–71, Trends in Drug Research,<br />

Amsterdam: Elsevier.<br />

Vos, R.M.E., VAN WELIE, R.T.H., PETERS, W.H.M., EVELO,<br />

C.T.A., BOOGAARDS, J.J.P., VERMEULEN, N.P.E. and VAN BLADEREN,<br />

P.J., 1991, Genetic deficiency <strong>of</strong> human class mu glutathione S-transferase<br />

isoenzymes in relation to the urinary excretion <strong>of</strong> the mercapturic adds <strong>of</strong> Zand<br />

E-1,3-dichloropropene. Arch. Toxicol., 65, 95–9.<br />

WARHOLM, M., JENSSON, H., TAHIR, M.K. and MANNERVIK, B., 1986,<br />

Purification and characterization <strong>of</strong> three distinct glutathione S-transferases<br />

from mouse liver, Biochemistry., 25, 4119–25.<br />

ZIELHUIS, R.L. and HENDERSON, P.TH., 1986, Definitions <strong>of</strong> monitoring<br />

activities and their relevance for the practice <strong>of</strong> occupational health, Int. Arch.<br />

Occup. Environ. Hlth, 57, 249–57.


3<br />

Metabolic Activation <strong>of</strong> <strong>Industrial</strong> Chemicals and<br />

Implications for Toxicity<br />

GERARD J.MULDER<br />

Leiden University, Leiden<br />

Introduction<br />

In the toxicity <strong>of</strong> industrial chemicals bioactivation (Anders, 1985) plays an<br />

important role. Obviously, its importance depends on the structure <strong>of</strong> the<br />

chemical as well as the toxic effect considered. Thus, inorganic compounds<br />

in general will not require bioactivation: metal salts or oxides will usually<br />

cause toxicity in the form in which they are taken up. However, even these<br />

chemicals may require further metabolism for maximum toxicity in the<br />

body: inorganic mercury may be converted to an organic form<br />

(methylmercury), and nitrate may be reduced to nitrite. It is also possible<br />

that in vivo complexes are being formed, such as between heavy metals<br />

ions and the protein, metallothionein, which may be more toxic (or cause<br />

more organ-selective toxicity) than the original, uncomplexed compound<br />

(Wang et al., 1993).<br />

Bioactivation thus mostly concerns the conversion <strong>of</strong> organic chemicals<br />

to more toxic products. On one hand this may result in stable metabolites<br />

that better fit a receptor binding site, resulting in (in principle) reversible<br />

interactions (Mulder, 1992). On the other hand, the metabolites may be<br />

quite reactive, resulting in essentially irreversible effects which are <strong>of</strong><br />

particular concern when they can escape correction, such as neoplasms or<br />

sensitization.<br />

Mechanisms <strong>of</strong> bioactivation<br />

<strong>Industrial</strong> chemicals have widely different structures. Often the preparations<br />

used contain a variable degree <strong>of</strong> impurities, or are mixtures. In this<br />

chapter only the toxicity <strong>of</strong> pure chemicals will be discussed; obviously<br />

when several compounds are present at the same time in a reaction mix or<br />

a commercial product, the final toxicity may be the result <strong>of</strong> complex<br />

interactions between the substituents, which may cause the toxicity to be<br />

more severe (but also much less serious) than expected.


The bioactivation to reactive intermediates by oxidative, cytochrome<br />

P450-mediated metabolism has been extensively studied. So much so, that<br />

it is <strong>of</strong>ten overlooked that conjugation reactions may similarly convert<br />

stable compounds into reactive, electrophilic metabolites (Anders and<br />

Dekant, 1994). This is <strong>of</strong> some practical importance, because many rapid<br />

in vitro toxicity screening tests, e.g. for genotoxicity, include only oxidative<br />

biotransformation capacity (microsomal fractions plus NADPH). In such<br />

screening systems the possibility that, for example, glucuronidation,<br />

sulfation or glutathione conjugation may activate a chemical is not<br />

assessed. Examples <strong>of</strong> bioactivation <strong>of</strong> industrial chemicals by glutathione<br />

conjugation are various halogenated hydrocarbons, while in 2naphthylamine<br />

toxicity glucuronidation may play a role. All in all,<br />

however, little information is available on the role <strong>of</strong> conjugation. As a<br />

consequence, it is unclear at present whether conjugation reactions are <strong>of</strong><br />

major concern for bioactivation <strong>of</strong> industrial chemicals in general. It<br />

certainly seems worth while for reasons more than just scientific curiosity<br />

to include conjugation reactions in test systems. This can be done by using,<br />

for example, intact hepatocytes (or other cells), or by using a mix <strong>of</strong><br />

cosubstrates for conjugation in combination with an S9 fraction (consisting<br />

<strong>of</strong> both cytosol and microsomal fraction). UDP glucuronic acid, a sulfate<br />

activating system, glutathione, acetyl-CoA and S-adenosylmethionine<br />

would cover the major conjugation reactions.<br />

A role <strong>of</strong> bioactivation in the toxicity <strong>of</strong> many chemicals has been<br />

demonstrated. Chemical groups that <strong>of</strong>ten are involved in mutagenic or<br />

carcinogenic effects have been identified (‘alerting groups’). However, as yet<br />

it is still impossible to predict with certainty the carcinogencity <strong>of</strong> a<br />

compound based only on its chemical structure, although a panel <strong>of</strong><br />

experts can make quite good guesses (Wachsman et al., 1993).<br />

In this chapter some <strong>of</strong> the major issues will be illustrated by the<br />

examples vinyl chloride, styrene (versus styrene oxide), benzene,<br />

dichloromethane, chlor<strong>of</strong>orm, 1,2-dibromoethane and 2-naphthylamine.<br />

Vinyl chloride<br />

G.J.MULDER 37<br />

High exposure <strong>of</strong> workers to vinyl chloride in the past has led to the<br />

realization that it may cause neoplasms in man, in particular<br />

haemangiosarcomas in the liver. Vinyl chloride is a genotoxic compound<br />

that acts as initiator <strong>of</strong> various types <strong>of</strong> tumors (Swaen et al., 1987).<br />

The major routes <strong>of</strong> bioactivation <strong>of</strong> vinyl chloride are shown in<br />

Figure 3.1. The most important first step is oxidation by (a) cytochrome<br />

P450 species, resulting in a rather reactive epoxide, which readily<br />

rearranges to chloroacetaldehyde. This may bind to DNA bases, especially<br />

the N6 <strong>of</strong> adenosine or the N4 <strong>of</strong> cytidine, yielding N-ethenoadducts.<br />

Glutathione provides protectionbecause it traps the reactive intermediates


38 METABOLIC ACTIVATION OF INDUSTRIAL CHEMICALS<br />

Figure 3.1 Bioactivation <strong>of</strong> vinylchloride.<br />

formed from vinyl chloride. Furthermetabolism <strong>of</strong> such conjugates leads to<br />

urinary products that can be used tomonitor vinyl chloride exposure in<br />

workers (Guengerich, 1992).<br />

The compound is mutagenic in many in vitro test systems, which require<br />

bioactivation by a microsomal preparation with co-factors for cytochrome<br />

P450. Whether other toxic effects that have been associated with vinyl<br />

chloride exposure in man, such as Raynauds syndrome or acro-osteolysis,<br />

also require bioactivation <strong>of</strong> vinyl chloride is unknown. In addition to its<br />

DNA adduct forming capacity, vinyl chloride also binds covalently to thiol<br />

groups in proteins. It is conceivable that such binding in specific cell types<br />

might lead to non-carcinogenic defects in organ functions.<br />

Styrene and styrene oxide<br />

Styrene metabolism and bioactivation are very similar to that <strong>of</strong> vinyl<br />

chloride: epoxidation by cytochrome P450 is the pathway <strong>of</strong> toxification<br />

(Figure 3.2). It can be detoxified by epoxide hydrolase and glutathione<br />

transferase activity. Mandelic acid excretion in urine can be used for<br />

exposure monitoring in man. Styrene oxide is a direct mutagen in several in<br />

vitro mutagenesis systems and it readily reacts with DNA in vitro.<br />

However, when animals are exposed to styrene in vivo very little if any<br />

DNA binding is observed. Moreover, styrene is not carcinogenic in animal<br />

experiments, although it is a (weak) mutagen in vitro, after bioactivation<br />

(Bond, 1989; Ecetoc, 1992). The explanation most likely is that the styrene


Figure 3.2 Bioactivation <strong>of</strong> styrene.<br />

Figure 3.3 Bioactivation <strong>of</strong> chlor<strong>of</strong>orm.<br />

oxide, generated in vivo inside a cell is such a good substrate for the phase<br />

2 enzymes, epoxide hydrolase and glutathione transferase, that virtually<br />

immediately upon its synthesis, it is further metabolized. Thus, presumably<br />

the build-up <strong>of</strong> an effective concentration in vivo is prevented. Whether<br />

other toxicity <strong>of</strong> styrene in, for example, oesophagus, stomach or<br />

forestomach is related to covalent binding <strong>of</strong> styrene oxide to protein thiol<br />

groups in those tissues is unclear at present.<br />

Styrene is an example <strong>of</strong> a compound <strong>of</strong> which the metabolism<br />

completely goes through a reactive intermediate (the epoxide); yet it does<br />

not cause the cancer that might be expected from its highly mutagenic<br />

metabolite. Accumulation <strong>of</strong> enough <strong>of</strong> this epoxide inside the cells for a<br />

detectable genotoxic effect may require a dose which is acutely toxic, and<br />

therefore can never be tested.<br />

Chlor<strong>of</strong>orm<br />

G.J.MULDER 39<br />

Chlor<strong>of</strong>orm is acutely toxic in the liver and the kidney. This is the result <strong>of</strong><br />

formation <strong>of</strong> a reactive intermediate (Figure 3.3), phosgene, which binds


40 METABOLIC ACTIVATION OF INDUSTRIAL CHEMICALS<br />

avidly to thiol and amine groups in protein. In mice the kidney toxicity is<br />

much more pronounced in males than in females; this sex-difference is due<br />

to the much higher activity <strong>of</strong> the bioactivating cytochrome P450 species in<br />

male mouse kidney than in the females (Pohl et al., 1984). Chlor<strong>of</strong>orm also<br />

increased the tumor incidence in the liver and kidney in some experiments<br />

(Reitz et al., 1990), at dose levels which damaged these organs. However,<br />

there are no indications <strong>of</strong> mutagenicity or genotoxicity in in vitro or<br />

animal in vivo systems. Therefore, most likely the increased tumor<br />

frequency in animals is due to tissue toxicity, leading to increased cell<br />

turnover and a mitogenic stimulus. This is an important distinction, at least<br />

in some countries such as The Netherlands, because for such chemicals a<br />

threshold approach is allowed, whereas for initiating chemicals a linear<br />

extrapolation for carcinogenic risk is used.<br />

Benzene<br />

Benzene presents something <strong>of</strong> a mystery in the evaluation <strong>of</strong> its toxicity<br />

mechanism (Swaen et al., 1989). Exposure to high levels <strong>of</strong> benzene has<br />

been associated with leukaemia in man. However, in vitro it shows little<br />

genotoxicity, and it hardly generates DNA adducts when it is given even at<br />

high dose to animals. A candidate for DNA damage could have been the 1,<br />

4-dihy-droxybenzene (hydroquinone) metabolite, which, however, does not<br />

form DNA adducts readily. Recently a ring-opened metabolite, the<br />

trans,trans-muconic dialdehyde has been proposed as a possible reactive<br />

metabolite <strong>of</strong> benzene (Figure 3.4). Whether it really plays a role in<br />

benzene toxicity is unclear as yet (Kline et al., 1993).<br />

Dichloromethane<br />

Dichloromethane can be metabolized by two pathways, an oxidative and a<br />

conjugative route. Oxidation catalyzed by P450 yields carbon monoxide<br />

(Figure 3.5). The glutathione pathway generates a reactive intermediate,<br />

which is mutagenic and has been implicated in the hepatocarcinogenic<br />

effect <strong>of</strong> dichloromethane in mice. It could be shown that the human liver<br />

has a negligible activity <strong>of</strong> the glutathione transferase involved, so that the<br />

risk for hepatocarcinogenesis in man is virtually non-existent (Green et al.,<br />

1988; Reitz et al., 1989; Dankovic and Bailer, 1994). This example<br />

illustrates how insight into the mechanism <strong>of</strong> bioactivation enables a more<br />

reliable species extrapolation in terms <strong>of</strong> hazard and risk.<br />

1,2-Dibromoethane<br />

This compound can be conjugated with glutathione to form a reactive<br />

thiiranium ion which forms adducts with DNA. This is the reason for the


Figure 3.4 Possible route <strong>of</strong> bioactivation <strong>of</strong> benzene.<br />

Figure 3.5 Bioactivation <strong>of</strong> dichloromethane.<br />

carcinogenic and mutagenic effects <strong>of</strong> 1,2-dibromoethane (Inskeep et al.,<br />

1986).<br />

2-Naphthylamine<br />

G.J.MULDER 41<br />

2-Naphthylamine causes bladder tumors in the dog and man, but not in<br />

mice and rats. The most likely cause is a complicated interplay between<br />

glucuroni dation and urinary pH. In all four species 2-naphthylamine is Nhydroxylated<br />

and subsequently N-glucuronidated. The resulting metabolite


42 METABOLIC ACTIVATION OF INDUSTRIAL CHEMICALS<br />

is excreted in urine. In man and dog the urine is slightly acidic, while in rat<br />

and mouse it is slightly alkaline. Under acidic conditions the glucuronide is<br />

hydrolyzed to generate the hydroxylamine in the bladder. In this case<br />

glucuronidation is not a bioactivation, but rather a targeting<br />

biotransformation: in man and dog the carcinogenic metabolite is targeted<br />

to the bladder, due to the (necessary!) acidic local pH (Kadlubar et al.,<br />

1981).<br />

Conclusions<br />

The above illustrates the importance <strong>of</strong> bioactivation in toxicity <strong>of</strong><br />

industrial chemicals. Is it possible to predict bioactivation from the<br />

structure? As outlined above, in some cases the compound contains<br />

structural elements which make bioactivation to a reactive intermediate<br />

quite likely. Whether it does play a role in toxicity then is still uncertain.<br />

Test systems to detect reactive intermediates depend on, for example, the<br />

availability <strong>of</strong> the radiolabeled compound; in fact, a very high specific<br />

radioactivity is required to detect low levels <strong>of</strong> binding. Alternatively,<br />

radiolabelled glutathione can be used for those reactive intermediates that<br />

readily bind to the thiol group <strong>of</strong> glutathione (Mulder and Le, 1988).<br />

Whether such systems can pick up every relevant toxic reactive<br />

intermediate remains to be seen.<br />

For extrapolation <strong>of</strong> one species to the other it is important to have<br />

insight into the metabolite that is responsible for the toxicity. Therefore, it<br />

is more than just <strong>of</strong> academic interest to know the mechanism <strong>of</strong> toxicity in<br />

safety assessment <strong>of</strong> industrial chemicals. Unfortunately, it is <strong>of</strong>ten not easy<br />

to establish such a mechanism beyond reasonable doubt: it may require too<br />

many rats to feel comfortable about it if we would have to do this for every<br />

chemical used industrially!<br />

References<br />

ANDERS, M.W. (Ed.), 1985, Bioactivation <strong>of</strong> Foreign <strong>Compounds</strong>, Orlando, FL:<br />

Academic Press.<br />

ANDERS, M.W. and DEKANT, W., 1994, Conjugation-dependent Carcinogenicity<br />

and Toxicity <strong>of</strong> Foreign <strong>Compounds</strong>, Orlando, FL: Academic Press.<br />

BOND, J.A., 1989, Review <strong>of</strong> the toxicology <strong>of</strong> styrene, CRC Crit. Rev. Toxicol 19,<br />

227–49.<br />

DANKOVIC, D.A. and BAILER, A.J., 1994, The impact <strong>of</strong> exercise and<br />

intersubject variability on dose estimates for dichloromethane derived from a<br />

physiologically based pharmacokinetic model, Fund. Appl. Toxicol, 22, 20–5.<br />

ECETOC, 1992, Technical report No. 52, Styrene toxicology. Investigations on the<br />

potential for carcinogenicity, Brussels: Ecetoc.


G.J.MULDER 43<br />

GREEN, T., PROVAN, W.M., COLLINGE, D.C. and GUEST, A.E., 1988,<br />

Macro molecular interactions <strong>of</strong> inhaled methylene chloride in rats and mice,<br />

Toxicol. Appl. Pharmacol, 93, 1–10.<br />

GUENGERICH, F.R., 1992, Roles <strong>of</strong> the vinylchloride oxidation products 1chlorooxirane<br />

and 2-chloroacetaldehyde in the in vitro formation <strong>of</strong> etheno<br />

adducts <strong>of</strong> nucleic acid bases, Chem. Res. Toxicol, 5, 2–5.<br />

INSKEEP, P.B., KOGA, N.K., CMARIK, J.L. and GUENGERICH, F.P., 1986,<br />

Covalent binding <strong>of</strong> 1,2-dihaloalkanes to DNA, Cancer Res., 46, 2839–44.<br />

KADLUBAR, F.F., UNRUH, L.E., FLAMMANG, T.J., SPARKS, D., MITCHUM,<br />

R.K. and MULDER, G.J., 1981, Alteration <strong>of</strong> urinary levels <strong>of</strong> the carcinogen,<br />

N-hydroxy-2-naphthylamine, and its N-glucuronide in the rat by control <strong>of</strong><br />

urinary pH, inhibition <strong>of</strong> metabolic sulfation, and changes in biliary excretion,<br />

Chem.-Biol. Interact. 33, 129–47.<br />

KLINE, S.A., ROBERTSON, J.F., GROTZ, V.L., GOLDSTEIN, B.D. and WITZ,<br />

G., 1993, Identification <strong>of</strong> 6-hydroxy-trans,trans-2,4-hexadienoic acid, a novel<br />

ring-opened urinary metabolite <strong>of</strong> benzene, Environm. Hlth Perspect., 101,<br />

310–12.<br />

MULDER, G.J., 1992, Pharmacological effects <strong>of</strong> drug conjugates: is morphine 6glucuronide<br />

an exception? Trends Pharmacol. Sci., 13, 302–4.<br />

MULDER, G.J. and LE, C.T., 1988, A rapid simple in vitro screening test to detect<br />

reactive intermediates <strong>of</strong> xenobiotics. Toxicol. In Vitro, 2, 225–30.<br />

POHL, L.R., GEORGE, J.W. and SATOH, H., 1984, Strain and sex differences in<br />

chlor<strong>of</strong>orm-induced nephrotoxicity. Drug Metab. Disposit., 12, 304–7.<br />

REITZ, R.H., MENDRALA, A.L. and GUENGERICH, F.P., 1989, In vitro<br />

metabo-lism <strong>of</strong> methylene chloride in human and animal tissues, Toxicol.<br />

Appl. Pharmacol, 97, 230–46.<br />

REITZ, R.H., MENDRALA, A.L. and CONOLLY, R.B., 1990, Estimating the risk<br />

<strong>of</strong> liver cancer associated with human exposures to chlor<strong>of</strong>orm using PbPK<br />

modeling, Toxicol. Appl. Pharmacol., 105, 443–59.<br />

SWAEN, G.M.H. et al., 1987, A scientific basis for the risk assessment <strong>of</strong> vinyl<br />

chloride, Regul. Toxicol. Pharmacol, 7, 120–7.<br />

SWAEN, G.M.H. et al., 1989, Carcinogenic risk assessment <strong>of</strong> benzene in outdoor<br />

air, Regul. Toxicol. Pharmacol., 9, 175–85.<br />

WACHSMAN, J.T., BRISTOL, D.W., SPALDING, J., SHELBY, M. and<br />

TENNANT, R.W., 1993, Predicting chemical carcinogenesis in rodents,<br />

Environm. Hlth Perspect., 101, 444–5.<br />

WANG, X.P., CHAN, H.M., GOYER, R.A. and CHERIAN, M.G., 1993,<br />

Nephrotoxicity <strong>of</strong> repeated injections <strong>of</strong> cadmium-metallothionein in rats,<br />

Toxicol. Appl. Pharmacol., 119, 11–16.


4<br />

Sizing up the Problem <strong>of</strong> Exposure Extrapolation:<br />

New Directions in Allometric Scaling<br />

D.BRUCE CAMPBELL<br />

Director International Scientific Affairs, Servier Research and<br />

Development, Slough<br />

Introduction<br />

The evaluation <strong>of</strong> the safety <strong>of</strong> industrial chemicals requires the<br />

administration <strong>of</strong> a range <strong>of</strong> doses to test animals over periods <strong>of</strong> time and<br />

the extrapolation in some meaningful way to man. Various risk assessment<br />

models have been suggested which attempt to measure an uncertainty or<br />

safety factor which can be used to extrapolate to man to obtain an<br />

acceptable daily intake (ADI) (Dourson and Stara, 1983). Other<br />

approaches are also used, such as benchmark dose, the smallest dose which<br />

produces a statistical increase in toxicity over the background level (Crump,<br />

1984), or more frequently the LOEL, the lowest observed dose which<br />

produces an adverse effect, and NOEL, the highest dose at which no<br />

adverse effect is observed. There are difficulties in the interpretation <strong>of</strong><br />

these exposure margins since there is <strong>of</strong>ten little information on: (1) the<br />

slope or intensity <strong>of</strong> the effect, (2) species differences in the sensitivity, (3)<br />

the possibility <strong>of</strong> cumulative or irreversible toxicities, etc. But perhaps the<br />

most important weakness in these estimates is the lack <strong>of</strong> knowledge <strong>of</strong> the<br />

actual circulating levels <strong>of</strong> the chemical(s) in the different species. This<br />

problem is particularly pertinent for industrial chemicals and environmental<br />

pollutants where it may be unethical to administer doses <strong>of</strong> these<br />

compounds to volunteers which are sufficiently high to measure the<br />

kinetics. It is <strong>of</strong> special concern since it is well known that there are large<br />

interspecies differences in the clearance <strong>of</strong> chemicals and that comparison <strong>of</strong><br />

doses in animals, expressed simply in terms <strong>of</strong> mg kg −1 , provides little<br />

information as to the actual exposure likely to occur. This is not surprising<br />

since small animals have relatively faster blood flow and larger organs than<br />

man when expressed as a percentage <strong>of</strong> body weight, and consequently<br />

clearance is more rapid and circulating levels <strong>of</strong> the administered<br />

compound are lower than could be expected during toxicity testing<br />

(Campbell and Ings, 1988).<br />

However since most mammals share similar physiological and<br />

biochemical actions these differences in physiological rates and sizes for


most processes in the mammalian body have been shown to be<br />

proportional to the body weight <strong>of</strong> the animal (Adolph, 1949; Calabrese,<br />

1983; Peters, 1983; Chappell and Mordenti, 1991) and can be related by<br />

allometry, a word from the Greek meaning the measurement (metry) <strong>of</strong><br />

changing size (allo). It has been shown that blood flow, organ size,<br />

metabolic and respiratory rate, and many other physiological and<br />

anatomical variables are related by the general allometric equation<br />

(Boxenbaum, 1982b):<br />

(4.1)<br />

where Y is the function to be measured, W the body weight <strong>of</strong> the animal,<br />

a the coefficient and b the exponent. For mammals, whilst a is different for<br />

each function, b is approximately 0.6–0.8 for rates, flows and clearances, 1.<br />

0 for volumes and organ sizes, and 0.25 for cycles and times. Thus<br />

metabolic rate can be calculated from 7.0·W 0.75 , liver blood flow from<br />

37·W 0.85 , blood weight from 0.055·W 0.99 , and respiratory rate from 0.<br />

019·W 0.26 . Since the blood flows and the weights <strong>of</strong> the liver and kidney,<br />

the two major organs <strong>of</strong> elimination, can be similarly allometrically scaled,<br />

it follows that the same formula could in principle be used for<br />

extrapolation <strong>of</strong> the clearance <strong>of</strong> chemicals between species.<br />

In the past there has been much discussion on the possibility <strong>of</strong><br />

predicting human kinetics and distribution from animal data, using<br />

allometry. For industrial chemicals relatively complex physiological models<br />

have been constructed using this knowledge <strong>of</strong> relative blood flows and<br />

organ size to predict what levels <strong>of</strong> exposure could be expected in man<br />

(Andersen et al., 1984), but little work has been published on comparative<br />

interspecies clearances which will dictate the circulating levels. For drugs,<br />

on the other hand, a number <strong>of</strong> reports have been published on the<br />

rationale for the use <strong>of</strong> allometric scaling <strong>of</strong> kinetics (Dedrick, 1973;<br />

Boxenbaum, 1982b, 1984, 1986; Mordenti, 1985, 1986; Sawada et al.,<br />

1985; Chappell and Mordenti, 1991) but many have been concerned with<br />

its theoretical aspects rather than with its practical use for prediction.<br />

When scaling has been used, the predictions have not always been<br />

accurate, and the method has therefore not had wide usage. This is<br />

unfortunate since the ability to predict what will be the blood levels in man,<br />

without the need to administer the compound, can potentially have many<br />

advantages in drug development and in the safety testing <strong>of</strong> industrial<br />

chemicals where dosing volunteers is <strong>of</strong>ten unacceptable.<br />

Methods<br />

D.BRUCE CAMPBELL 45<br />

A meta-analysis <strong>of</strong> the papers related to this subject has been made from<br />

those published over the last 20 years. Data before this have largely been<br />

rejected due to the poor design <strong>of</strong> the studies or lack <strong>of</strong> analytical


46 SIZING UP THE PROBLEM OF EXPOSURE EXTRAPOLATION<br />

precision. In the main the data have come from drugs but the same general<br />

considerations would hold for environmental chemicals.<br />

Wherever possible the only compounds included in the analysis have<br />

been those where unbound clearance after systemic administration has been<br />

reported, unless it has been shown that there are little interspecies<br />

differences in protein binding or that absorption is known to be complete<br />

in all the animals. In the past these provisos have not always been met,<br />

leading to incorrect interpretation <strong>of</strong> the data. In most reports the<br />

allometric scaling has used results from at least four species but in some<br />

cases up to 11 have been included. Practically this would involve an<br />

enormous resource and would be difficult when many compounds are<br />

being investigated. For this analysis it has been assumed that only one<br />

species will initially be used and the aim <strong>of</strong> this analysis was to find which<br />

single species would provide the best prediction <strong>of</strong> clearance compared to<br />

that found in man.<br />

Three methods have been used using data, wherever possible, from<br />

mouse, rat, rabbit, dog and monkey (macaques) in a total <strong>of</strong> 60<br />

compounds, with human unbound clearances ranging from 4 to 150 909 ml<br />

min −1 .<br />

Simple allometric equation<br />

Figure 4.1 shows a typical allometric relationship for the clearance <strong>of</strong> the<br />

anticancer drug, fotemustine, showing that equation (4.1) can be made<br />

linear for the determination <strong>of</strong> the variables by logarithmically<br />

transforming the body weight (W) and clearances (CL), as shown in<br />

equation (4.2) where the exponent b can be calculated from the slope <strong>of</strong><br />

the linear regression.<br />

(4.2)<br />

From this analysis <strong>of</strong> all the available papers, where this has been<br />

undertaken with more than four species using data taken from 29<br />

compounds, it was possible to show that the mean exponent (b) is<br />

approximately 0.70±0.15 for unbound clearance, but with a range <strong>of</strong> 0.92–<br />

0.28. This mean value is to be expected since it is comparable to the<br />

exponent for the allometric equation relating physiological rates and<br />

clearances to weight as for metabolic rate, body surface area, hepatic and<br />

renal blood flow, etc. Therefore it would seem that even without a specific<br />

knowledge <strong>of</strong> the clearance in a number <strong>of</strong> different species, it could be<br />

assumed that the exponent <strong>of</strong> 0.7 is a common factor for all chemicals, if it<br />

has not been previously determined. The coefficient a can subsequently be<br />

determined for each compound from only one species according to<br />

equation (4.1), and a predictive value for man determined.


Body surface area (BSA)<br />

It has been suggested that the body surface area provides a good measure<br />

<strong>of</strong> overall metabolic rate and that this may be a better measure <strong>of</strong> relative<br />

clearance between species (Chiou and Hsu, 1988). The BSA has therefore<br />

been cal culated for each species using Meehs Formula, BSA=0.103·W 0.67<br />

(Spector, 1956) and the ratio <strong>of</strong> human BSA to animal BSA multiplied by<br />

the animal clearance, to determine the predicted human clearance.<br />

Life span correction<br />

D.BRUCE CAMPBELL 47<br />

Figure 4.1. Allometric scaling <strong>of</strong> Fotemustine clearance compared with the body<br />

weight in various species.<br />

For some drugs, particularly those which are extensively metabolised but<br />

have a low hepatic clearance, such as phenytoin, antipyrine or caffeine<br />

(Boxenbaum, 1982b; Bonati et al., 1984–5), these simple scaling methods<br />

seem to be poorly predictive for man and an allometric correction using<br />

maximum life potential (MLP) has been used to improve the accuracy<br />

(Figure 4.2). Although the allometric approach using body weight alone is


48 SIZING UP THE PROBLEM OF EXPOSURE EXTRAPOLATION<br />

Figure 4.2. Comparison <strong>of</strong> the allometric interspecies scaling for phencyclidine<br />

using: (top) clearance (CL), and (bottom) clearance corrected for maximum life<br />

potential (MLP) in seven species (redrawn from Owens et al., 1987).<br />

valid for many physiological functions it is poorly predictive <strong>of</strong> longevity<br />

or maximum life potential in man. Using a derived equation based on body<br />

weight alone, humans should only live for 26.6 years, clearly an<br />

underestimate. In fact Sacher (1959) has shown that a better measurement<br />

<strong>of</strong> life span can be calculated using not only body weight but also brain<br />

weight (equation (4.3)), and with this correction the MLP for man<br />

becomes 113 years (Boxenbaum and De Souza, 1988).


(4.3)<br />

Simplistically it has been suggested that these differences in longevity can<br />

be explained by the assumption that in any one species there is a<br />

predetermined or fixed amount <strong>of</strong> total ‘body metabolic potential’ and<br />

once this is used up the animal dies (Boddington, 1978). Boxenbaum (1986)<br />

has extrapolated this concept to include intrinsic hepatic metabolism<br />

suggesting that there is a certain quantity <strong>of</strong> ‘hepatic pharmacokinetic<br />

stuff’ per unit <strong>of</strong> body weight available in a life-time which can be<br />

interrelated by the formula:<br />

(4.4)<br />

where CL is the unbound clearance, and c is a constant for each compound.<br />

Thus, the longer the animal lives, the slower this ‘stuff’ is used up.<br />

Examination <strong>of</strong> the data available from 13 disparate compounds<br />

(Table 4.1), where at least four species have been investigated, shows the<br />

MLP correction has produced good results with an exponent b equal to<br />

unity. Thus this would suggest that the relative clearance between species is<br />

directly proportional to their body weight (W) and MLP, and that animal<br />

(CL (A)) and human clearance (CL (H)) can be simply related according to<br />

equation (4.4).<br />

(4.5)<br />

The maximum life potential (MLP) has been calculated for each animal<br />

from Sacher’s formula (equation (4.3)) (mouse=2.7 y, rat=4.7 y, dog=20 y,<br />

rabbit=8 y, monkey=22 y and human=113 y).<br />

For each drug where the appropriate information was available, the<br />

human clearance has been calculated from each species using the above<br />

approaches and compared with that observed (Table 4.2), and the<br />

percentage prediction measured as:<br />

Results<br />

D.BRUCE CAMPBELL 49<br />

The data from 60 different compounds were used in this ongoing analysis<br />

and as could be expected more data were available for the rat (n=47)<br />

compared to mouse (n=27) and dog (n=28), rabbit (n=24), or monkeys<br />

(n=17). In four cases, valproic acid, diazepam, ceftizoxime and<br />

theophylline, different results were found and data have been analysed<br />

separately. For two classes <strong>of</strong> drugs, β-lactams and benzodiazepines, data<br />

from a number <strong>of</strong> compounds were available (n=6 and 12, respectively), but<br />

only mean values were used in this analysis to minimise a class <strong>of</strong><br />

compounds bias in the results.


50 SIZING UP THE PROBLEM OF EXPOSURE EXTRAPOLATION<br />

Table 4.1 Comparison <strong>of</strong> exponential values for b with MLP corrected clearance<br />

(CL u ·MLP=aW b )<br />

From Figure 4.3 it can be seen that for most species the use <strong>of</strong> the simple<br />

exponent 0.7 provided the worst prediction, particularly in the mouse and<br />

dog, which overestimated the human clearance by approximately 600 and<br />

400 per cent, respectively. The rat and rabbit (100–150 per cent) were<br />

better but the monkey was best giving a small overestimate (36 per cent). The<br />

body surface area calculation for most animals gave a better result<br />

particularly for the rat (48 per cent) and monkey (−28 per cent), but the<br />

best method overall is the use <strong>of</strong> the maximum life potential correction<br />

which provided reasonable predictions, within 50 per cent, for all species<br />

with the exception <strong>of</strong> the mouse (89 per cent). The mean accuracy values<br />

only provide part <strong>of</strong> the picture on predictions and the variation, range and<br />

outliers can give additional information on precision and confidence <strong>of</strong> the<br />

analyses. Table 4.3 shows that although there is reasonable accuracy with<br />

the rat, rabbit and dog, the coefficients <strong>of</strong> variations and range <strong>of</strong> values<br />

for these species are large, particularly in the dog, even though the mean<br />

value is reasonable. However for the monkey most estimates <strong>of</strong> human<br />

clearance fall within close proximity to the mean provid ing good<br />

confidence in the data. Similarly the number <strong>of</strong> all compounds which have<br />

a predictability <strong>of</strong> more than 100 per cent error was large for the dog (18 per<br />

cent) and mouse (11 per cent), less for the rat and rabbit, but none were<br />

found for the monkey. In the rat, where the largest number <strong>of</strong> compounds<br />

were examined (n=56), there is a good correlation (r=0.81, p


Figure 4.3. Mean prediction values (percentage error) for human clearance<br />

calculated for various species using: exponent 0.7, body surface area (BSA), and<br />

maximum life potential correction (MLP).<br />

D.BRUCE CAMPBELL 51<br />

For these life span corrections, equation (4.3) has been used to calculate<br />

MLP, but monkeys in captivity, in contract organisations and zoos<br />

(Carmac, 1994), appear to live longer than the calculated 22 years and<br />

ages <strong>of</strong> 35 years are not uncommon. Substituting this longer life span into<br />

the clearance MLP correction improves the mean accuracy to −14 per cent,<br />

but the range increases and 2 per cent <strong>of</strong> compounds now give a prediction<br />

greater than 100 per cent. Attempts to combine predictions from two or<br />

more animals did not improve the accuracy <strong>of</strong> the predictions but did<br />

marginally improve the confidence <strong>of</strong> these values, particularly when the<br />

data from rat and monkey were averaged, from a confidence interval <strong>of</strong><br />

±20 and ±23 for rat and monkey respectively, when used alone, to ±15<br />

when the results were combined.<br />

From this analysis <strong>of</strong> the data it would appear that measurement <strong>of</strong> the<br />

clearance <strong>of</strong> a drug in the monkey together with a correction for MLP<br />

differences, provide the best overall estimate <strong>of</strong> human clearance with the<br />

greatest confidence in the results, although for many compounds the rat or<br />

even the rabbit are good alternatives. The mouse and the dog, on the other<br />

hand, seem to be poorer animal models to extrapolate to human kinetics.<br />

Discussion<br />

There has in the past been a hesitation to use allometric scaling to predict<br />

the clearance in man, but it would appear from this review <strong>of</strong> the literature<br />

that this approach can be used for predictive purposes with an acceptable<br />

degree <strong>of</strong> accuracy, even when the clearance is measured in only one<br />

species. To put this in perspective, if the actual human clearance was 500 ml<br />

min−1 , the predicted clearance using rat or monkey with MLP correction<br />

would be a oximately 300 ml min 1 ppr<br />

− with a 95 per cent confidence,


52 SIZING UP THE PROBLEM OF EXPOSURE EXTRAPOLATION<br />

Table 4.2 Human unbound clearances <strong>of</strong> the compounds used in this analysis


D.BRUCE CAMPBELL 53<br />

a Campbell DB, 1993 unpublished data.<br />

b CL=812 ml min−1


54 SIZING UP THE PROBLEM OF EXPOSURE EXTRAPOLATION<br />

Table 4.3 Interspecies comparisons <strong>of</strong> human clearance predictions expressed as<br />

percentage from observed clearances using maximum life potential corrections<br />

a MLP=22 years.<br />

b MLP=35 years.<br />

c Percentage <strong>of</strong> compounds with a predicted human clearance greater than 100% <strong>of</strong><br />

that observed.<br />

Figure 4.4. Relationship between observed human clearance and that calculated<br />

from the rat using a maximum life potential (MLP) correction (n=56) (—— line <strong>of</strong><br />

identity).<br />

ranging from 240 to 360 ml min− 1.<br />

The monkey appears to be slightly<br />

better than the rat and rabbit in terms <strong>of</strong> the accuracy <strong>of</strong> prediction, and<br />

with a few exceptions may be phylogenetically more acceptable. This is<br />

perhaps not surprising since most studies which have examined species<br />

differences in metabolism indicate that the monkey is more similar to man<br />

compared to the rat (Caldwell, 1981). All the primate data reported, as far<br />

as can be ascertained, have come from the Rhesus or Cynomolgus, Old<br />

World Macaque monkeys. The same considerations may not be true for<br />

New World monkeys, such as the squirrel or marmoset, but few kinetic<br />

comparisons have been made with these species.<br />

In practice, prediction <strong>of</strong> human clearance would involve measuring the<br />

intravenous or intramuscular kinetics, namely the infinite area under the<br />

curve, for each investigatory compound in two to four animals, together<br />

with an estimate <strong>of</strong> the in vitro protein binding in the animal under<br />

investigation and in human plasma, to obtain the free intrinsic clearance


and then multiply the animal clearance by the ratio <strong>of</strong> weight and MLP,<br />

approximately 13 for the rat and 3.5 for a macaque monkey. Of course, as<br />

shown by these data, there can be exceptions, and the monkey and indeed<br />

the rat may not be a suitable species to undertake allometric scaling for all<br />

compounds. However there is an increasing use <strong>of</strong> in vitro systems such as<br />

isolated microsomes, hepatocytes or hepatic slices, to compare the<br />

metabolic pr<strong>of</strong>iles <strong>of</strong> compounds in animals. If undertaken in conjunction<br />

with allometric scaling, pr<strong>of</strong>ound interspecies differences in the rates and<br />

extent <strong>of</strong> metabolism compared to humans could be observed and provide<br />

information on which is the most suitable species to use for scaling. Since<br />

the allometric scaling for volume appears for most compounds to be<br />

directly proportional to body weight with an exponent <strong>of</strong> approximately 1.<br />

0, half-life can also be easily calculated thereby providing all the necessary<br />

kinetic parameters to simulate plasma levels after repeated dosing in man.<br />

With this information the absolute need to undertake kinetic analysis <strong>of</strong><br />

industrial chemicals in volunteers would be reduced since the exposure<br />

calculated by this procedure is considerably better than that employed<br />

presently using uncertainty factors, giving errors in excess <strong>of</strong> 1000 per<br />

cent.<br />

Further studies are <strong>of</strong> course needed to confirm these initial<br />

observations, particularly with those chemicals used in industry or potential<br />

environmental pollutants, but perhaps this re-evaluation shows that<br />

allometry, when correctly used, may well have a practical role in the<br />

evaluation <strong>of</strong> their potential risk to man.<br />

References<br />

D.BRUCE CAMPBELL 55<br />

ADOLPH, E.F., 1949, Quantitative relations in the physiological constituents <strong>of</strong><br />

mammals, Science, 109, 579–85.<br />

ANDERSEN, M.E., CLEWELL, H.J.III, GARGAS, M.L., SMITH, F.A. and<br />

REITZ, R.H., 1984, Physiologically-based pharmacokinetics and the risk<br />

assessment process for methylene chloride, Toxicol. Appl. Pharmacol., 87,<br />

185–205.<br />

BÄÄRNHIELM, C., DAHLBÄCK, H. and SKǺNBERG, I., 1986, In vivo<br />

pharmacokinetics <strong>of</strong> felodipine predicted from in vitro studies in rat, dog and<br />

man, Acta Pharmacol. Toxicol, 59, 113–22.<br />

BACHMANN, K., 1989, Predicting toxicokinetic parameters in humans from<br />

toxicokinetic data acquired from three small mammalian species, J. Appl.<br />

Toxicol., 9(5), 331–8.<br />

BODDINGTON, M.J., 1978, An absolute metabolic scope for activity, J. Theor.<br />

Biol., 75, 443–9.<br />

BONATI, M., LATINI, R., TOGNONI, G., YOUNG, J.F. and GARATTINI, S.,<br />

1984–5, Interspecies comparison <strong>of</strong> in vivo caffeine pharmacokinetics in man,<br />

monkey, rabbit, rat and mouse, Drug Metab. Rev., 15(7), 1355–83.


56 SIZING UP THE PROBLEM OF EXPOSURE EXTRAPOLATION<br />

BOXENBAUM, H., 1982a, Comparative pharmacokinetics <strong>of</strong> benzodiazepines in<br />

dog and man, J. Pharmacokin. Biopharm., 10, 417–26.<br />

BOXENBAUM, H., 1982b, Interspecies scaling, allometry, physiological time, and<br />

the ground plan <strong>of</strong> pharmacokinetics, J. Pharmacokin. Biopharm., 10(2), 201–<br />

27.<br />

BOXENBAUM, H., 1984, Interspecies pharmacokinetic scaling and the<br />

evolutionarycomparative paradigm, Drug Metab. Rev., 15(5, 6), 1071–121.<br />

BOXENBAUM, H., 1986, Time concepts in physics, biology and pharmacokinetics,<br />

J. Pharm. Sci., 75(11), 1053–62.<br />

BOXENBAUM, H. and FERTIG, J.B., 1984, Scaling <strong>of</strong> antipyrine intrinsic<br />

clearance <strong>of</strong> unbound drug in 15 mammalian species, Europ. J. Drug Metab.<br />

Pharmacokin., 9(2), 177–83.<br />

BOXENBAUM, H. and DE SOUZA, R., 1988, Physiological models, allometry,<br />

neoteny, space time and pharmacokinetics, in Peale, A. and Resagno, A. (eds)<br />

Pharmacokinetics: Mathematical and Statistical Approaches, New York:<br />

Plenum Publishing.<br />

CALABRESE, E.J., 1983, Principles <strong>of</strong> Animal Extrapolation, New York: John<br />

Wiley.<br />

CALDWELL, J., 1981, The current status <strong>of</strong> attempts to predict species differences<br />

in drug metabolism, Drug Metab. Rev., 12, 221–37.<br />

CAMPBELL, D.B. and INGS, R.M.J., 1988, New approaches to the use <strong>of</strong><br />

pharmacokinetics in toxicology and drug development, Human Toxicol, 7,<br />

469–79.<br />

CARMAC, M., 1994, Personal communication, London Zoo: Head Keeper <strong>of</strong><br />

Primates.<br />

CHAPPELL, W.R. and MORDENTI, J., 1991, Extrapolation <strong>of</strong> toxicological and<br />

pharmacological data from animals to humans, Adv. Drug Res., 20, 1–116.<br />

CHIOU, W.L. and Hsu, F-H. 1988, Correlation <strong>of</strong> unbound plasma clearances <strong>of</strong><br />

fifteen extensively metabolized drugs between humans and rats, Pharm. Res., 5<br />

(10), 668–72.<br />

CHUNG, M., RADWANSKI, E., LOEBENBERG, D., LIN, C-C., ODEN, E.,<br />

SYMCHOWICZ, S., GURAL, R.P. and MILLER, G.H., 1985, Interspecies<br />

pharmacokinetic scaling <strong>of</strong> Sch 34343, J. Antimicrob. Chemother., 15<br />

(Supp.C), 227–33.<br />

CRUMP, K.S., 1984, A new method for determining available daily intakes,<br />

Fundam. Appl. Toxicol., 4, 854–71.<br />

DEDRICK, R.L., 1973, Animal scale-up, J. Pharmacokin. Biopharm., 1(5), 435–61.<br />

DOURSON, M.L. and STARA, J.F., 1983, Regulatory history and experimental<br />

support <strong>of</strong> uncertainty (safety) factors, Regul. Toxicol. Pharmacol, 3, 224–38.<br />

DUTHU, G.S., 1985, Interspecies correlation <strong>of</strong> the pharmacokinetics <strong>of</strong><br />

erythromycin, oleandomycin and tylosin. J. Pharm. Sci., 74(9), 943–6.<br />

GASPARI, P. and BONATI, M., 1990, Interspecies metabolism and<br />

pharmacokinetic scaling <strong>of</strong> theophylline disposition, Drug Metab. Rev., 22(2,<br />

3), 179–207.<br />

HINDERLING, P.H., DILEA, C., KOZIOL, T. and MILLINGTON, G., 1993,<br />

Comparative kinetics <strong>of</strong> sematilide in four species, Drug Metab. Dispos., 21(4),<br />

662–9.


D.BRUCE CAMPBELL 57<br />

IBRAHIM, S.S. and BOUDINOT, F.D., 1988, Pharmacokinetics <strong>of</strong> 2′,3′dideoxycytidine<br />

in rats: application to interspecies scale-up, J. Pharm.<br />

Pharmacol, 41, 829–34.<br />

KLOTZ, U., ANTONIN, K.H. and BIECK, P.R., 1976, Pharmacokinetics and<br />

plasma binding <strong>of</strong> diazepam in man, dog, rabbit, guinea pig and rat, J. Pharm.<br />

Exper. Therap., 199(1), 67–73.<br />

KURIHARA, A., NAGANUMA, H., HISAOKA, M., TOKIWA, H. and<br />

KAWAHARA, Y., 1992, Prediction <strong>of</strong> human pharmacokinetics <strong>of</strong> panipenembetamipron,<br />

a new carbapenem, from animal data, Antimicrob. Agents<br />

Chemother., 36(9), 1810–16.<br />

LAPKA, R., REJHOLEC, V., SECHSER, T., PETERKOVA, M. and SMID, M.<br />

1989, Interspecies pharmacokinetic scaling <strong>of</strong> metazosin, a novel alphaadrenergic<br />

antagonist, Biopharm. Drug Dispos., 10, 581–9.<br />

LEBEL, M., PAONE, R.P. and LEWIS, G.P., 1983, Effect <strong>of</strong> probenecid on the<br />

pharmacokinetics <strong>of</strong> ceftizoxime, J. Antimicrob. Chemother., 12, 147–55.<br />

LINDSTEDT, S.L. and CALDER, W.A., 1981, Body size, physiological time and<br />

longevity <strong>of</strong> homeothermic animals, Quart. Rev. Biology, 56, 1–16.<br />

McGOVERN, J.P., WILLIAMS, M.G. and STEWART, J.C., 1988, Interspecies<br />

comparison <strong>of</strong> acivicin pharmacokinetics, Drug Metab. Dispos., 16(1), 18–22.<br />

MITSUHASHI, Y., SUGIYAMA, Y., OZAWA, S., NITANAI, T., SASAHARA, K.,<br />

NAKAMURA, K-I., TANAKA, M. et al. 1990, Prediction <strong>of</strong> ACNU plasma<br />

concentration-time pr<strong>of</strong>iles in humans by animal scale-up, Cancer Chemother.<br />

Pharmacol, 27, 20–6.<br />

MORDENTI, J., 1985, Forecasting cephalosporin and monobactam antibiotic halflives<br />

in humans from data collected in laboratory animals, Antimicrob. Agents<br />

Chemother., 27(6), 887–91.<br />

MORDENTI, J., 1986, Man versus beast: pharmacokinetics in mammals, J.<br />

Pharm. Sci., 75(11), 1028–40.<br />

MROSZCZAK, E.J., LEE, F.W., COMBS, D., SARNQUIST, F.H., HUANG, B-L.,<br />

WU, A.T., TOKES, L.G. et al., 1987, Ketorolac thromethamine absorption,<br />

distribution, metabolism, excretion and pharmacokinetics in animals and<br />

humans, Drug Metab.Dispos., 15(5), 618–26.<br />

MURAKAWA, T., SAKAMOTO, H., FUKADA, S., NAKAMOTO, S., HIROSE,<br />

T., ITOH, N. and NISHIDA, M., 1980, Pharmacokinetics <strong>of</strong> ceftizoxime in<br />

animals after parenteral dosing, Antimicrob. Agents Chemother., 17(2), 157–<br />

64.<br />

OWENS, S.M., HARDWICK, W.C. and BLACKALL, D., 1987, Phencyclidine<br />

pharmacokinetic scaling among species, J. Pharm. Exper. Therap., 242(1), 96–<br />

101.<br />

PAXTON, J.W., KIM, S.N. and WHITFIELD, L.R., 1990, Pharmacokinetic and<br />

toxicity scaling <strong>of</strong> the antitumor agents amsacrine and CI-921, a new analogue,<br />

in mice, rats, rabbits, dogs and humans, Cancer Res., 50, 2692–7.<br />

PETERS, H.P., 1983, The Ecological Implications <strong>of</strong> Body Size, Cambridge:<br />

Cambridge University Press.<br />

PUIGDEMONT, A., GUITART, R., DE MORA, F. and ARBOIX, M., 1991,<br />

Prediction <strong>of</strong> the disposition <strong>of</strong> propafenone in humans and dogs from<br />

pharmacokinetic parameters in other animal species, J. Pharm. Sci., 80(12),<br />

1106–9.


58 SIZING UP THE PROBLEM OF EXPOSURE EXTRAPOLATION<br />

REIGNER, B.C., BOIS, F.Y. and TOZER, T.N., 1993, Pentachlorophenol<br />

carcinogenicity: extrapolation <strong>of</strong> risk from mice to humans, Human Exp.<br />

Toxicol., 12, 215–25.<br />

RITSCHEL, W.A., VACHHARAJANI, N.N., JOHNSON, R.D. and HUSSAIN, A.<br />

S., 1991, Interspecies scaling <strong>of</strong> the pharmacokinetic parameters <strong>of</strong> coumarin<br />

among six different mammalian species, Meth. Find Exper. Clin. Pharmacol, 13<br />

(10), 697–702.<br />

SACHER, G.A., 1959, Relation <strong>of</strong> lifespan to brain weight and body weight in<br />

mammals, in Wolstenholme, G.E.W. and O’Connor, M. (Eds), The Life Span<br />

<strong>of</strong> Animals, pp. 115–41. Boston, Mass: Little Brown.<br />

SAWADA, Y., HANANO, M., SUGIYAMA, Y. and IGA, T., 1984, Prediction <strong>of</strong><br />

the disposition <strong>of</strong> β-lactam antibiotics in humans from pharmacokinetic<br />

parameters in animals, J. Pharmacokin. Biopharm., 12(3), 241–61.<br />

SAWADA, Y., HANANO, M., SUGIYAMA, Y. and IGA, T., 1985, Prediction <strong>of</strong><br />

the disposition <strong>of</strong> nine weakly acidic and six weakly basic drugs in humans<br />

from pharmacokinetic parameters in rats, J. Pharmacokin. Biopharm., 13(5),<br />

477–92.<br />

SPECTOR, W.S., 1956, Handbook <strong>of</strong> Biological Data, Philadelphia:<br />

W.B.Saunders.<br />

SWABB, E.A. and BONNER, D.P., 1983, Prediction <strong>of</strong> aztreonam<br />

pharmacokinetics in humans based on data from animals, J. Pharmacokin.<br />

Biopharm., 11(3), 215–23.<br />

TSUNEKAWA, Y., HASEGAWA, T., NADAI, M., TAKAGI, K. and NABESHIMA,<br />

T., 1992, Interspecies differences and scaling for the pharmacokinetics <strong>of</strong><br />

xanthine derivatives, J. Pharm. Pharmacol, 44, 594–9.


PART TWO<br />

Reactive industrial chemicals


5<br />

Metabolism <strong>of</strong> Reactive Chemicals<br />

PETER J.van BLADEREN 1,2 and BEN van OMMEN 1<br />

1 TNO <strong>Toxicology</strong>, Zeist<br />

2 Agricultural University, Wageningen<br />

Introduction<br />

For the purpose <strong>of</strong> the present paper, a reactive chemical will be defined as<br />

a strongly electrophilic agent. Such compounds can bind to the numerous<br />

macromolecular targets in the cell, and thus elicit toxic effects. Binding to<br />

DNA can result in mutations or cancer, binding to proteins or membrane<br />

components to cytotoxicity or specific forms <strong>of</strong> toxicity.<br />

A scale could be drawn up for the reactivity <strong>of</strong> electrophiles. However, it<br />

is not certain that those compounds on the high end <strong>of</strong> the scale, i.e. the<br />

most reactive, would also be the most toxic. On the contrary, these<br />

compounds might be expected to react quickly with water and thus not<br />

reach their target molecules. For the purpose <strong>of</strong> classifying the reactivity <strong>of</strong><br />

electrophiles, the most useful is the theory <strong>of</strong> s<strong>of</strong>t and hard acids and bases<br />

(e.g. Commandeur and Vermeulen, 1990). In principle, the preferential<br />

targets for electrophiles can be derived. Furthermore, an electrophile<br />

showing the highest affinity for the relatively hard nitrogen and oxygen<br />

nucleophiles <strong>of</strong> DNA may pose a higher risk for mutations and cancer than<br />

one reacting preferentially with s<strong>of</strong>t sulfur nucleophiles such as found in<br />

proteins and glutathione.<br />

The following classes <strong>of</strong> electrophiles will be discussed: (1) quinones,<br />

which can both arylate as well as cause toxicity through redox cycling; (2)<br />

derivatives with an actual leaving group such as methylene chloride and<br />

ethylene dibromide, and (3) reagents such as isothiocyanates, isocyanates<br />

and α,β-unsatu-rated<br />

ketones and aldehydes.<br />

The enzymes involved in activation and detoxication<br />

To become toxic, almost all <strong>of</strong> the chemicals to which man is exposed,<br />

including the carcinogens, need metabolic activation. The reactive<br />

intermediates that are formed during metabolism are responsible for<br />

binding to cellular macromolecules which very likely elicit the toxic


P.J.VAN BLADEREN AND B.VAN OMMEN 61<br />

response. In general, other biotransformation enzymes can detoxify these<br />

metabolites. Thus, the concentration <strong>of</strong> the ultimate carcinogen, or<br />

toxicant in general, is the result <strong>of</strong> a delicate balance between the rate <strong>of</strong><br />

activation and the rate <strong>of</strong> detoxification. Although toxicological processes<br />

can be much more complex, interindividual differences in susceptibility are<br />

certainly also a result <strong>of</strong> interindividual differences in this balance between<br />

metabolic activation and detoxification.<br />

The enzymes which are to a large extent responsible for the formation <strong>of</strong><br />

reactive metabolites belong to the family <strong>of</strong> cytochromes P-450. However,<br />

for almost all enzymes involved in biotransformation, examples have been<br />

described <strong>of</strong> activation <strong>of</strong> specific classes <strong>of</strong> chemicals. The main classes <strong>of</strong><br />

enzymes involved in detoxifying chemicals which are reactive per se as well<br />

as reactive metabolites are the epoxide hydrolases and the glutathione Stransferases.<br />

NADPH quinone reductase is involved in the reduction <strong>of</strong><br />

quinones.<br />

Epoxide hydrolases<br />

Metabolites which contain an epoxide moiety may undergo hydrolytic<br />

cleavage to less reactive vicinal dihydrodiols. This reaction is catalyzed by<br />

the enzyme epoxide hydrolase (EH), which was first thought to be<br />

exclusively located in the endoplasmic reticulum (microsomal epoxide<br />

hydrolase, mEH; Oesch, 1972). In later studies on the mammalian<br />

metabolism <strong>of</strong> certain alkyl epoxides, the existence <strong>of</strong> a cytosolic EH (cEH)<br />

was demonstrated (Gill et al., 1974). The two forms <strong>of</strong> EH have<br />

complementary substrate specificity, in that many epoxides, e.g. arene<br />

oxides, which are good substrates for mEH are poor substrates for cEH,<br />

and vice versa, e.g. trans-disubstituted oxiranes are good substrates for cEH<br />

but not for mEH (Hammock and Hasagawa, 1983). Other studies have<br />

pointed to the fact that the common nomenclature <strong>of</strong> ‘microsomal’ and<br />

‘cytosolic’ epoxide hydrolase is not semantically precise: metabolic and<br />

immunochemical studies demonstrated the existence <strong>of</strong> membrane-bound<br />

forms <strong>of</strong> cEH (Guenthner and Oesch, 1983), whereas mEH-like activity<br />

was detected in cytosolic fractions <strong>of</strong> human tissue (Schladt et al., 1988).<br />

Glutathione S-transferases<br />

Glutathione is involved in a variety <strong>of</strong> vital cellular reactions. First, a large<br />

number <strong>of</strong> the various classes <strong>of</strong> xenobiotics to which man is exposed—<br />

industrial, therapeutic as well as naturally occurring chemicals—are<br />

metabolized in vivo to reactive intermediates. Such electrophilic<br />

metabolites may bind to cellular macromolecules and thus cause toxicity.<br />

The formation <strong>of</strong> glutathione conjugates, both by spontaneous reaction<br />

between the reactive species and glutathione as well as catalyzed by the


62 METABOLISM OF REACTIVE CHEMICALS<br />

glutathione S-transferases, is the main detoxification mechanism for<br />

electrophiles in mammalian cells (Chasseaud, 1979). Secondly, via<br />

glutathione peroxidase and the glutathione S-transferases, hydrogen<br />

peroxide and organic peroxides are detoxified, yielding glutathione disulfide<br />

as one <strong>of</strong> the products (Prohaska, 1980). Thirdly, glutathione and the<br />

glutathione S-transferases play a role in the biosynthesis <strong>of</strong> such important<br />

endogenous compounds as prostaglandins and leukotriene C4 (Söderstrom<br />

et al., 1985; Ujihara et al., 1988). In fact, in the latter case one may argue<br />

that an endogenous compound is activated by conjugation with<br />

glutathione, since leukotriene C4 is a mediator <strong>of</strong> the adverse reactions<br />

associated with asthmatic attacks (Samuelson, 1988).<br />

The GSTs are a family <strong>of</strong> isoenzymes with broad and overlapping<br />

substrate selectivity. Although membrane-bound forms <strong>of</strong> GST have been<br />

detected (Morgenstern et al., 1988), GST activity is mainly located in the<br />

cytosol. GSTs are dimers <strong>of</strong> subunits and within a dimer, each subunit<br />

functions independently <strong>of</strong> the other (Mannervik and Jensson, 1982). The<br />

GSTs are now known to be a multi-gene family <strong>of</strong> isoenzymes, which can<br />

be divided into four classes (alpha, mu, pi and theta), based on similarity in<br />

structural, physical and catalytic properties <strong>of</strong> their subunits (Ketterer and<br />

Mulder, 1990; Vos and Van Bladeren, 1990). In addition to their crucial role<br />

in catalyzing glutathione conjugation, GSTs may also be important in<br />

intracellular binding and/or transport <strong>of</strong> endogenous and xenobiotic nonsubstrate<br />

ligands (Listowsky et al., 1988).<br />

The glutathione conjugates initially formed from electrophilic species are<br />

further processed via -glutamyltranspeptidase which splits <strong>of</strong>f the<br />

glutamate residue, and dipeptidases which remove the glycine moiety. The<br />

resultant cysteine S-conjugates are then acetylated to give so-called<br />

mercapturic acids which are excreted into the urine (Jakoby, 1980).<br />

Interestingly, mercapturic acids were the first metabolites derived from<br />

xenobiotics to be recognized as such (Baumann and Preusse, 1879).<br />

In recent years it has become increasingly evident that glutathione<br />

conjugation is also involved in the formation <strong>of</strong> toxic metabolites from a<br />

variety <strong>of</strong> chemicals (Monks et al., 1990b). These metabolites display a<br />

wide spectrum <strong>of</strong> toxic effects, ranging from cytotoxicity to genotoxicity.<br />

The various mechanisms elucidated for the toxic action <strong>of</strong> the conjugates<br />

can be grouped as follows: (1) directly toxic glutathione conjugates may be<br />

formed from vicinal and geminal dihaloalkanes, via the formation <strong>of</strong> sulfur<br />

halfmustards; (2) from several types <strong>of</strong> glutathione conjugates active<br />

metabolites may be formed by further metabolic steps: conjugates <strong>of</strong><br />

hydroquinones can be oxidized to give reactive quinones, and conjugates<br />

derived from haloalkenes are transformed into electrophilic species by the<br />

action <strong>of</strong> cysteine conjugate β-lyase. For both hydroquinones and<br />

haloalkenes the selective nephrotoxicity observed is the result <strong>of</strong> the<br />

targeting <strong>of</strong> the conjugates to the kidneys; (3) glutathione conjugates may


P.J.VAN BLADEREN AND B.VAN OMMEN 63<br />

serve as transporting and targeting agents for compounds that react<br />

reversibly with gluathione such as isothiocyanates, isocyanates and α,<br />

βunsaturated<br />

ketones (Van Bladeren, 1988).<br />

Glutathione S-transferase polymorphism<br />

Genetic variation in the expression <strong>of</strong> GST isoenzymes has been studied<br />

almost solely in man. Considerable variation, possibly indicating a<br />

polymorphism, has been observed for the human liver alpha class<br />

isoenzymes. The ratio <strong>of</strong> GSTA1 and GSTA2 subunits, as determined by<br />

HPLC, was found to range from 0.5 to over 10 (Van Ommen et al., 1990).<br />

However, a division into two groups, with average ratios <strong>of</strong> 1.6±0.3 and 3.<br />

8±0.6 could be made, suggesting an alpha class polymorphism. In view <strong>of</strong><br />

the fact that subunits GSTA1 and GSTA2 together make up a major<br />

portion <strong>of</strong> the GST protein in human liver this potential polymorphism<br />

merits further attention. For class mu isoenzymes a clear polymorphism<br />

has been observed in humans: iso-enzyme GSTM1a-1a was found to be<br />

expressed in only 60% <strong>of</strong> the samples analyzed (Board, 1981). In this study<br />

no account was taken <strong>of</strong> the fact that a second mu class isoenzyme,<br />

isoenzyme GSTM1b-1b was also suggested to play a part in this<br />

polymorphism. In a study on the excretion <strong>of</strong> the mercapturate derived<br />

from 1,3-dichloropropene in exposed workers, however, no difference was<br />

observed between mu-positive and mu negative subjects (Vos et al., 1991).<br />

Quinones and their glutathione conjugates<br />

Two modes <strong>of</strong> reactivity can form the basis <strong>of</strong> the toxicity associated with<br />

quinones: (i) their ability to undergo ‘redox cycling’ and to thereby create<br />

an oxidative stress (Kulkarni et al., 1978), and (ii) their electrophilicity<br />

allowing them to react directly with cellular nucleophiles such as protein<br />

and non-protein sulfhydryls (Dierickx, 1983). Since glutathione is the<br />

major non-protein sulfhydryl present in cells, it comes as no surprise that it<br />

is intimately involved in the biological effects <strong>of</strong> quinones. On the one<br />

hand, glutathione can act as a reducing agent, detoxifying quinones by<br />

converting them to hydroquinones with the concomitant formation <strong>of</strong><br />

glutathione disulfide. On the other hand quinone and hydroquinonethioethers<br />

are formed. Recently considerable evidence has been gathered,<br />

indicating that a variety <strong>of</strong> these thioethers possess biological activity<br />

(Dierickx, 1983; Koga et al., 1986).<br />

The target sites for the biological (toxicological) activity <strong>of</strong><br />

quinonethioethers is to a large extent determined by the glutathione moiety:<br />

as will be discussed, the main targets are the kidney (Monks et al., 1985)<br />

and various enzymes using glutathione as a (second) substrate, e.g. the<br />

glutathione S-transferases (Van Ommen et al., 1988). Bromobenzene is


64 METABOLISM OF REACTIVE CHEMICALS<br />

toxic to proximal renal tubules. The nephrotoxic effect <strong>of</strong> o-bromophenol<br />

and bromo hydroquinone was found to be considerably higher, indicating<br />

that these compounds were situated along the main bioactivation route<br />

(Monks et al., 1985). Subsequent elegant work by Monks and Lau has<br />

shown that in fact the nephrotoxicity is caused by the glutathione<br />

derivatives <strong>of</strong> bromohydroquinone (Lau and Monks, 1990). Interestingly,<br />

the relative toxicity <strong>of</strong> the quinoneglutathione conjugates increases as the<br />

extent <strong>of</strong> glutathione addition increases, i.e. the diglutathionyl derivative is<br />

more toxic than the monoconjugate (Monks et al., 1988b). The tissue<br />

selectivity is a consequence <strong>of</strong> their targeting to renal proximal tubule cells<br />

by the brushborder -glutamyl transpeptidase. AT-125, a selective inhibitor<br />

<strong>of</strong> this enzyme in vivo, protects the kidney from the toxic effects <strong>of</strong> the<br />

conjugates. The toxicity <strong>of</strong> these hydroquinone conjugates is apparently<br />

not mediated by cysteine conjugate β-lyase catalyzed formation <strong>of</strong> thiols.<br />

The inhibitor <strong>of</strong> the lyase, amino-oxyacetic acid, had only minor effects on<br />

the extent <strong>of</strong> toxicity, and the putative product, 6-bromo-2,5dihydroxythiophenol,<br />

needed activation by oxidation before it exerted any<br />

biological effect (Monks et al., 1990b). Thus, the effects <strong>of</strong> these<br />

conjugates apparently are a consequence <strong>of</strong> their oxidation to the<br />

corresponding quinones.<br />

Several isomers <strong>of</strong> 2-bromo-glutathionyl as well as the<br />

bromodiglutathionyl hydroquinones were isolated and tested. Instead <strong>of</strong> a<br />

direct correlation <strong>of</strong> toxicity with the electrochemical properties <strong>of</strong> these<br />

compounds, it was found that the diglutathionyl derivative, which is by far<br />

the most toxic, was the most stable to oxidation at pH 7.4 (Monks and<br />

Lau, 1990). The paradox was clarified by Monks and Lau by determining<br />

the oxidation potentials <strong>of</strong> the breakdown products for the mercapturic<br />

acid pathway: hydrolysis <strong>of</strong> the glutathione moiety gives rise to the cysteine<br />

derivative, which is more readily oxidized than the parent compound<br />

(Monks and Lau, 1990). Apparently two detoxication pathways are<br />

possible for these cysteine derivatives: N-acetylation results in formation <strong>of</strong><br />

the mercapturic acid which again is relatively resistant to oxidation, but<br />

oxidative cyclization <strong>of</strong> cysteinylglycine and cysteine derivatives has been<br />

found to give 1,4-benzothiazines, which do not possess any apparent toxic<br />

properties (Monks and Lau, 1990). The action <strong>of</strong> -glutamyl<br />

transpeptidase can thus result in both activation as found for 2bromohydroquinone<br />

derivatives, but also in detoxication as was observed<br />

for 2,5-dichloro-3-(glutathion-S-yl)hydroquinone and 2,5,6-trichloro-3glutathion-S-yl)hydroquinone<br />

(Mertens et al., 1991). The ease with which<br />

the 1,4-benzothiazines are formed is very likely the determining factor in this<br />

case. A similar pathway has been worked out for p-aminophenol, a known<br />

nephrotoxic metabolite <strong>of</strong> acetaminophen (Eckert et al., 1989, 1990).<br />

Bioactivation <strong>of</strong> halogenated benzenes has long been thought to be the<br />

result <strong>of</strong> oxidation to an epoxide. However, recent studies have shown that


P.J.VAN BLADEREN AND B.VAN OMMEN 65<br />

the covalent binding to cellular macromolecules is not only the result <strong>of</strong> the<br />

first oxidative step, but also <strong>of</strong> the second, the formation <strong>of</strong> a quinone or<br />

hydroquinone from the initially formed phenol. The quinone in turn can be<br />

detoxified by glutathione conjugation. However, although glutathione<br />

protects the liver against toxicity due to these quinones, the conjugates are<br />

transported to the kidney and are there activated to new reactive<br />

intermediates. Thus, increasing the relative amount <strong>of</strong> glutathione Stransferases<br />

in this case would not really protect the organism, but merely<br />

change the target organ <strong>of</strong> the active metabolites.<br />

Chemicals with a leaving group<br />

Methylene chloride<br />

Both vicinal and geminal haloalkanes are bioactivated via conjugation with<br />

glutathione. The glutathione-dependent metabolism <strong>of</strong> the important<br />

industrial solvent dichloromethane yields S-chloromethyl-glutathione as the<br />

initial metabolite (Ahmed and Anders, 1976). This intermediate is held<br />

responsible for the carcinogenicity <strong>of</strong> dichloromethane in the mouse.<br />

Interestingly, this compound does not cause tumors in rats, and this has<br />

been related to the fact that the rate <strong>of</strong> metabolism via the glutathione<br />

pathway, catalyzed by the glutathione S-transferases, is much lower in rat<br />

tissue than in mouse tissue. Man has been postulated to resemble the rat in<br />

this respect and is thus presumably safe from the carcinogenic effects <strong>of</strong><br />

methylene chloride (ECETOC, 1988). When it does not react with cellular<br />

macromolecules, the intermediate S-chloromethyl-glutathione is converted<br />

non-enzymatically to S-hydroxymethyl-glutathione, which easily eliminates<br />

formaldehyde and regenerates glutathione (Ahmed and Anders, 1978).<br />

The glutathione S-transferase isoenzyme involved in the formation <strong>of</strong> Schloromethylglutathione<br />

belongs to class theta. Interestingly, a<br />

considerable amount <strong>of</strong> interindividual variation could be observed in a<br />

group <strong>of</strong> 22 individuals (Bogaards et al., 1993).<br />

1,2-Dibromoethane and 1,2-dichloroethane<br />

The vicinal dihaloalkanes are exemplified by 1,2-dibromoethane and 1,2dichloroethane,<br />

which are mutagenic, carcinogenic as well as nephrotoxic<br />

(Van Bladeren et al., 1980; Wong et al. 1982; Guengerich et al., 1984;<br />

Elfarra and Anders, 1985; Cheever et al., 1990). The metabolism <strong>of</strong> these<br />

compounds involves two pathways, cytochrome P-450 dependent<br />

oxidation and glutathione S-transferase catalyzed formation <strong>of</strong> glutathione<br />

conjugates. The oxidative pathway results in chloro- and<br />

bromoacetaldehyde, respectively. These aldehydes are electrophilic and


66 METABOLISM OF REACTIVE CHEMICALS<br />

thought to be responsible for the covalent binding <strong>of</strong> 1,2-dichloro- and 1,2dibromoethane<br />

metabolites to protein (Inskeep and Guengerich, 1984).<br />

Glutathione conjugation results in the formation <strong>of</strong> S-2haloethylglutathione<br />

derivatives, which are sulfur halfmustards and as such<br />

highly reactive metabolites (Jean and Reed, 1989). The formation <strong>of</strong> these<br />

conjugates is catalyzed by the glutathione S-transferases, and both in rat<br />

and man the alpha-class isoenzymes have been found to be the most<br />

efficient in this catalysis (Cmarik et al., 1990).<br />

The glutathione pathway is responsible for the mutagenicity (Van<br />

Bladeren et al., 1980), the DNA-binding (Koga et al., 1986) as well as very<br />

likely the carcinogenicity (Cheever et al., 1990) <strong>of</strong> 1,2-dichloro- and 1,2dibromoethane.<br />

The S-2-haloethylglutathione derivatives are strong<br />

alkylating agents (e.g. Jean and Reed, 1989). Their electrophilicity is<br />

attributable to neighboring-group assistance. The halogen atom is displaced<br />

by the sulfur atom on the next carbon atom, to form a highly reactive<br />

episulfonium ion. The intermediacy <strong>of</strong> this reactive species is supported by<br />

stereochemical studies as well as NMR data (Van Bladeren et al., 1979;<br />

Dohn and Casida, 1987; Peterson et al., 1988).<br />

The relative importance <strong>of</strong> the oxidative and glutathione-dependent<br />

pathway in vivo is difficult to determine, since both pathways give rise to<br />

the formation <strong>of</strong> the same 2-hydroxyethylmercapturate. Using<br />

tetradeutero-1,2-dibromoethane, the ratio <strong>of</strong> the pathways has been<br />

calculated as 4:1 (Van Bladeren et al., 1981b). However, isotope effects<br />

might have a considerable influence on this ratio (White et al., 1983).<br />

The major DNA-adduct derived from 1,2-dibromoethane has been<br />

identified by Guengerich and coworkers to be S-(2-(N7-guanyl)-ethyl)<br />

glutathione (Ozawa and Guengerich, 1983; Koga et al., 1986). In addition,<br />

the structure <strong>of</strong> one <strong>of</strong> several minor adducts was recently found to be S-(2-<br />

(Nl-adenyl)ethyl) glutathione (Dong-Hyun et al., 1990). A series <strong>of</strong> S-2haloethylglutathione<br />

and -cysteine derivatives has been synthesized: all<br />

were found to react with DNA, specifically with guanine residues. As<br />

expected for a mechanism known to involve an intermediate episulfonium<br />

ion, adduct levels were similar for chloro- and bromo-substituted<br />

derivatives. However, in Salmonella typhimurium TA100 a large variation<br />

was observed in the ratio <strong>of</strong> mutations <strong>of</strong> adducts, indicating that the<br />

structure <strong>of</strong> the adduct has a major influence on the mutagenicity<br />

(Humphreys et al., 1990).<br />

Not all vicinal dihaloalkanes seem to give rise to the formation <strong>of</strong><br />

episulfonium ions. Methyl substitution for instance effectively hinders the<br />

mutagenicity through this pathway (Van Bladeren, et al., 1981a) and<br />

studies on 1,2-dibromopropane (Zoetemelk et al., 1986) and hexadeuterol,2-dichloro-propane<br />

(Bartels and Timchalk, 1990) indicate that the<br />

resulting mercapturic acids are only formed through an oxidative pathway.<br />

However, for the heavily used agricultural chemical l,2-dibromo-3-


P.J.VAN BLADEREN AND B.VAN OMMEN 67<br />

chloropropane evidence has been accumulating recently, implicating a<br />

glutathione-mediated activation pathway in the renal and testicular toxicity<br />

associated with this compound (Pearson et al., 1990). Interestingly,<br />

consecutive formation <strong>of</strong> two episulfonium ions can occur, and in fact bis-<br />

DNA-adducts have been identified (Humphreys et al., 1991). 1,2-<br />

Dibromochloropropane could thus cross-link DNA strands as the initial<br />

step leading to cell death.<br />

Isoenzyme selectivity for both primary reactions has been studied<br />

extensively. The alpha and theta class glutathione S-transferases are<br />

responsible for the conjugation <strong>of</strong> EDB both in rats and man. For both <strong>of</strong><br />

these enzymes enormous differences in levels between individuals have been<br />

found, which may be due to genetic differences, but are certainly also<br />

influenced by induction. One might expect individuals with an increased<br />

relative amount <strong>of</strong> glutathione S-transferases to be at increased risk.<br />

Reversible glutathione conjugates acting as transporting<br />

agents<br />

Numerous substrates for glutathione conjugation exist where a formal<br />

addition takes place: both the glutathionyl residue and the hydrogen atom<br />

are added to the acceptor molecule. From a chemical point <strong>of</strong> view, this<br />

reaction should be relatively easily reversible. Of course, the extent <strong>of</strong> the<br />

occurrence <strong>of</strong> the reverse reaction depends on the position <strong>of</strong> the<br />

equilibrium and is influenced by such conditions as the concentration <strong>of</strong><br />

the reactants and the pH. The biological consequences <strong>of</strong> this reaction<br />

sequence would be that the original electrophile is detoxified initially, but<br />

not permanently: it can be released again and thus appear in unexpected<br />

parts <strong>of</strong> the body. The glutathione conjugate serves as a storage or<br />

transport form for the alkylating agent. Systemic effects <strong>of</strong> highly reactive<br />

compounds might be explained in this way.<br />

For both isothiocyanates and isocyanates evidence for this pathway has<br />

been obtained. Benzyl and allyl isothiocyanate are both naturally occurring<br />

compounds that are excreted mainly as mercapturic acids in urine after<br />

administration to rats (Brüsewitz et al., 1977). However, the mercapturate<br />

in urine is unstable under basic conditions and reforms the free<br />

isothiocyanate. The glutathione, cysteine as well as N-acetyl-cysteine<br />

conjugates derived from these isothiocyanates are all toxic in vitro<br />

(Bruggeman et al., 1986, Temmink et al., 1986). In vivo, the fact that the<br />

conjugates are somewhat more unstable in urine probably plays a role in<br />

the effects. Benzyl isothiocyanate is used for the treatment <strong>of</strong> bladder<br />

infections (Brüsewitz et al., 1977), while allyl iso-thiocyanate causes<br />

bladder tumors in male rats (Dunnick et al., 1982).<br />

The extremely reactive and toxic methyl isocyanate, used in the<br />

manufacture <strong>of</strong> carbamate pesticides, was released into the atmosphere in


68 METABOLISM OF REACTIVE CHEMICALS<br />

large amounts during a disaster in 1984. To explain the systemic effects <strong>of</strong><br />

exposure to this compound, Baillie and coworkers hypothesized that these<br />

are mediated by the glutathione conjugates (Pearson et al., 1990). In fact, a<br />

rapid distribution <strong>of</strong> radioactivity throughout the body was found for rats<br />

exposed to 14C-methyl isocyanate vapor (Ferguson et al., 1988), the<br />

glutathione conjugate was identified in bile (Pearson et al, 1990) and the<br />

mercapturic acid was identified as a major urinary metabolite (Slatter et<br />

al., 1991) <strong>of</strong> rats dosed with methyl isocyanate. As was found for the<br />

isothiocyanates, in aqueous solution the synthetic glutathione conjugates<br />

are in equilibrium with the free electrophiles and glutathione: when an<br />

excess <strong>of</strong> cysteine is added to the solution, the corresponding cysteine<br />

conjugate is formed rapidly (Pearson et al., 1990). It should be realized<br />

however, that although thiols are the prime targets <strong>of</strong> iso- thiocyanates and<br />

isocyanates, the reactions with oxygen and nitrogen nucleophiles also<br />

occur and give rise to adducts that are much more stable (Pearson et al.,<br />

1991).<br />

The veterinary drug furazolidone is metabolized to a reactive metabolite<br />

that possesses an α,<br />

β-unsaturated ketone functionality. A reversible, socalled<br />

Michael adduct <strong>of</strong> this metabolite with glutathione was identified<br />

and has been suggested to play a role in the toxic effects <strong>of</strong> furazolidone<br />

(Vroomen et al., 1987). In fact residues <strong>of</strong> this metabolite covalently bound<br />

to microsomal protein could be trapped by an excess <strong>of</strong> mercaptoethanol<br />

and the glutathione conjugate gives rise to covalent binding to microsomal<br />

protein (Vroomen et al., 1988). Similarly, 2-methylfuran is metabolized to<br />

acetyl acrolein. The glutathione conjugate derived from this metabolite is<br />

unstable, and in fact toxicity <strong>of</strong> 2-methylfuran is potentiated by increasing<br />

glutathione levels by the administration <strong>of</strong> the cysteine precursor L-2oxothiazolidine-4-carboxylate<br />

(Ravindranath and Boyd, 1991).<br />

Thus, the reversibility <strong>of</strong> glutathione conjugation reactions warrants<br />

further investigation. The fact that reactive intermediates can be reformed<br />

might have important implications for the explanation <strong>of</strong> effects at sites<br />

distant from the site <strong>of</strong> initial exposure and/or initial conjugation.<br />

Conclusion<br />

Reactive chemicals can be detoxified fairly efficiently by several ubiquitous<br />

biotransformation enzymes. However, numerous cases have been reported<br />

where the initial detoxification is not the end <strong>of</strong> the story. The various<br />

pathways that the initially formed metabolites may undergo can result in<br />

unexpected toxicities at sites distant from the point <strong>of</strong> entry into the body<br />

<strong>of</strong> the electrophilic xenobiotic or the site <strong>of</strong> formation <strong>of</strong> the electrophilic<br />

metabolite.


P.J.VAN BLADEREN AND B.VAN OMMEN 69<br />

References<br />

AHMED, A.E. and ANDERS, M.W., 1976, Drug Metab. Dispos. 4, 357–61.<br />

AHMED, A.E. and ANDERS, M.W. 1978, Biochem. Pharmacol, 27, 2021–5.<br />

BARTELS, M.J. and TIMCHALK, C., 1990, Xenobiotica, 20, 1035–42.<br />

BAUMANN, E. and PREUSSE, C., 1879, Ber. Dtsch. Chem. Ges., 12, 806.<br />

BLADEREN, P.J.VAN, 1988, Trends Pharm. Sci., 9, 295–8.<br />

BLADEREN, P.J.VAN, GEN, A.VAN DER, BREIMER, D.D. and MOHN, G.R.,<br />

1979, Biochem. Pharmacol., 28, 2521–4.<br />

BLADEREN, P.J.VAN, BREIMER, D.D.ROTTEVEEL-SMIJS, G.M.T., JONG, R.<br />

A.W.DE, BUYS, W., GEN, A.VAN DER and MOHN, G.R., 1980, Biochem.<br />

Pharmacol., 29, 2975–82.<br />

BLADEREN, P.J.VAN, BREIMER, D.D., ROTTEVEEL-SMIJS, G.M.T., DE<br />

KNIFF, P., MOHN, G.R., MEETEREN, B.VAN, BUYS, W. and GEN, A.VAN<br />

DER, 1981a, Carcinogenesis, 2, 499–505.<br />

BLADEREN, P.J.VAN, BREIMER, D.D., HUIJGEVOORT, J.A.T.C.M VAN,<br />

VERMEULEN, N.P. E. and GEN, A. VAN DER, 1981b, Biochem.<br />

Pharmacol, 30, 2499–502.<br />

BOARD, P.G. 1981, Am. J. Hum. Genet., 33, 36–43.<br />

BOGAARDS, J.J.P., OMMEN, B.VAN and BLADEREN, P.J.VAN, 1993, Biochem<br />

Pharmacol., 45, 2166–9.<br />

BRUGGEMAN, J.M., TEMMINK, J.M.H. and BLADEREN, P.J.VAN 1986,<br />

Toxicol. Appl. Pharmacol, 83, 349–59.<br />

BRÜSEWITZ, G., CAMERON, B.D., CHASSEAUD, L.F., GORLER, K.,<br />

HAWKINS, D.R., KOCH, K. and MENNICKE, W.H., 1977, Biochem J., 162,<br />

99–107.<br />

CHASSEAUD, L.F., 1979, Adv. Cancer Res.,29, 175–274.<br />

CHEEVER, K.L., CHOLAKIS, J.M., MONAEM EL-HAWARI, A., KOVATCH,R.<br />

M. and WEISBURGER, E.K., 1990, Fundam. Appl Toxicol, 14, 243–61.<br />

CMARIK, J.L., INSKEEP, P.B., MEREDITH, M.J., MEYER, D.J., KETTERER, B.<br />

and GUENGERICH, F.P., 1990, Cancer Res.50, 2747–52.<br />

COMMANDEUR, J.N. and VERMEULEN, N.P.E., 1990, Chem. Res. Toxicol, 3,<br />

22–218.<br />

DIERICKX, P.J., 1982, Pharmacol. Res. Commun., 15, 581–91.<br />

DIERICKX, P.J., 1983, Res. Commun. Chem. Pathol. Pharmacol. 41, 517–20.<br />

DOHN, D.R. and CASIDA, J.E. 1987, Bioorg. Chem., 15, 115–24.<br />

DONG-HYUN, K., HUMPHREYS, W.G. and GUENGERICH, F.P., 1990, Chem.<br />

Res. Toxicol, 3, 587–94.<br />

DUNNICK, J.R., PREJEAN, J.D., HASEMAN, J., THOMPSON, R.B., GILES,H.<br />

D., and MACCONNELL, E.E. 1982, Fundam. Appl. Toxicol, 2, 114–20.<br />

ECKERT, K.G., ELBERS, F.R. and EYER, P., 1989, Biochem. Pharmacol., 38,<br />

3253–9.<br />

ECKERT, K.G., EYER, P., SONNEBICHLER, J. and ZETL I., 1990, Xenobiotica,<br />

20, 333–50.<br />

ELFARRA, A.A. and ANDERS M.W., 1985, J. Pharmacol. Expt. Ther., 233, 512–<br />

16.


70 METABOLISM OF REACTIVE CHEMICALS<br />

European Chemical Industry Ecology & <strong>Toxicology</strong> Centre (ECETOC), 1988,<br />

Technical report no. 26.<br />

FERGUSON, J.S., KENNEDY, A.L., STOCK, M.F., BROWN, W.E. and ALARIE,<br />

Y. 1988, Toxicol. Appl. Pharmacol, 94, 104–17.<br />

GILL, S.S., HAMMOCK B.D. and CASIDA, J.E., 1974, J. Agric. Food Chem., 22,<br />

386–95.<br />

GUENGERICH, F.P., CRAWFORD, M.W. Jr., DOMORADZKI, J.Y.,<br />

MACDONALD, T.L. and WATANABE, P.G., 1984, Toxicol. Appl.<br />

Pharmacol., 55, 303– 17.<br />

GUENTHNER, T.M. and OESCH, F., 1983, J. Biol. Chem., 258, 15054–61.<br />

HAMMOCK, B.D. and HASAGAWA, L.S., 1983, Biochem. Pharmacol., 32, 1155–<br />

64.<br />

HUMPHREYS, W.G., DONG-HYUN, K., CMARIK, J.L., SHIMADA, T. and<br />

GUENGERICH, F.P., 1990, Biochemistry, 29, 10342–50.<br />

HUMPHREYS, W.G., DONG-HYUN, K. and GUENGERICH, F.P., 1991, Chem.<br />

Res. Toxicol., 4, 445–53.<br />

INSKEEP, P.B. and GUENGERICH, F.P., 1984, Carcinogenesis, 5, 805–8.<br />

JAKOBY, W.B. 1980, in Jakoby, W.B. (Ed.) Enzymatic Basis <strong>of</strong> Detoxication San<br />

Diego: Academic Press.<br />

JEAN, P.A. and REED, D.J., 1989, Chem. Res. Toxicol, 2, 455–60.<br />

KETTERER, B and MULDER, G.J., 1990, in Mulder, G.J., (Ed.) Conjugation<br />

Reactions in Drug Metabolism, pp. 307–64 London: Taylor and Francis.<br />

KOGA, N., INSKEEP, P.B., HARRIS., T.M. and GUENGERICH, F.P., 1986,<br />

Biochemistry, 25, 2192–8.<br />

KULKARNI, A.P., MOTOYAMA, N., DAUTERMAN, W.C. and HODGSON, E.,<br />

1978, Bull. Environm. Contam. Toxicol. 20, 227–32.<br />

LAU, S.S. and MONKS, T.J., 1990, Toxicol. Appl. Pharmacol., 103, 121–32.<br />

LAWLEY, P.D. and THATCHER, C.J. 1970, Biochem. J., 116, 693–707.<br />

LISTOWSKY, I., ABROMOVITZ, M., HOMMA, H and NIITSU, Y. 1988, Drug<br />

Metab. Rev., 19, 305–18.<br />

MANNERVIK, B. and JENSSON, H., 1982, J. Biol. Chem., 257, 9909–12.<br />

MERTENS, J.M., TEMMINK, J.H.H., BLADEREN, P.J.VAN, JONES, T.W., Lo,<br />

H.H., LAU, S.S. and MONKS, T.J., 1991, Toxicol. Appl. Pharmacol, 110, 45–<br />

61.<br />

MONKS, T.J., and LAU, S.S., 1990, Toxicol. Appl. Pharmacol, 103, 557–63.<br />

MONKS, T.J., LAU, S.S., HIGHET, R.J. and GILLETTE, J.R., 1985, Drug. Metab.<br />

Dispos., 13, 553–9.<br />

MONKS, T.J. HIGHET, R.J., and LAU, S.S. 1988a, Mol Pharmacol, 34, 492–500.<br />

MONKS, T.J., HIGHET, R.J., and LAU, S.S. 1990a, Mol. Pharmacol, 38, 121–7.<br />

MONKS, T.J., HIGHET, R.J., CHU, P.S. and LAU, S.S. 1988b, Mol. Pharmacol.,<br />

34, 15–22.<br />

MONKS, T.J., ANDERS, M.W., DEKANT, W., STEVENS, J.L., LAU, S.S. and<br />

BLADEREN, P.J.VAN, 1990b, Toxicol. Appl. Pharmacol., 106, 1–19.<br />

MORGENSTERN, R., LUNDQVIST, G., HANCOCK, V. and DEPIERRE, J.W.,<br />

1988, J. Biol. Chem., 263, 6671–5.<br />

OESCH, F., 1972, Xenobiotica, 3, 305–40.


P.J.VAN BLADEREN AND B.VAN OMMEN 71<br />

OMMEN, B.VAN, BESTEN, C.DEN, RUTTEN, A.C.M., PLOEMEN, J.H.T.M.,<br />

Vos, R.M.E., MÜLLER, M. and BLADEREN, P.J.VAN, 1988, J. Biol Chem.,<br />

263, 12939–12942.<br />

OMMEN, B.VAN, BOGAARDS, J.J.P., PETERS, W.H.M., BLAAUBOER, B. and<br />

BLADEREN, P.J.VAN, 1990, Biochem. J., 269, 609–13.<br />

OZAWA, N. and GUENGERICH, F.P., 1983, Proc. Nat. Acad. Sci. USA, 80,<br />

5266– 70.<br />

PEARSON, P.G., SODERLUND, E.J., DYBING, E, and NELSON, S.D., 1990a,<br />

Biochemistry, 29, 4971–81.<br />

PEARSON, P.G., SLATTER, G.T., RAKSHED, M.S., MAN, D.M., GRILLO,M. P.<br />

and BAILLIE, T.A., 1990b, Biochem. Biophys. Res. Commun., 166, 245– 50.<br />

PEARSON, P.G., SLATTER, J.G., RASHED, M.S., HAN, D.H., and BAILLIE,<br />

TH.A, 1991, Chem. Res. Toxicol, 4, 436–44.<br />

PETERSON, L.A., HARRIS, T.M., GUENGERICH, F.P., 1988, J. Am. Soc., 110,<br />

3284–91.<br />

PROHASKA, J.R., 1980, Biochim. Biophys. Acta., 611, 87–98.<br />

RAVINDRANATH, V. and BOYD, M.R. 1991, Biochem. Pharmacol., 41,(9),<br />

1311–18.<br />

SAMUELSSON, B., 1988, Science, 220, 568–75.<br />

SCHLADT, L., THOMAS, H., HARTMANN, R. and OESCH, F., 1988, Eur. J.<br />

Biochem., 176, 715–23.<br />

SLATTER, J.G., RASHED, M.S., PEARSON, P.G., HAN, D.H, and BAILLIE,<br />

TH.A., 1991, Chem. Res. Toxicol, 4, 157–61.<br />

SMITH, M.T., EVANS, C.G., THOR, H. and ORRENIUS, S., 1985, Oxidative<br />

Stress, 91–113.<br />

SÖDERSTRÖM, M., MANNERVIK, B., ORNING, L. and HAMMARSTRÖM,<br />

S., 1985, Biochem. Biophys. Res. Commun., 128, 265–70.<br />

TEMMINK, J.H.M., BRUGGEMAN, I.M. and BLADEREN, P.J.VAN, 1986,<br />

Arch. Toxicol., 59, 103–10.<br />

UJIHARA, M., TSUCHIDA, S., SATOH, K., SATO, K. and URADE, Y. 1988,<br />

Arch. Biochem. Biophys., 264, 428–37.<br />

Vos, R.M.E., and VAN BLADEREN, P.J. 1990, Chem.-Biol. Interact. 75, 241–65.<br />

Vos, R.M.E., VAN WELIE, R.T.H., PETERS, W.M., EVELO, C.T.A.,<br />

BOGAARDS, J.J.P., VERMEULEN, N.P.E. and VAN BLADEREN, P.J., 1991,<br />

Arch. Toxicol. 65, 95–9.<br />

VROOMEN, L.H.M., GROTEN, J.P., MUISWINKEL, K.VAN, VELDHUIZEN, A.<br />

VAN and BLADEREN, P.J.VAN, 1987, Chem. Biol. Interact. 64, 167–79.<br />

VROOMEN, L.H.M., BERGHMANS, M.C.J., GROTEN, J.P., KOEMAN, J.H.,<br />

and BLADEREN, P.J.VAN, 1988, Toxicol. Appl. Pharmacol, 95, 53–60.<br />

WHITE, R.D., GANDOLFI, A.J., BOWDEN, G.T. and SIPES, I.G., 1983, Toxicol.<br />

Appl. Pharmacol., 69, 170–8.<br />

WITZ, G., 1989, Free Radical Biol. Med., 7, 333–49.<br />

WONG, L.C.K., WINSTON, J.M., HONG, C.B. and PLOTNICK, H., 1982,<br />

Toxicol. Appl. Pharmacol. 63, 155–65.<br />

ZOETEMELK, C.E.M., OEI, I.H., MEETEREN-WALCHI, B., ONKENHOUT,<br />

W., GEN, A.VAN DER and BREIMER, D.D., 1986, Drug Metab. Dispos. 14,<br />

601–7.


6<br />

Methods for the Determination <strong>of</strong> Reactive<br />

<strong>Compounds</strong><br />

PETER SAGELSDORFF<br />

CIBA-GEIGY Ltd, Basel<br />

Introduction<br />

It has well been recognised for a long time that adverse effects <strong>of</strong> chemicals<br />

are associated with their reactivity whereby many unreactive chemicals are<br />

metabolised in the cell to a reactive intermediate. Reactive intermediates<br />

are generally electrophiles which undergo reactions with cellular<br />

nucleophiles and the toxicological response is <strong>of</strong>ten the consequence <strong>of</strong> the<br />

covalent binding <strong>of</strong> a chemical to cellular macromolecules. This chapter<br />

will provide a brief survey <strong>of</strong> current methods for the determination <strong>of</strong><br />

protein and DNA adducts generated by reactive compounds, and will<br />

discuss some useful applications <strong>of</strong> these technologies.<br />

Source <strong>of</strong> reactive metabolites<br />

Reactive chemicals are generally strong electrophilic agents. These<br />

compounds can be reactive per se (direct electrophiles), such as methylmethanesulphonate,<br />

epoxides or strained lactones. On the other hand,<br />

unreactive chemicals can be enzymatically converted to electrophilic agents<br />

(indirect electrophiles), such as aromatic amines and nitroarenes to the<br />

corresponding nitrenium ions, polycyclic aromatic hydrocarbons to diol<br />

epoxides or N-nitroso compounds to carbenium ions (Figure 6.1; Magee et<br />

al., 1975; Weissburger and Williams, 1975; Lutz, 1979).<br />

Interaction <strong>of</strong> reactive compounds with cellular<br />

constituents<br />

As electrophiles, these reactive compounds undergo reactions with<br />

nucleophiles. The nature <strong>of</strong> the toxicological response is dependent on the<br />

biological macromolecule affected. Reactions with water or glutathione,<br />

two <strong>of</strong> the most abundant cellular nucleophiles, in most cases, lead to an<br />

inactivation <strong>of</strong> the respective reactive compound.


P.SAGELSDORFF 73<br />

Figure 6.1 Examples <strong>of</strong> electrophilic compounds. The arrows indicate the suspected<br />

electrophilic centres (from Lutz, 1979).


74 METHODS FOR THE DETERMINATION OF REACTIVE COMPOUNDS<br />

Figure 6.2 Nucleophilic centres in nucleobases and DNA ‘adduct library’ indicating<br />

the preferential binding sites on guanine for several classes <strong>of</strong> chemicals. The most<br />

reactive targets are indicated by an arrow (from Lutz, 1979; Beach and Gupta,<br />

1992).<br />

The most important nucleophilic centres in proteins are the side chains<br />

<strong>of</strong> the amino acids cysteine, methionine, histidine and tyrosine, and the<br />

amino group <strong>of</strong> the N-terminal amino acid. Reactions <strong>of</strong> electrophiles with<br />

proteins lead to the formation <strong>of</strong> protein adducts. This results in general or<br />

specific cytotoxicity depending on whether the function <strong>of</strong> a particular<br />

protein is disturbed (Lawley, 1976; Brooks, 1977). Adduct formation with<br />

blood proteins can result in the formation <strong>of</strong> immunogens and subsequent<br />

allergenic responses.<br />

Finally, reactions with DNA predominantly occur with the nucleobases<br />

adenine, cytosine, thymine and guanine, whereby the most important<br />

nucleophile in DNA is guanine (Figure 6.2). Adduct formation with<br />

nucleobases in DNA is recognised as a crucial step in the formation <strong>of</strong><br />

mutations and cancer (Lutz, 1979).


Methods for the determination <strong>of</strong> adducts<br />

Adduct formation <strong>of</strong> reactive compounds with protein or DNA can easily<br />

be detected by the use <strong>of</strong> radiolabelled test compounds. However, the<br />

radiolabelled compounds are <strong>of</strong>ten not available. In addition, real exposure<br />

situations and unknown mixtures <strong>of</strong> compounds can not be assessed. For a<br />

number <strong>of</strong> chemicals, therefore, alternative methods for adduct<br />

determinations have been developed during the past.<br />

Reactions with proteins can be assessed by analysing haemoglobin or<br />

albumin adducts. These proteins can easily be isolated in large quantities<br />

(100 mg haemoglobin, 30 mg albumin per ml blood) and with sufficient<br />

purity from the blood <strong>of</strong> treated animals or occupationally exposed<br />

humans. Methods for the determination <strong>of</strong> DNA adducts generally require<br />

a higher sensitivity, since DNA from treated animals or exposed humans is<br />

only available in small amounts (1–2 mg per g tissue, 4 µg from white<br />

blood cells per ml blood).<br />

Protein adducts<br />

Physical methods<br />

Aromatic amines and nitroarenes<br />

The key step in the metabolic activation <strong>of</strong> arylamines to the respective<br />

nitrenium ions involves N-hydroxylation. The N-hydroxylamines can be<br />

further oxidised in erythrocytes to the corresponding nitroso compounds<br />

with a concurrent production <strong>of</strong> methaemoglobin. On the other hand,<br />

nitroarenes can be metabolically reduced to the corresponding<br />

nitrosoarenes. The nitrosoarenes covalently bind to the thiol group <strong>of</strong><br />

cysteine residues and rearrange to give stable sulphinic acid amides.<br />

Mild alkaline treatment can be used to hydrolyse these adducts. The<br />

liberated parent amines can be extracted and analysed by HPLC with<br />

specific detection methods, such as electrochemical or fluorescence<br />

detection. In order to improve the sensitivity <strong>of</strong> the assay, the extracted<br />

adducts can be derivatised with electrophores and analysed by GC with<br />

electron capture detection or by GC/MS (Bailey et al., 1990; Skipper and<br />

Tannenbaum, 1990; Sabbioni, 1992, 1994).<br />

Polycyclic aromatic hydrocarbons<br />

P.SAGELSDORFF 75<br />

Polycyclic aromatic hydrocarbons are oxidised by cytochrome P450 to<br />

epoxides, which are rapidly hydrolysed. However, further oxidation to the


76 METHODS FOR THE DETERMINATION OF REACTIVE COMPOUNDS<br />

Figure 6.3 Modified Edman degradation <strong>of</strong> alkylated N-terminal valine in<br />

haemoglobin (Törnqvist et al., 1986).<br />

respective diol epoxides results in the formation <strong>of</strong> relatively stable<br />

electrophiles which also alkylate cysteine residues in proteins.<br />

Upon mild acid treatment these adducts are liberated as the respective<br />

tetrols. Similar to adducts from aromatic amines, the tetrols can be<br />

extracted and analysed by HPLC with specific detection methods, or by GC<br />

with electron capture detection or by GC/MS after derivatisation with<br />

electrophores (Shugart and Kao, 1985; Weston et al., 1989; Day et al.,<br />

1990).<br />

Alkylating agents<br />

Adducts <strong>of</strong> alkylating agents with the thiol group <strong>of</strong> cysteine, histidine or<br />

the N-terminal amino acids resist alkaline or acid hydrolysis. To determine<br />

the alkylated amino acids the protein is hydrolysed with 6 N HCl and the<br />

amino acids are separated on a anion exchange column. The fractions<br />

containing the alkylated amino acids are derivatised with electrophores and<br />

analysed by GC/MS (van Sittert et al., 1985; Bailey et al., 1987).


Alternatively, the alkylated N-terminal valine <strong>of</strong> haemoglobin can<br />

selectively be cleaved <strong>of</strong>f by a modified Edman degradation with<br />

pentafluorophenyl isothiocyanate (PFPITC). Since alkylation <strong>of</strong> the amino<br />

group favours the reaction, conditions can be selected to exclusively<br />

liberate alkylated N-terminal amino acids whilst leaving the non-adducted<br />

N-terminal valine intact (Törnqvist et al., 1986). The resulting<br />

pentafluorophenyl thiohydantoine (PFPTH) derivative can be extracted and<br />

quantified by GC/MS (Figure 6.3).<br />

Immunological methods<br />

Immunological methods have been developed for the quantification <strong>of</strong><br />

some adducts <strong>of</strong> aromatic amines, polycyclic aromatic hydrocarbons and<br />

alkylating agents. However, these methods involve a couple <strong>of</strong> time<br />

consuming steps for the isolation <strong>of</strong> an appropriate antibody. The<br />

respective haemoglobin adduct has to be chemically synthesised, an animal<br />

has to be immunised with the modified haemoglobin and, later on,<br />

polyclonal antibodies can be isolated from the blood <strong>of</strong> the immunised<br />

animal. In order to produce monoclonal antibodies, which normally have a<br />

better specificity and sensitivity, spleen cells <strong>of</strong> the immunised animal are<br />

fused with myeloma cells and the antibodies can be isolated from the cell<br />

culture.<br />

The methods for the determination <strong>of</strong> adducts include competitive<br />

radioimmunoassays and solid phase assays (ELISA, USERIA). The protein<br />

is partially hydrolysed, adsorbed on a solid surface and treated with the<br />

primary antibody. An anti-antibody which is directed against the primary<br />

antibody, radiolabelled, or conjugated to a fluorescent dye or an indicator<br />

enzyme, is added and the amount <strong>of</strong> bound label is quantified (Santella et al.,<br />

1986; Lee and Santella, 1988).<br />

DNA adducts<br />

Physical methods<br />

Aromatic amines and nitroarenes<br />

P.SAGELSDORFF 77<br />

The hydroxylamines produced by enzymatic hydroxylation <strong>of</strong> aromatic<br />

amines or by reduction <strong>of</strong> nitrosoarenes are further conjugated (Osulphatation,<br />

O-acetylation, O-glucuronidation). The conjugates can<br />

decompose to the respective nitrenium ions which add predominantly to<br />

the C8 <strong>of</strong> guanine.<br />

Similarly to protein adducts <strong>of</strong> these compounds, the adducts can be<br />

liberated from DNA by alkaline hydrolysis or hydrazinolysis, extracted and


78 METHODS FOR THE DETERMINATION OF REACTIVE COMPOUNDS<br />

quantified by HPLC with fluorescence or electrochemical detection. In<br />

order to improve the sensitivity the extracted adducts can be derivatised<br />

with electrophores and analysed by GC/MS (Bakthavachalam et al., 1991;<br />

Lin et al., 1991).<br />

Polycyclic aromatic hydrocarbons<br />

The diol epoxides enzymatically produced from polycyclic aromatic<br />

hydrocarbons mainly adduct at the exocyclic amino group <strong>of</strong> guanine. The<br />

adducts can be liberated from DNA by acid hydrolysis, extracted and<br />

quantified by HPLC with fluorescence or electrochemical detection or by<br />

GC with electron capture detection or GC/MS after suitable derivatisation<br />

with electrophores (Rahn et al., 1982; Shugart and Kao, 1985; Weston et<br />

al., 1989).<br />

Alkylating agents<br />

Alkylating agents mainly alkylate the N7 <strong>of</strong> guanine but also give rise to<br />

the formation <strong>of</strong> other N- and O-alkyl nucleobase adducts. The DNA bases<br />

are liberated by hydrolysis and analysed for the presence <strong>of</strong> adducts by<br />

HPLC with electrochemical detection or they are extracted, derivatised<br />

with electrophores and analysed by GC/MS (Minnetian et al., 1987; Groot<br />

et al., 1994). Some alkylating agents and small epoxides lead to the<br />

formation <strong>of</strong> cyclic nucleobase adducts which exhibit strong fluorescence.<br />

Enzymatic or acid hydrolysis can be used for the liberation <strong>of</strong> the DNA<br />

constituents and the fluorescent adducts can be analysed by HPLC with<br />

fluorescence detection (Fedtke et al., 1990; Steiner et al., 1992a).<br />

Immunological methods<br />

Immunological methods for the determination <strong>of</strong> DNA adducts essentially<br />

follow the procedure as outlined already for protein adducts: generation <strong>of</strong><br />

an antibody, absorption <strong>of</strong> the DNA on a solid surface, incubation with the<br />

antibody and a labelled anti-antibody. However, for the production <strong>of</strong> the<br />

antibody an additional step has to be performed. The immune system<br />

normally does not respond to small molecules. Therefore, the chemically<br />

synthesised base or nucleoside adduct has to be coupled to a carrier protein,<br />

in order to obtain an immunogen (Perera et al., 1986; Santella, 1988;<br />

Poirier, 1993).<br />

Postlabelling<br />

One <strong>of</strong> the most popular assays for determination <strong>of</strong> DNA adducts is the<br />

postlabelling assay. The DNA is enzymatically hydrolysed to the four


natural deoxynucleoside-3′-monophosphates (dNp) and the dNp adducts.<br />

The adducted dNp carrying bulky or aromatic substituents are enriched by<br />

extraction with butanol in the presence <strong>of</strong> a phase transfer agent or by<br />

selective digestion <strong>of</strong> the natural (unadducted) dNp with nuclease P1. The<br />

enriched adducted dNp are labelled with [ 32 P]- or [ 33 P]ATP (Figure 6.4).<br />

Polynucleotide kinase T4 is used to catalyse the transfer <strong>of</strong> the labelled<br />

phosphate group from ATP to the 5′ position <strong>of</strong> the dNp. The labelled<br />

deoxynucleoside-3′,5′-bisphosphates are then separated by multidirectional<br />

TLC on polyethyleneimine coated cellulose. Radioactive impurities and<br />

unused ATP are running to the top with phosphate buffer (D1), whereas<br />

nucleotides carrying aromatic or bulky adducts are retained at or near the<br />

origin. The part containing the impurities and unused ATP is cut <strong>of</strong>f, and<br />

the adducts are chromatographed in D3 (opposite to D1) and D4<br />

(perpendicular to D3) with ammonia and ammonia/ propanol or urea<br />

containing buffers. Adduct spots are visualised and quantified by<br />

autoradiography and Cherenkov counting or by phosphor imaging (Gupta,<br />

1985; Reddy and Randerath, 1986; Beach and Gupta, 1992).<br />

Alternatively, the enriched nucleotide adducts can be chemically<br />

derivatised with fluorescent labels and analysed by HPLC with fluorescence<br />

detection. However, this method does not reach the sensitivity <strong>of</strong> the<br />

radioactive assay (Sharma and Jain, 1991; Jain and Sharma, 1993).<br />

Comparison <strong>of</strong> different methods<br />

P.SAGELSDORFF 79<br />

The methods used for the determination <strong>of</strong> protein and DNA adducts are<br />

summarised in Tables 6.1 and 6.2. Special attention is drawn to the cost <strong>of</strong><br />

equipment and time required for analysis.<br />

HPLC methods with electrochemical or fluorescent detection are<br />

relatively insensitive and only applicable with compounds which are<br />

strongly fluorescent or electrochemically active. Since the costs for the<br />

equipment used and the time consumption are relatively low, these<br />

methods are attractive in certain cases. GC with electron capture detection<br />

or GC/MS <strong>of</strong>fers better sensitivity. However, the method requires<br />

derivatisation. In addition, the costs for the equipment <strong>of</strong> the GC/MS<br />

methods are quite high. Immunoassays are very sensitive, but involve a<br />

number <strong>of</strong> time consuming steps for the preparation <strong>of</strong> an appropriate<br />

antibody, and are only possible if the structure <strong>of</strong> the respective adduct is<br />

known. The postlabelling method for DNA adducts <strong>of</strong>fers the best<br />

sensitivity, with low equipment costs and low to medium time<br />

consumption. However, the standard method only detects bulky or<br />

aromatic adducts.


80 METHODS FOR THE DETERMINATION OF REACTIVE COMPOUNDS<br />

Figure 6.4 Schematic representation <strong>of</strong> the postlabelling assay for determination <strong>of</strong><br />

DNA adducts (Gupta, 1985; Reddy and Randerath, 1986; Beach and Gupta, 1992).<br />

Examples/applications<br />

In the following sections some useful applications <strong>of</strong> adduct<br />

determinations, which have been performed in our laboratory, will be<br />

presented.


Table 6.1 Methods for the determination <strong>of</strong> protein adducts<br />

a An approximate mean sensitivity is given in pmol (=10 −12 mol) adducts/g<br />

haemoglobin.<br />

Table 6.2 Methods for the determination <strong>of</strong> DNA adducts<br />

Lack <strong>of</strong> bioavailability <strong>of</strong> 3,3′-dichlorobenzidine from<br />

diarylide pigments<br />

P.SAGELSDORFF 81<br />

a An approximate mean sensitivity is given in fmol (=10 −15 mol) adducts/mg DNA.<br />

3,3′-Dichlorobenzidine is an important intermediate in the production <strong>of</strong><br />

diarylide pigments and azo dyes. Some <strong>of</strong> these pigments have been tested<br />

in long term studies and shown to exert no specific toxicological effects and<br />

to be not carcinogenic to experimental animals (ETAD Report, 1990).<br />

However, there might be a theoretical hazard after metabolic splitting <strong>of</strong> the<br />

pigments into DCB, a known animal carcinogen (IARC, 1982). DCB and<br />

its N-acetylated metabolite are N-hydroxylated and oxidised to the<br />

corresponding nitroso compound which binds to haemoglobin. Since no<br />

repair <strong>of</strong> haemoglobin adducts occurs, these adducts cumulate during the


82 METHODS FOR THE DETERMINATION OF REACTIVE COMPOUNDS<br />

Figure 6.5 HPLC/ECD pr<strong>of</strong>iles obtained after hydrolysis and extraction <strong>of</strong><br />

haemoglobin samples isolated from an untreated rat (control) and from rats treated<br />

for 4 weeks with DCB (2 mg kg −1 ), Direct Red 46 (160 mg kg −1 ), Pigment Yellow<br />

13 (400 mg kg −1 ) and Pigment Yellow 17 (400 mg kg −l ) as well as <strong>of</strong> commercially<br />

available bovine haemoglobin (Hb-bovine).<br />

life span <strong>of</strong> the erythrocyte. Haemoglobin adduct formation, therefore, was<br />

used to monitor the liberation <strong>of</strong> DCB from diarylide pigments.<br />

Rats were treated by daily oral gavage for 4 weeks with the pigment at<br />

daily dose levels <strong>of</strong> 400 mg kg −1 body weight. As a positive control,<br />

animals were treated accordingly with DCB (2 mg kg −1 ) or with Direct Red<br />

46 (160 mg kg −1 ), asoluble azo dye with known bioavailability <strong>of</strong> DCB.<br />

After termination <strong>of</strong> the treatment, haemoglobin was isolated and<br />

hydrolysed in 0.1 N sodium hydroxide. The liberated DCB and<br />

monoacetyl-DCB were extracted with toluene/2-propanol and analysed by<br />

HPLC with electrochemical detection. With 2 mg DCB kg −1 body weight<br />

DCB and monoacetyl-DCB adducts were clearly detectable, amounting up


to 50 ng g −1 haemoglobin (Figure 6.5). No macromolecular adducts were<br />

detectable in the rats treated with the two diarylide pigments. The limits <strong>of</strong><br />

determination would correspond to a daily DCB dose <strong>of</strong> 0.3–0.5 mg kg −1<br />

body weight, indicating that DCB was not liberated from the pigments at a<br />

determination limit <strong>of</strong> 0.3% <strong>of</strong> the DCB equivalents, whereas the<br />

bioavailability <strong>of</strong> DCB in the rats treated with the azo dye could clearly be<br />

confirmed.<br />

Formation <strong>of</strong> glycidaldehyde from glycidylethers<br />

Bisphenol A diglycidylether (BPADGE) is widely used as component <strong>of</strong><br />

epoxy resins. The chemical reactivity <strong>of</strong> this class <strong>of</strong> compounds is a<br />

prerequisite for their technical use, and accounts for the sensitising,<br />

mutagenic and in some cases carcinogenic properties <strong>of</strong> many epoxy resin<br />

monomers. It was suggested that the metabolic inactivation <strong>of</strong> BPADGE by<br />

hydrolysis <strong>of</strong> epoxides may form an equilibrium with its metabolic<br />

activation by oxidative dealkylation <strong>of</strong> the intact glycidyl side chain<br />

followed by the release <strong>of</strong> glycidaldehyde. Cutaneous treatment <strong>of</strong> mice<br />

with glycidaldehyde led to the formation <strong>of</strong> one major epidermal DNA<br />

adduct which was identified as HMEdA<br />

(hydroxymethylethenodeoxyadenosine, Steiner et al., 1992a). This cyclic<br />

deoxyadenosine adduct is strongly fluorescent and can be quantified by<br />

fluorescence measurements.<br />

In order to investigate the formation <strong>of</strong> glycidaldehyde from BPADGE,<br />

mice were treated with BPADGE (2 mg) and the fluorescent<br />

glycidaldehydeDNA adducts formed in epidermal DNA were compared<br />

with those obtained after treatment with glycidaldehyde (2 mg). After 24–<br />

96 h epidermal DNA was isolated, enzymatically digested to the<br />

deoxynucleoside-3'-monophosphates and analysed for the presence <strong>of</strong><br />

HMEdA by HPLC with fluorescence detection (excitation at 231 nm,<br />

emission at 420 nm). In glycidaldehyde treated mice 166 adducts per 10 6<br />

nucleotides could be detected after an exposure time <strong>of</strong> 24 h (Figure 6.6)<br />

whereas with epidermal DNA from BPADGE treated mice 0.2– 0.8<br />

adducts per 10 6 nucleotides were found. This adduct level would be equal<br />

to a dose <strong>of</strong> 10 µg glycidaldehyde, indicating that, at the most, 1.1% <strong>of</strong> the<br />

glycidaldehyde moiety in BPADGE were bioavailable for DNA-adduct<br />

formation (Steiner et al., 1992b).<br />

Determination <strong>of</strong> reactive compounds in unknown<br />

mixtures<br />

P.SAGELSDORFF 83<br />

A challenging task is the analysis <strong>of</strong> reactive metabolities in unknown<br />

mixtures <strong>of</strong> different compounds. In order to assess the impact <strong>of</strong> chemical<br />

pollution on aquatic organisms, rainbow trouts were continuously exposed


84 METHODS FOR THE DETERMINATION OF REACTIVE COMPOUNDS<br />

to the diluted effluent discharges <strong>of</strong> a chemical production plant for 3<br />

months. The plant produced different dyes and chemicals and the waste<br />

water therefore could be contaminated with a variety <strong>of</strong> aliphatic and<br />

aromatic amines and some cyclic aromatic hydrocarbons. After termination<br />

<strong>of</strong> the treatment, liver and gill DNA from exposed and control trouts was<br />

analysed by [ 32 P]postlabelling for the presence <strong>of</strong> DNA adducts.<br />

The DNA was enzymatically hydrolysed to the nucleotides. Adducted<br />

nucleotides were extracted with butanol in the presence <strong>of</strong> the phase<br />

transfer agent tetrabutylammonium chloride and postradiolabelled with<br />

[ 32 P]ATP and PNK. The labelled nucleotides were separated by<br />

multidirectional TLC with 1.0 M phosphate buffer, pH 6.6, in D1, 0.4 M<br />

ammonia in D3 and 4 N ammonia/propanol (1.2:1) in D4. A final<br />

development in direction D4 with 1.0 M phosphate buffer, pH 6.6, was<br />

used as background clean up.<br />

In the trouts exposed to control water no DNA adducts were detectable,<br />

neither in the livers nor in the gills (Figure 6.7). In contrast, in the trouts<br />

exposed to the highest concentration <strong>of</strong> the waste water, at least 4 DNA<br />

adducts could be found in the livers and in the gills. The overall DNA<br />

adduct level in the exposed trouts was relatively low (1 adduct per 10 8<br />

nucleotides, which indicated only a minimal cancer risk for the exposed<br />

fish.<br />

Limitations<br />

However, the methods presented for adduct determination have their<br />

limitations. For protein adduct determination the most popular method is<br />

by HPLC with electrochemical or fluorescence detection after hydrolysis<br />

and extraction <strong>of</strong> the adducts. This is due to the low cost and time<br />

consumption <strong>of</strong> the method. This method is hampered by the possibility <strong>of</strong><br />

interferences, which can elute in the range <strong>of</strong> the compounds <strong>of</strong> interest. For<br />

an exclusion <strong>of</strong> haemoglobin adducts formation at low levels it is therefore<br />

crucial to obtain additional information about the chromatographic peaks<br />

<strong>of</strong> interest, such as for example, by GC/MS.<br />

DNA adducts are <strong>of</strong>ten assessed by [ 32 P]postlabelling. This method is<br />

limited by low yields <strong>of</strong> the enrichment and labelling procedures and by<br />

choosing the appropriate chromatographic conditions for the resolution <strong>of</strong><br />

the labelled adducts. The lack <strong>of</strong> detectability <strong>of</strong> some DNA adducts,<br />

although they may contain aromatic moieties, enforces the use <strong>of</strong> a positive<br />

standard in order to check for the yield <strong>of</strong> the enrichment and the labelling<br />

reaction, and to check for appropriate chromatographic conditions to<br />

resolve the adducts.


P.SAGELSDORFF 85<br />

Figure 6.6 HPLC/fluorescence analysis <strong>of</strong> epidermal DNA hydrolysates from a<br />

control (a) and a BPADGE treated mouse (b), and UV trace <strong>of</strong> synthetic HMEdAp<br />

and HMEdGp (c).


86 METHODS FOR THE DETERMINATION OF REACTIVE COMPOUNDS<br />

Figure 6.7 TLC chromatograms <strong>of</strong> DNA adducts in gills and livers <strong>of</strong> rainbow<br />

trouts, exposed for 3 months to waste water or control water. Top: control water,<br />

liver DNA (left chromatogram), gill DNA (right chromatogram); bottom: waste<br />

water, liver DNA (left chromatogram), gill DNA (right chromatogram).<br />

Conclusions<br />

Each method, although inherently chemically-specific, has its advantages<br />

and limitations depending on the adduct-type. The continued rapid<br />

development <strong>of</strong> the technologies described for assessing biomarkers should<br />

result in more accurate assessment <strong>of</strong> the intracellular reactions <strong>of</strong><br />

chemicals and thereby provide information about the mechanism <strong>of</strong><br />

toxicity <strong>of</strong> a compound under investigation.


Acknowledgements<br />

Grateful thanks to Drs Markus Joppich and Regula Joppich-Kuhn for<br />

haemoglobin adduct analyses and Dr Sandra Steiner for the development<br />

<strong>of</strong> the fluorescence assay for HMEdAp.<br />

References<br />

P.SAGELSDORFF 87<br />

BAILEY, E., FARMER, P.B. and SHUKER, D.E.G., 1987, Estimation <strong>of</strong> exposure<br />

to alkylating carcinogens by the GC-MS determination <strong>of</strong> adducts to<br />

haemoglobin and nucleic acid bases in urine, Arch. Toxicol., 60, 187–91.<br />

BAILEY, E., BROOKS, A.G., BIRD, I., FARMER, P.B. and STREET, B., 1990,<br />

Monitoring exposure to 4,4′-methylenedianiline by the GC/MS determination<br />

<strong>of</strong> adducts to haemoglobin, Anal. Biochem., 190, 175–81.<br />

BAKTHAVACHALAM, J., ABEL-BAKY, S. and GIESE, R.W., 1991, Release <strong>of</strong> 2amin<strong>of</strong>luorene<br />

from N-(deoxyguanosine-8-yl)-2-amin<strong>of</strong>luorene by<br />

hydrazinolysis, J.Chromatogr., 538, 447–51.<br />

BEACH, A.C. and GUPTA, R.C., 1992, Human biomonitoring and the 32 Ppostlabelling<br />

assay, Carcinogenesis, 13, 1053–74.<br />

BROOKS, P., 1977, The role <strong>of</strong> covalent binding in carcinogenicity, in Jollow, D.J.<br />

et al. (Eds) Biological Reactive Intermediates, pp. 470–480, New York:<br />

Plenum.<br />

DAY, B.W., NAYLOR, S., GAN, L.-S., SHALI, Y., NGUYEN, T.T., SKIPPER, P. L.,<br />

WISHNOK, J.S. and TANNENBAUM, S.R., 1990, Molecular dosimetry <strong>of</strong><br />

polycyclic aromatic hydrocarbon epoxides and diol epoxides via haemoglobin<br />

adducts, Cancer Res., 50, 4611.<br />

ETAD Report, 1990, On the carcinogenic potential <strong>of</strong> diarylide azo pigments based<br />

on 3,3′-dichlorobenzidine, T 2028-CA, Toxicological Subcommittee <strong>of</strong> ETAD<br />

(TSC).<br />

FEDTKE, N., BOUCHERON, J.A., WALKER, V.E. and SWENBERG, J.A., 1990,<br />

Vinyl chloride induced DNA adducts: II: formation and persistence <strong>of</strong> 7-(2'oxoethyl)<br />

guanine and N 2 ,3-ethenoguanine in rat tissue DNA. Carcinogenesis,<br />

11, 1278–1292.<br />

GREEN, L.C., SKIPPER, P.L., TURESKY, R.J., BRYANT, M.S. and<br />

TANNENBAUM, S.R., 1984, In vivo dosimetry <strong>of</strong> 4-aminobiphenyl in rats via<br />

a cysteine adduct in haemoglobin, Cancer Res., 44, 4254–9.<br />

GROOT, A.J.L., JANSEN, J.G., VAN WALKENBURG, C.F.M. and ZEELAND,<br />

A.A., 1994, Molecular dosimetry <strong>of</strong> 7-alkyl- and O 6 -alkylguanine in DNA by<br />

electrochemical detection, Mutat. Res., 307, 61–6.<br />

GUPTA, R.C., 1985, Enhanced sensitivity <strong>of</strong> 32 P-postlabelling analysis <strong>of</strong> aromatic<br />

carcinogen: DNA adducts, Cancer Res., 45, 5656–62.<br />

IARC, 1982, Monographs <strong>of</strong> the carcinogenic risk <strong>of</strong> chemicals to humans: some<br />

industrial chemicals and dyestuffs. 3,3′-Dichlorobenzidine and its<br />

hydrochloride, International Agency for the Research on Cancer, Vol. 29, pp.<br />

239–56.<br />

JAIN, R. and SHARMA, M., 1993, Fluorescence postlabelling assay <strong>of</strong> DNA<br />

damage induced by N-methyl-nitrosourea, Cancer Res., 53, 2771–4.


88 METHODS FOR THE DETERMINATION OF REACTIVE COMPOUNDS<br />

LAWLEY, P.D., 1976, Carcinogenesis by alkylating agents, in Searle C.E. (Ed.),<br />

Chemical Carcinogens, pp. 83–244, ACS Monograph 173, Washington, DC:<br />

American Chemical Society.<br />

LEE, M.L. and SANTELLA, M., 1988, Quantitation <strong>of</strong> protein adducts as a<br />

marker <strong>of</strong> genotoxic exposure: immunologic detection <strong>of</strong> benzo[a]pyreneglobin<br />

adducts in mice, Carcinogenesis, 9, 1773–7.<br />

LIN, D.-X., LAY, J.O. JR, BRYANT, M. and KADLUBAR, F.F., 1991, Analysis <strong>of</strong><br />

4-aminobiphenyl-DNA adducts by alkaline hydrolysis and negative ion GC/<br />

MS, in Biomonitoring and Susceptibility Markers in Human Cancer:<br />

Applications in Molecular Epidemiology and Risk Assessment, P-20, Haway,<br />

USA: KailuaKono.<br />

LUTZ, W.K., 1979, In vivo covalent binding <strong>of</strong> organic chemicals to DNA as a<br />

quantitative indicator in the process <strong>of</strong> chemical carcinogenesis, Mutat. Res.,<br />

65, 289–356.<br />

MAGEE, P.N., PEGG, A.E. and SWANN, P.F., 1975, Molecular mechanisms <strong>of</strong><br />

chemical carcinogenesis, in Grundmann E. (Ed.), Handbuch der allgemeinen<br />

Pathologie, Vol. VI/6, pp. 329–419, Berlin: Springer.<br />

MINNETIAN, O., SAHA, M. and GIESE, R.W., 1987, Oxidation-elimination <strong>of</strong> a<br />

DNA base from its nucleoside to facilitate determination <strong>of</strong> alkyl chemical<br />

damage to DNA by GC/MS with electrophore detection, J.Chromatogr., 410,<br />

453–7.<br />

PERERA, F., SANTELLA, R. and POIRIER, M., 1986, Biomonitoring <strong>of</strong> workers<br />

exposed to carcinogens: immunoassay to benzo[a]pyrene-DNA adducts as a<br />

prototype, J. Occup. Med., 28, 1117–23.<br />

POIRIER, M.C., 1993, Antisera specific for carcinogen-DNA adducts and<br />

carcinogenmodified DNA: applications for detection <strong>of</strong> xenobiotics in<br />

biological samples, Mutat. Res., 288, 31–8.<br />

RAHN, R.O., CHANG, S.S., HOLLAND, J.M. and SHUGART, L.R., 1982, A<br />

fluorimetric HPLC assay for quantitating the binding <strong>of</strong> benzo[a]pyrene<br />

metabolites to DNA, Biochem. Biophys. Res. Commun., 109, 262–8.<br />

REDDY, M.V. and RANDERATH, K., 1986, Nuclease P1-mediated enhancement<br />

<strong>of</strong> sensitivity <strong>of</strong> 32 P-postlabelling test for structurally diverse DNA adducts,<br />

Carcinogenesis, 7, 1543–51.<br />

SABBIONI, G., 1992, Quantitative structure activity relationship <strong>of</strong> the Noxidation<br />

<strong>of</strong> aromatic amines, Chem.-Biol. Interact., 81, 91–117.<br />

SABBIONI, G., 1994, Haemoglobin binding <strong>of</strong> nitroarenes and quantitative<br />

structure activity relationships, Chem. Res. Toxicol, 7, 267–74.<br />

SANTELLA, R.M., 1988, Application <strong>of</strong> new techniques for the detection <strong>of</strong><br />

carcinogen adducts to human population monitoring, Mutat. Res., 205, 271–<br />

82.<br />

SANTELLA, R.M., LIN, C.D. and DHARMARAJA, N., 1986, Monoclonal<br />

antibodies to a benzo[a]pyrene diolepoxide modified protein, Carcinogenesis,<br />

7, 441–4.<br />

SHARMA, M. and JAIN, R., 1991, Nuclease P1-mediated fluorescence<br />

postlabelling assay <strong>of</strong> AAF modified DNA model d(TACGTA) and calf-thymus<br />

DNA, Biochem. Biophys. Res. Commun., 177, 151–8.


P.SAGELSDORFF 89<br />

SHUGART, L. and KAO, J., 1985, Examination <strong>of</strong> adduct formation in vivo in the<br />

mouse between benzo[a]pyrene and DNA <strong>of</strong> skin and haemoglobin <strong>of</strong> red<br />

blood cells. Environm. Health Perspect., 62, 223–6.<br />

VAN SITTERT, N.J., DE JONG, G., CLARE, M.G., DAVIES, R., DEAN, B.J.,<br />

WREN, L.J. and WRIGHT, A.S., 1985, Cytogenetic, immunological and<br />

haematological effects in workers in an ethylene oxide manufacturing plant,<br />

Br. J. Ind. Med., 42, 19–46.<br />

SKIPPER, P.L. and TANNENBAUM, S.R., 1990, Protein adducts in molecular<br />

dosimetry <strong>of</strong> chemical carcinogens, Carcinogenesis, 11, 507–18.<br />

STEINER, S., CRANE, A.E. and WATSON, W.P., 1992a, Molecular dosimetry <strong>of</strong><br />

DNA adducts in C3H mice treated with glycidaldehyde, Carcinogenesis, 13,<br />

119– 24.<br />

STEINER, S., HÖNGER, G. and SAGELSDORFF, P., 1992b, Molecular dosimetry<br />

<strong>of</strong> DNA adducts in C3H mice treated with bisphenol A diglycidylether,<br />

Carcinogenesis, 13, 969–72.<br />

TÖRNQVIST, M., MOWRER, J., JENSEN, S. and EHRENBERG, L., 1986,<br />

Monitoring <strong>of</strong> environmental cancer initiators through haemoglobin adducts<br />

by a modified Edman degradation method, Anal. Biochem., 154, 255–66.<br />

WEISSBURGER, J.H. and WILLIAMS, G.N., 1975, Metabolism <strong>of</strong> chemical<br />

carcinogens, in Becker, F.F. (Ed.), Cancer, Vol. 1, pp. 185–234, New York:<br />

Plenum.<br />

WESTON, A., BOWMAN, E.D., ROWE, M.L., MANCHESTER, D.K. and<br />

HARRIS, C.C., 1989, Fluorescence and mass spectral evidence for the<br />

formation <strong>of</strong> benzo[a]pyrene anti-diol-epoxide-DNA and -haemoglobin<br />

adducts in humans, Carcinogenesis, 10, 251–7.


PART THREE<br />

Pulmonary toxicology <strong>of</strong> industrial<br />

chemicals


7<br />

Studies to Assess the Carcinogenic Potential <strong>of</strong><br />

Man-Made Vitreous Fibers<br />

THOMAS W.HESTERBERG, GERALD R.CHASE,<br />

RICHARD A.VERSEN and ROBERT ANDERSON<br />

Schuller International, Inc., Littleton, CO<br />

Introduction<br />

Man-made vitreous fibers (MMVFs) are a class <strong>of</strong> materials which have<br />

found many applications in both residential and industrial settings. MMVFs<br />

are fibrous inorganic substances that are made primarily from rock, clay,<br />

slag or glass. Sometimes referred to as man-made mineral fibers<br />

(MMMFs), the major classes <strong>of</strong> MMVF are refractory ceramic fibers<br />

(RCFs), fibrous glass, rock (stone) wool and slag wool.<br />

RCF, the smallest category <strong>of</strong> MMVF, represents only about 1–2 per<br />

cent <strong>of</strong> the world production <strong>of</strong> MMVF. It is made by melting Al 2O 3 and<br />

SiO 2 in about equal amounts or by melting kaolin clay and then ‘spinning’<br />

or ‘blowing’ this molten material into fibers. Most RCF is used as a high<br />

temperature furnace insulation. World production <strong>of</strong> RCF in 1990 was<br />

about 80 million 1b. Fibrous glass is the largest category <strong>of</strong> the MMVFs<br />

and is used in insulation, air handling, filtration and sound absorption. The<br />

thermal, acoustical and fire resistant properties <strong>of</strong> these products have led<br />

to their widespread use in a variety <strong>of</strong> residential and commercial<br />

applications. Production <strong>of</strong> fibrous glass in North America in 1989 was<br />

approximately 1.8 million t. Slag and rock wool are composed primarily <strong>of</strong><br />

calcium, magnesium, aluminum and silica. Since 1975, most slag wool has<br />

been produced from the waste slag that resulted from the reduction <strong>of</strong> iron<br />

ore to iron. Rock wool fibers are made from basaltic rocks with additives<br />

such as limestone or dolomite. Slag and rock wool are used in residential<br />

and commercial low and high temperature insulation and in acoustical<br />

ceiling tiles and wall panels. About 75% <strong>of</strong> slag wool production is used in<br />

acoustical ceiling tile manufacture in North America.<br />

Animal studies and epidemiological studies have been conducted to<br />

assess the potential biological effects <strong>of</strong> MMVFs. This research has been<br />

reviewed by the International Agency for Research on Cancer (IARC, 1988),<br />

the International Programme on Chemical Safety (IPCS, 1988), and the US<br />

Environmental Protection Agency (Vu, 1988). These reviews are consistent


92 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

in the judgement that chronic inhalation studies <strong>of</strong> airborne fibers provide<br />

the best model for assessing potential risk to man (McClellan et al., 1992).<br />

In assessing the carcinogenic risks <strong>of</strong> exposure to any possible<br />

occupational hazard, research is pursued through several different scientific<br />

techniques. Studies <strong>of</strong> mortality (analysis <strong>of</strong> death rates) are used to<br />

evaluate the potential carcinogenicity associated with direct human<br />

exposure. Animal exposure studies are used to not only evaluate the<br />

potential carcinogenicity but to also investigate the mechanisms <strong>of</strong> disease<br />

development. <strong>Industrial</strong> hygiene and engineering studies are used for<br />

quantifying exposures.<br />

Epidemiological studies<br />

By general agreement among experts (IARC, 1988; IPCS, 1988), two major<br />

historical cohort studies are considered to have comprehensively addressed<br />

the mortality experience <strong>of</strong> workers engaged in the production <strong>of</strong> FG, rock<br />

wool and slag wool: a European study conducted by the International<br />

Agency for Research on Cancer (IARC), and a University <strong>of</strong> Pittsburgh<br />

study conducted in the USA. The discussion here will concentrate on those<br />

two studies. For a summary <strong>of</strong> other studies, the reader is referred to the<br />

IARC review (IARC, 1988). There are no published reports <strong>of</strong> the<br />

mortality experience <strong>of</strong> RCF workers. Epidemiological studies <strong>of</strong> workers<br />

engaged in the manufacture <strong>of</strong> all major classes <strong>of</strong> MMVF are underway or<br />

are continuing. Morbidity studies <strong>of</strong> the respiratory health <strong>of</strong> workers are<br />

not discussed here.<br />

The IARC study<br />

IARC researchers reported their study at the WHO Occupational Health<br />

Conference on the Biological Effects <strong>of</strong> Man-Made Mineral Fibres at<br />

Copenhagen in 1982, with a follow-up in 1986 (Simonato et al., 1987). The<br />

updated study is also published in the Scandinavian Journal <strong>of</strong> Work,<br />

Environment & Health, Volume 12, Supplement 1, 1986. The mortality <strong>of</strong><br />

23609 workers (2836 deaths) employed in 13 European factories engaged<br />

in the production <strong>of</strong> MMVF (including 11 852 fibre glass production<br />

workers at six plants in five countries and 10 115 rock wool/slag wool<br />

production workers at seven plants in four countries) has been studied<br />

(Saracci et al., 1984) and updated (Simonato et al., 1987). The authors<br />

reported an ‘excess <strong>of</strong> lung cancer among rock-wool/slag workers<br />

employed during an early technological phase before the introduction <strong>of</strong><br />

dust-suppressing agents’, and concluded that ‘fiber exposure, either alone or<br />

in combination with other exposures, may have contributed to the elevated<br />

risk’. The authors also reported that ‘no excess <strong>of</strong> the same magnitude was<br />

evident for glass-wool production, and the follow-up <strong>of</strong> the continuous-


filament cohort was too short to allow for an evaluation <strong>of</strong> possible longterm<br />

effects’. It was also noted that ‘there was no evidence <strong>of</strong> an increased<br />

risk for pleural tumors or non-malignant respiratory diseases’. An update <strong>of</strong><br />

this study is underway.<br />

The University <strong>of</strong> Pittsburgh study<br />

This study was also reported at the WHO Occupational Conference on<br />

Biological Effects <strong>of</strong> Man-Made Mineral Fibres at Copenhagen in 1982 and<br />

the follow-up Conference in 1986 (Simonato et al., 1987). Subsequent to<br />

the 1986 Conference, additional analyses were completed and included in<br />

the manuscript published for the proceedings (Enterline et al., 1987). The<br />

study has been updated and published (Marsh et al., 1990). The University<br />

<strong>of</strong> Pittsburgh researchers’ comprehensive mortality review <strong>of</strong> more than<br />

16000 workers— many with long-term exposure up to 40 years—was<br />

undertaken at 17 US fiber-glass, rock-wool and slag-wool manufacturing<br />

plants, including 14800 fiber-glass workers in 11 plants. The original<br />

report, given in 1982, covered the mortality experience from the 1940s to<br />

the end <strong>of</strong> 1977. The same group <strong>of</strong> workers was followed through 1982<br />

(reported in October 1986, with additional analyses available in June<br />

1987). The June 1987 report contained, for the first time, local area<br />

mortality statistics for each <strong>of</strong> the plants as the basis for studying the<br />

mortality experience. Experts agree that, barring unusual circumstances,<br />

local area comparisons are most appropriate. The study has been further<br />

updated through 1985, with publication in 1990. For respiratory cancer, in<br />

the latest update there was a small but statistically significant increase for<br />

fiber glass production workers. However, aside from the issue <strong>of</strong><br />

uncontrolled potential confounding, the study provides no evidence to date<br />

that respiratory cancer mortality is related to fiber glass exposure. There<br />

was a somewhat larger statistically significant excess <strong>of</strong> respiratory cancer<br />

mortality reported for slag wool and rock wool production workers. The<br />

absence <strong>of</strong> any clear exposure-response relationship for any <strong>of</strong> the fiber<br />

groups studied led the authors to conclude that ‘overall, the evidence <strong>of</strong> a<br />

relationship between exposure to man-made mineral fibers and respiratory<br />

cancer appears to be somewhat weaker than in the previous update’.<br />

Consistent with the IARC study, no increase in the occurrence <strong>of</strong><br />

mesothelioma has been observed in this cohort. This study has now been<br />

expanded to include well over 30000 workers from 14 fiber-glass and six<br />

rock wool and slag wool facilities.<br />

Other epidemiological studies<br />

T.W.HESTERBERG ET AL. 93<br />

In addition to the two major studies highlighted above, a number <strong>of</strong> other<br />

studies have been conducted as well. Many <strong>of</strong> them widely overlap these


94 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

major studies, comprise sub-groups within them, or represent smaller<br />

worker populations outside <strong>of</strong> them.<br />

A Canadian study was reported by Shannon at the WHO Occupational<br />

Health Conference on Biological Effects <strong>of</strong> Man-Made Mineral Fibers at<br />

Copenhagen in 1982 and 1986 (Shannon et al., 1987). It followed 2557<br />

male workers at a Canadian glass wool plant through 1977 and was later<br />

updated to extend the follow-up to the end <strong>of</strong> 1984. In the updated study,<br />

the authors reported a statistically significant excess <strong>of</strong> lung cancer. In<br />

discussing this excess, the authors concluded that the interpretation <strong>of</strong> the<br />

information was difficult since there was no relationship between the<br />

excess <strong>of</strong> lung cancer and the length <strong>of</strong> time since first exposure to the<br />

fibrous glass manufacturing environment.<br />

Two recent case-control studies have addressed the lung cancer mortality<br />

<strong>of</strong> FG and slag wool production workers. Chiazze et al. (1992) have<br />

investigated the potential impact <strong>of</strong> confounding factors such as smoking<br />

and other occupational exposures for workers at the oldest and largest US<br />

fiber glass manufacturing facility. In particular, Chiazze helped clarify the<br />

heavy smoking patterns in those workers and verified the large impact that<br />

smoking has on their lung cancer experience. Wong et al., (1991)<br />

investigated the potential impact <strong>of</strong> smoking on the lung cancer deaths at<br />

nine US slag wool manufacturing plants. Wong also found heavy smoking<br />

among the slag wool workers and advanced the understanding <strong>of</strong> the<br />

modest increase in lung cancer seen in the historical cohort studies cited<br />

above.<br />

Users <strong>of</strong> MMVFs generally have experienced mixed exposures, making<br />

the study <strong>of</strong> any potential health effects <strong>of</strong> MMVF difficult, if possible at<br />

all. For example, in a study <strong>of</strong> Swedish construction workers, Engholm et al.<br />

(1987) discussed the difficulty caused by overlapping <strong>of</strong> reported exposures<br />

to asbestos and MMVFs. In addition, essential employment and exposure<br />

histories for users <strong>of</strong> MMVFs are lacking.<br />

The mortality studies <strong>of</strong> FG workers, while showing a small but<br />

statistically significant increase in lung cancer, have failed to show any<br />

consistent relationship with exposure to FG (i.e. no dose-response<br />

relationships have been found). It is recognized that uncontrolled<br />

occupational and/or non-occupational confounding factors may be<br />

associated with the slight increase. The IARC review (IARC, 1988)<br />

concluded that there is ‘inadequate evidence’ for carcinogenicity in<br />

humans. Other reviews have reached similar conclusions. In addition,<br />

reports subsequent to the IARC review have further clarified potential<br />

confounding factors and, if anything, shown weaker evidence <strong>of</strong> a<br />

relationship between exposure and lung cancer.<br />

The cohort mortality studies <strong>of</strong> rock wool and slag wool workers have<br />

shown a somewhat larger statistically significant excess <strong>of</strong> lung cancer<br />

deaths, but have also provided no clear dose-response relationship with


fiber exposure. While the IARC review (IARC, 1988) concluded that there<br />

is ‘limited evidence’ for carcinogenicity in humans, reports subsequent to<br />

the IARC review have further clarified potential confounding factors and,<br />

if anything, shown weaker evidence <strong>of</strong> a relationship between exposure and<br />

lung cancer.<br />

Experimental studies<br />

Toxicologic studies <strong>of</strong> MMVFs have been conducted in both in vitro and in<br />

vivo systems. In addition, the physical and chemical characteristics thought<br />

to correlate with toxicity have been examined. The in vitro studies have<br />

been conducted using cells from the lungs <strong>of</strong> animals as well as bacterial<br />

and cell lines. Two categories <strong>of</strong> whole-animal studies have been reported:<br />

studies using artificial methods to implant high concentrations <strong>of</strong> fibers in<br />

the abdomen, pleura or trachea <strong>of</strong> animals; and inhalation studies <strong>of</strong><br />

maximum tolerated doses and multiple dose levels <strong>of</strong> fibers.<br />

Cell culture studies<br />

T.W.HESTERBERG ET AL. 95<br />

The use <strong>of</strong> cell culture systems for studying the toxic effects <strong>of</strong> fibers has<br />

been recently reviewed (Hesterberg et al., 1993a). A number <strong>of</strong> studies<br />

have shown that fiber length and diameter are important in determining<br />

the toxicity <strong>of</strong> mineral fibers <strong>of</strong> various chemical compositions to cells<br />

grown in culture (Chamberlain et al., 1979, Tilkes and Beck, 1980;<br />

Hesterberg and Barrett, 1984; Hesterberg et al., 1993a; Hart et al., 1994).<br />

Chemical composition has also been shown to be critical to the toxicity <strong>of</strong><br />

fibers to rat tracheal epithelial cells (Ririe et al., 1985) and human<br />

bronchial epithelial cells grown in culture (Kodama et al., 1993). MMVFs<br />

have also been shown to induce neoplastic transformation (Hesterberg and<br />

Barrett, 1984; Poole et al., 1986) and genetic damage to cells in culture<br />

(Sincock and Seabright, 1975; Oshimura et al., 1984). Cell culture models<br />

are important for understanding the mechanisms <strong>of</strong> fiber toxicity and, with<br />

further development, have potential for use as part <strong>of</strong> a battery <strong>of</strong> shortterm<br />

screening tests to assess the toxic and tumorigenic potential <strong>of</strong><br />

mineral fibers. However, it was recently shown that cytotoxicity <strong>of</strong> different<br />

compositions MMVFs to Chinese hamster ovary (CHO) cells in culture did<br />

not correlate with the in vivo toxicity <strong>of</strong> theses MMVFs (Hart et al., 1994).<br />

This may be related to CHO cells being aneuploid, preneoplastic and not a<br />

normal target cell for fiber toxicity in vivo. Future in vitro studies <strong>of</strong> MMVF<br />

toxicity should focus on the use <strong>of</strong> cell types that represent the relevant<br />

target tissues, and cells should be as close to normal as possible.


96 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

Implantation studies<br />

Using various types and dimensions <strong>of</strong> fibers, researchers have studied the<br />

effects <strong>of</strong> ‘artificially’ exposed animals by surgically implanting fibrous<br />

material in the pleural (chest) and abdominal cavities <strong>of</strong> laboratory<br />

animals, and by injecting fibers directly into the trachea (Pott et al., 1987;<br />

Stanton et al., 1981). Those studies have shown that high levels <strong>of</strong> most<br />

fibrous materials <strong>of</strong> certain dimensions, regardless <strong>of</strong> their physical or<br />

chemical makeup, can induce tumors in laboratory animals. From these<br />

study results, scientists have also hypothesized that biological activity<br />

correlates with fiber length and diameter, since ‘long, thin’ fibers are the<br />

most active. The actual chemical composition appears to play only a minor<br />

role, if any, in such ‘artificial exposure’ experiments (Stanton et al., 1981).<br />

Injection <strong>of</strong> fibers bypasses the normal defense mechanisms <strong>of</strong> the lung<br />

and can produce abnormal fiber distribution, fiber clumping, and overload<br />

doses (McClellan et al., 1992). Furthermore, when fibers are injected into<br />

the pleura or peritoneum <strong>of</strong> an animal, leaching, degradation,<br />

fragmentation or any other transformations are unlikely to be the same as<br />

after inhalation. The weaknesses <strong>of</strong> intracavitary injection studies <strong>of</strong><br />

fibrous materials limit their relevance for human risk assessment (IPCS,<br />

1988; Vu, 1988; Dement et al., 1990; WHO, 1992; McClellan et al.,<br />

1992).<br />

Recent animal inhalation studies <strong>of</strong> MMVFs<br />

Inhalation is the only natural route <strong>of</strong> exposure for fiber entry and<br />

distribution to the target organs in man. Animal inhalation studies are<br />

more relevant than intracavity administration studies for risk assessment<br />

because the exposure conditions <strong>of</strong> inhalation experiments more closely<br />

approach the circumstances <strong>of</strong> human exposure.<br />

Background<br />

In June 1988, a series <strong>of</strong> inhalation studies was initiated at Research and<br />

Consulting Company (RCC) in Geneva, Switzerland, to evaluate the<br />

biological effects <strong>of</strong> different compositions <strong>of</strong> MMVF. These included<br />

RCFs, common insulation fiber glass, and rock and slag wool fibers. These<br />

studies used recently perfected state-<strong>of</strong>-the-art technologies for fiber sizeseparation,<br />

fiber l<strong>of</strong>ting and nose-only inhalation exposure. A more<br />

detailed description <strong>of</strong> the techniques used and the results from these<br />

studies are found elsewhere (Hesterberg et al., 1991, 1993b; Mast et al.,<br />

1995 McConnell et al., 1994). The animal models selected were those with<br />

demonstrated capacity to develop asbestosrelated disease following<br />

inhalation exposure. The studies were conducted in accordance with


standard techniques for chronic toxicity/carcinogenicity studies, including<br />

dose and latency considerations. Rats were exposed for 2 years and<br />

hamsters for 18 months. The animals were observed for their lifetime or until<br />

20% survival <strong>of</strong> the test group was reached. Positive and shamexposed<br />

negative controls were included in the protocol.<br />

Fiber aerosols<br />

In designing these animal inhalation studies, the techniques <strong>of</strong> fiber<br />

preparation, aerosolization, exposures, measurement, quantification and<br />

determination <strong>of</strong> actual target organ dose were critical factors. Fiber<br />

dimensions that permitted deposition into the distal lung regions (i.e.<br />

respirable fibers) for the model used were selected. The characteristics <strong>of</strong> the<br />

fiber aerosol in actual work areas for man was an important consideration<br />

in determining experimental exposure. For example, an average fiber size<br />

<strong>of</strong> 1×20 µm has been measured during simulated RCF work practices. The<br />

critical need to use fibers pre-selected for their size and to verify the actual<br />

size distributions <strong>of</strong> the fiber exposure aerosol was met throughout the<br />

study. Non-fibrous particles (shot) in the aerosol were reduced to the<br />

maximum extent possible. Furthermore, fiber preparation, handling and<br />

aerosolization did not alter the physical-chemical characteristics <strong>of</strong> the<br />

fiber, since as will be discussed later, these are known to be critical<br />

determinants <strong>of</strong> fiber toxicity.<br />

Nose-only rather than whole-body exposure was used for several<br />

reasons, including the impossibility <strong>of</strong> preparing the huge quantities <strong>of</strong><br />

specially sized fibers that would be required for 2 years <strong>of</strong> whole-body<br />

exposure. Additionally, nose-only exposure levels permitted better control<br />

<strong>of</strong> exposure levels and host entry.<br />

Selection <strong>of</strong> exposure concentrations<br />

It was important that at least three exposure concentrations be used in the<br />

chronic inhalation study in order to assess the dose-response relationships<br />

<strong>of</strong> any induced changes. The highest concentration selected was the<br />

‘Maximum tolerated dose’ (MTD), while lower concentrations were 50 per<br />

cent <strong>of</strong> the MTD and multiples <strong>of</strong> the projected occupational and<br />

environmental exposure levels.<br />

Experimental design, time lines<br />

T.W.HESTERBERG ET AL. 97<br />

Groups <strong>of</strong> three or six randomly selected animals from each exposure<br />

group were killed at 3, 6, 12, 18 and 24 (rats only) months to follow the<br />

progression <strong>of</strong> histopathological changes and to determine lung fiber<br />

burdens. An additional six ‘recovery’ animals were removed from each


98 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

exposure group at 3, 6, 12 and 18 (rats only) months and held without<br />

further treatment until the end <strong>of</strong> the exposure period, when they were<br />

killed to assess progression or regression <strong>of</strong> lung lesions and lung retention<br />

and clearance <strong>of</strong> fibers after cessation <strong>of</strong> exposure. To assure quality<br />

control, the l<strong>of</strong>ting technique and exposure level were consistently<br />

monitored during the study by both gravimetric measurement and fiber<br />

counting techniques. The terminal sacrifice was carried out when only 20 per<br />

cent <strong>of</strong> the animals survived. A complete necropsy was performed on each<br />

animal. Gross pathological examination and diagnoses were performed<br />

using a dissecting microscope. Uniform sections <strong>of</strong> the left lung and right<br />

diaphragmatic lobe were embedded in paraffin, cut at a thickness <strong>of</strong> 4 mm<br />

and replicate sections were routinely stained with hematoxylin and eosin<br />

(H&E) and Masson-Goldner’s trichrome stain for collagen staining to<br />

assess the presence <strong>of</strong> lung fibrosis. In addition, sections were made from<br />

all grossly visible lesions from that and other portions <strong>of</strong> the lung.<br />

Proliferative lesions <strong>of</strong> the pulmonary parenchyma were designated as<br />

bronchoalveolar hyperplasia, pulmonary adenoma or adenocarcinoma.<br />

Other types <strong>of</strong> lesions, including those in the pleura were noted where<br />

appropriate. All research and analyses were conducted using good<br />

laboratory practices.<br />

Lung fiber burden<br />

Immediately after necropsy, the infracardiac lobe <strong>of</strong> each animal’s lung was<br />

removed and frozen for later analysis <strong>of</strong> lung fiber burden. To recover<br />

fibers from the lung, the tissue was rapidly dehydrated with acetone and<br />

ashed using a low-temperature process. Recovered fibers were dispersed in<br />

distilled water and examined using scanning electron microscopy. Number,<br />

dimensions and other physical characteristics <strong>of</strong> the inhaled lung fibers<br />

were determined, and reported as fibers per mg <strong>of</strong> dry lung weight.<br />

Results from recent animal inhalation studies <strong>of</strong> MMVFs<br />

Refractory ceramic fibers<br />

In the first RCC studies, rodents were exposed to the MTD <strong>of</strong> the sizeselected<br />

RCF test fiber, 30 mg m −3 and approximately 200–250 fibers cm<br />

−3 . Rats were exposed for 6 h per day, 5 days a week to aerosols containing<br />

one <strong>of</strong> four different types <strong>of</strong> RCF: kaolin, RCF 1; zirconia, RCF 2; high<br />

purity kaolin, RCF 3; and ‘after service’ (a kaolin based ceramic fiber<br />

containing 27% crystalline silica that had previously been exposed to high<br />

temperature), RCF 4. Hamsters were exposed to only kaolin RCF fibers.<br />

Positive controls (chrysotile asbestos) and negative controls (filtered air)


Table 7.1 Summary <strong>of</strong> lung pathology findings in RCF hamster inhalation study<br />

a WHO fibers=fibers with length/diameter ≥3, length >5 µm, and diameter


100 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

Table 7.2 Summary <strong>of</strong> lung pathology findings in RCF MTD (30 mg m−3) rat<br />

inhalation<br />

a WHO fibers=fibers with length/diameter ≥3, length >5 µm, and diameter


asbestos (10 mg m −3 ) were included in the study. The crocidolite exposure<br />

had to be stopped at 10 months due to excessive mortality resulting from<br />

lung toxicity. The results are summarized in Table 7.5. Crocidolite<br />

exposure resulted in lung fibrosis, a significant increase in lung tumors, and<br />

a single mesothelioma. Rock wool, but not slag wool, exposure at 16 and<br />

30 mg m −3 resulted in minimal lung fibrosis. However, neither rock wool<br />

nor slag wool exposure resulted in mesotheliomas or a significant increase<br />

in lung tumors.<br />

Lung burden analyses<br />

T.W.HESTERBERG ET AL. 101<br />

Table 7.3 Combined summary <strong>of</strong> lung pathology findings: chrysotile and RCF1<br />

from RCF MTD study in rats; and RCF multidose study in rats<br />

a WHO fibers=fibers with length/diameter ≥3, length >5 µm, and diameter


102 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

Table 7.4 Summary <strong>of</strong> lung pathology findings in fibrous glass inhalation study in<br />

rats<br />

a WHO fibers=fibers with length/diameter ≥3, length >5 µm, and diameter 5 µm, and a diameter 10 µm in length in the lung were similar for each <strong>of</strong> the<br />

different MMVF types (Figures 7.2(a) and 7.2(b)). However, greater<br />

numbers <strong>of</strong> long fibers (>20 µm. long) were found in the lungs <strong>of</strong> rats<br />

exposed to RCF 1 and MMVF 21 (rock wool) than for the other fiber<br />

types (Figure 7.2(c)). Even though lung levels <strong>of</strong> long MMVF 21 fibers<br />

were higher than long RCF 1 fibers, lung fibrosis occurred much later for<br />

MMVF 21 (18 vs 6 months for RCF 1) and no mesotheliomas or significant<br />

increase in lung tumors were observed for MMVF 21. This indicates that<br />

the lung pathogenic potential <strong>of</strong> a fiber may be determined by more than<br />

dose and dimension.


T.W.HESTERBERG ET AL. 103<br />

Table 7.5 Summary <strong>of</strong> lung pathology findings in rock and slag wool inhalation<br />

study in rats<br />

a WHO fibers=fibers with length/diameter ≥3, length >5 µm, and diameter


104 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

disappearance <strong>of</strong> long fibers. More studies are required to determine if a<br />

fiber’s ability to be leached is a critical determinant <strong>of</strong> its ultimate toxicity<br />

to the lung.<br />

Figure 7.1 Length distributions (a) and diameter distributions (b) <strong>of</strong> fibers from the<br />

lungs <strong>of</strong> rats exposed for 13 weeks to the five different MMVFs in the chronic<br />

inhalation studies. To permit clearance <strong>of</strong> the upper airways, rats were killed a


T.W.HESTERBERG ET AL. 105<br />

minimum <strong>of</strong> 24 h after the exposure was stopped; the right accessory lobe was<br />

frozen and later low temperature ashed for fiber recovery. Fiber lengths were<br />

determined using phase contrast optical microscopy, while fiber diameters were<br />

determined using scanning electron microscopy.<br />

Results from previous MMVF inhalation studies<br />

The results from two previous RCF inhalation studies (Davis et al., 1984;<br />

Smith et al., 1987) differ from the more recent RCC studies presented here.<br />

Davis et al., (1984) reported RCF exposure <strong>of</strong> rats resulted in an average <strong>of</strong><br />

5 per cent pulmonary fibrosis, pulmonary tumors in eight <strong>of</strong> 48 rats, and<br />

one peritoneal mesothelioma. The lower fibrosis and tumor response in the<br />

Davis study may have resulted from the lower exposure concentration used;<br />

8.4 mg m −3 compared to 30 mg m −3 in the present study. In addition, the<br />

use <strong>of</strong> fibers that were not presized, the use <strong>of</strong> whole-body exposure, or the<br />

fiber generation technique, which may have crushed some <strong>of</strong> the fibers,<br />

may account for the lack <strong>of</strong> consistency with the present study. Smith et<br />

al., (1987) exposed hamsters and rats to RCF at 200 f cm −3 , 6h a day, 5<br />

days a week, for 24 months. The rat study showed no significant increase<br />

in neoplasms and minimal pulmonary fibrosis in 22% <strong>of</strong> the exposed<br />

animals. In the hamster study, RCF produced only one mesothelioma in 50<br />

animals and no fibrosis was observed. It is difficult to explain why there<br />

was little response to RCF in the Smith studies, but it may be related to the<br />

different aerosol and exposure technology used or to the low exposure<br />

level; 12 mg m −3 compared to 30 mg m −3 in the present study.<br />

Previous inhalation studies <strong>of</strong> FG using rodents agree with the findings<br />

<strong>of</strong> the RCC studies. Fiber glass has been tested by inhalation in guinea pigs<br />

(Gross et al., 1970), hamsters (Lee et al., 1981; Smith et al., 1987), and<br />

rats (Gross et al., 1970; Lee et al., 1981; McConnell et al., 1984; Wagner<br />

et al., 1984; Mitchell et al. 1986; LeBouffant et al., 1987; Muhle et al.,<br />

1987; Smith et al., 1987). None <strong>of</strong> these studies identified a significant<br />

increase in either fibrosis or neoplasms following glass fiber inhalation in<br />

spite <strong>of</strong> FG lung burdens in excess <strong>of</strong> several hundred thousand fibers per<br />

mg dry lung tissue. In three <strong>of</strong> the above studies, the chronic inhalation<br />

toxicity <strong>of</strong> rock and slag wool were also examined (Wagner et al., 1984;<br />

LeBouffant et al., 1987; Smith et al., 1987). As was seen with fibrous glass,<br />

all three studies demonstrated no tumorigenic response by this route <strong>of</strong><br />

exposure.


106 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

Figure 7.2 (continued over) Lung burdens <strong>of</strong> (a) WHO fibers, (b) fibers >10 µm in<br />

length, and (c) fibers >20 µm in length per mg dry lung tissue from the lungs <strong>of</strong> rats<br />

continuously exposed to 30 mg m −3 <strong>of</strong> the five different MMVFs. Rats were killed<br />

at least 24 h after the exposure was stopped, the right accessory lobe was frozen<br />

and later low temperature ashed for fiber recovery.<br />

<strong>Industrial</strong> hygiene studies<br />

RCF<br />

<strong>Industrial</strong> hygiene monitoring data obtained on a regular basis at locations


Figure 7.2 Continued<br />

where RCF products are manufactured show that exposures are generally<br />

below 1.0 f cm −3 , typically below 0.2 f cm −3 . In a recent study, RCF levels<br />

during various end-user operations ranged from 0.12 to 1.55 f cm −3 with<br />

an overall mean and SD <strong>of</strong> 0.74±0.49 f cm −3 (Lees et al., 1993b). Other<br />

end-user studies have indicated that RCF exposures can exceed 5 f cm −3 or<br />

higher if appropriate engineering controls and work practices are not<br />

followed (Schuller, 1985–1988).<br />

Fiber glass<br />

T.W.HESTERBERG ET AL. 107<br />

Recently, studies which examined human aerosol exposure to fiber glass in<br />

manufacturing, installation and removal, and in ambient air were reviewed<br />

(Hesterberg and Hart, 1994). In most cases, human exposures to airborne<br />

fiber glass during manufacturing and installation fell well below the OSHAproposed<br />

permissible exposure limit (PEL) <strong>of</strong> 1 f cm −3 air (OSHA, 1992).<br />

Airborne fiber concentrations during FG manufacturing operations are<br />

typically less than 0.2 f cm −3 , with the majority being less than 0.1 f cm −3 .<br />

Exceptions include manufacture <strong>of</strong> finer diameter fiber glass and blowing<br />

installation <strong>of</strong> loose fiber glass that is either milled or lacks binder. Airborne<br />

levels averaging greater than 1 f cm −3 have been reported in the production<br />

<strong>of</strong> finer diameter fiber glass (TIMA, 1990), while blowing installation <strong>of</strong><br />

loose fiber glass without binder resulted in a task length average (TLA) <strong>of</strong><br />

7.67 f cm −3 , and an 8-h TWA <strong>of</strong> 1.96 f cm −3 (Lees et al., 1993a). Blowing<br />

installation <strong>of</strong> loose mineral wool also resulted in higher aerosol levels; a


108 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

TLA <strong>of</strong> 1.94 f cm −3 and an 8-h TWA <strong>of</strong> 0.97 f cm −3 (Lees et al., 1993a).<br />

Removal <strong>of</strong> fiber glass insulation created an aerosol <strong>of</strong> 0.042 f cm −3 (Jacob<br />

et al., 1993). Fiber concentrations <strong>of</strong> 0.004 f cm −3 were reported for<br />

buildings recently insulated with FG (Jacob et al., 1992). However this figure<br />

includes all types <strong>of</strong> fibers as it was obtained using optical microscopy. The<br />

background level prior to fiber glass installation was 0.001 f cm −3 .<br />

Ambient environmental exposures to airborne vitreous fibers were<br />

extremely low; exposure levels <strong>of</strong> product-related vitreous fibers reported<br />

for outdoor air was 0.0007 f cm −3 (Tiesler and Draeger, 1994).<br />

In addition to manufacturing and field use surveys, release <strong>of</strong> fibrous<br />

glass during actual use <strong>of</strong> products, particularly fiber released from air<br />

filter media, has been monitored. To determine possible exposure <strong>of</strong><br />

building occupants to fibrous glass, ambient air was sampled in a number<br />

<strong>of</strong> public buildings in which fibrous glass air filtration products had been<br />

installed. These evaluations showed no significant release <strong>of</strong> fibers from the<br />

filters (Balzer et al., 1971; Cholak and Schafer, 1971).<br />

To evaluate the efficiency <strong>of</strong> fibrous glass filter blankets, several high<br />

volume air samples were collected at various points in the ductwork <strong>of</strong> a<br />

large <strong>of</strong>fice complex at the intake and the exhaust prior to changing the<br />

filter media, and at the exhaust 23 days after installation <strong>of</strong> the new filter.<br />

Analyses <strong>of</strong> the samples using electron microscopy indicate little initial<br />

fiber release which decreases rapidly thereafter to the limit <strong>of</strong> detection<br />

(Schuller, 1987).<br />

Rock and slag wool<br />

Airborne concentrations <strong>of</strong> dust and fibers reported from US mineral wool<br />

plants is generally higher than in US glass wool facilities. This includes both<br />

airborne fibers and total particulate matter. Fiber levels reported ranged<br />

from 0.01 to 1.4 f cm −3 , compared with 0.1–0.3 f cm −3 for glass wool.<br />

Total particulate matter sample results ranged from 0.05 to 23.6 mg m −3 in<br />

the mineral wool facilities and 0.09–8.48 mg m −3 for glass wool (Esmen et<br />

al., 1980).<br />

Comparison <strong>of</strong> Human MMVF exposures used in the<br />

recent rat chronic inhalation studies<br />

When using animal inhalation studies for assessment <strong>of</strong> potential risk to<br />

human health <strong>of</strong> airborne fibers, it is critical to demonstrate that the<br />

characteristics and concentrations <strong>of</strong> the experimental fiber aerosols are<br />

comparable with those in human exposure situations. It is also important<br />

for risk assessment that the actual target organ dose in the animal model<br />

reach or exceed that found in exposed humans. To illustrate, consider<br />

levels <strong>of</strong> fiber glass published in a number <strong>of</strong> recent reports. A qualitative


T.W.HESTERBERG ET AL. 109<br />

Table 7.6 Representative airborne levels <strong>of</strong> fiber glass in workplace and rat<br />

inhalation study<br />

a Outdoor data from Tiesler and Draeger (1994). Product-related fibers counted<br />

using NIOSH A Rules.<br />

b Data from Jacob et al., (1993). Airborne levels resulting from manufacturing<br />

operations using FG insulation.<br />

c Data from Lees et al., (1993a). Installation <strong>of</strong> residential insulation.<br />

d Jacob et al. (1992) reported that levels returned to background within hours after<br />

Batt installation.<br />

All other data are averages from the various studies herein cited.<br />

and quantitative comparison was made <strong>of</strong> the aerosol and lung fibers in the<br />

rat inhalation study with those in various human exposure situations<br />

(Hesterberg and Hart, 1994). A comparison <strong>of</strong> the reported aerosol fiber<br />

levels in various human settings with those used in the rat inhalation study<br />

is shown in Table 7.6. FG levels in the rat aerosol were more than five<br />

orders <strong>of</strong> magnitude higher than the reported level for outdoor air, and at<br />

least three orders <strong>of</strong> magnitude higher than for average airborne levels for<br />

many occupational settings (e.g. over 2000fold higher than FG batt<br />

installation). The rat aerosol was 75-fold more concentrated than the<br />

highest reported average TWA for airborne fiber levels in an occupational<br />

setting, i.e. blowing installation <strong>of</strong> unbound fiber glass (the potential for<br />

higher airborne levels has been recognized for some time, and<br />

recommended work practices call for the use <strong>of</strong> respirators in such<br />

circumstances). Despite the range in products and occupational settings,<br />

fiber dimensions in most <strong>of</strong> the human exposures examined were fairly<br />

similar to those found in the rat inhalation study aerosol (Hesterberg and<br />

Hart, 1994). The fiber dimensions <strong>of</strong> aerosolized rock and slag wool<br />

collected from workplace air during the installation <strong>of</strong> batts or blowing <strong>of</strong><br />

loose fibers have similar mean diameters to that <strong>of</strong> fiber glass (1.0–1.6<br />

µm). However, the mean lengths appear to be greater (30–50 µm) than for<br />

most workplace samples <strong>of</strong> fiber glass.<br />

Hesterberg and Hart (1994) also compared the lung burdens <strong>of</strong> rats<br />

exposed in the recent fiber glass inhalation study in rats with lung burdens<br />

found in workers involved in MMMF (primarily FG) manufacturing<br />

(McDonald et al. 1990). As shown in Table 7.7, rat fiber glass lung<br />

burdens vastly exceeded that <strong>of</strong> the workers reported by McDonald et al.,


110 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

which was not significantly elevated above reference levels. Fibers per mg<br />

dry lung for the rat after lifetime exposure was >4000-fold higher than for<br />

the fiber glass worker, average exposure 11 years (the average time from<br />

last employment in MMMF manufacturing and death was 12 years). Lung<br />

fiber dimensions in the rat study were comparable to those <strong>of</strong> fibers<br />

recovered from the lung tissue <strong>of</strong> fiber glass manufacturing workers. From<br />

these comparisons, it can be concluded that the exposure levels used in the<br />

recent rat inhalation studies unequivocally achieved the goal <strong>of</strong> the studies<br />

to exceed human exposures by several orders <strong>of</strong> magnitude.<br />

Summary and conclusions<br />

MMVFs are among the most studied commercial products due to their<br />

widespread use and the concern for potential health effects <strong>of</strong> respirable<br />

fibers. In recent animal inhalation studies RCF produced lung fibrosis,<br />

mesotheliomas, and significant increases in lung tumors. However, it is<br />

believed that any potential cancer risk from RCF exposure can be<br />

minimized, if not eliminated, because <strong>of</strong> the small number <strong>of</strong> workers<br />

exposed and the ability to use respiratory protection and engineering<br />

controls to limit worker exposure. Both human and animal inhalation<br />

studies have shown no association between fiber glass exposure and<br />

disease. Although high exposure levels <strong>of</strong> rock wool (several orders <strong>of</strong><br />

magnitude higher than most reported workplace exposures) produced<br />

minimal lung fibrosis in rats, no mesotheliomas and no significant increase<br />

in lung tumors were observed. Slag wool produced no fibrosis or increase<br />

in tumors in the animal studies. The cohort mortality studies <strong>of</strong> rock wool<br />

and slag wool workers have also provided no clear dose-response<br />

relationship with fiber exposure.<br />

Results from the combined animal inhalation studies showed that<br />

differences in lung fiber burdens and lung clearance rates could not explain<br />

the differences observed in the toxicologic effects <strong>of</strong> MMVFs. These<br />

findings clearly indicate that dose, dimension and durability (i.e. the<br />

persistence <strong>of</strong> fibers in the rat lung) are not the only determinants <strong>of</strong> fiber<br />

toxicity; chemical composition and the surface physicochemical properties<br />

<strong>of</strong> the fibers may also play an important role. Exposure levels from animal<br />

inhalation studies were at least three orders <strong>of</strong> magnitude higher than for<br />

average airborne levels reported for many occupational settings.


Table 7.7 Reported lung fiber levels from fiber glass workers and rat inhalation study<br />

T.W.HESTERBERG ET AL. 111<br />

a Lung fibers: for humans, NIOSH A rules; for rats, total fibers (all fibers length/diameter >3:1).<br />

b For humans, NIOSH A rules; for rats, WHO respirable fibers, comparable to A rules because there were no diameters >3 µm in<br />

rats.<br />

c Hesterberg et al., (1993b), Rat fiber exposure was 5 days week•1 , 6 h day•1 for lifetime (2 years).<br />

d McDonald et al., (1990). Negative controls had not worked with FG and were matched with each FG worker for age and<br />

locale.<br />

e Occupational exposures averaged 11 years, followed by average <strong>of</strong> 12 years without exposure prior to death.<br />

f 101 were FG workers; 11 were mineral wool workers.<br />

g Not reported.


112 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

References<br />

BALZER, J.L., COOPER, W.C. and FOWLER, D.P., 1971, Fibrous glass-lined air<br />

transmission systems: an assessment <strong>of</strong> their environmental effects, Am. Ind.<br />

Hyg. Assoc. J., 32, 521–18.<br />

BOORMAN, G.A. and EUSTIS, S.L., 1990, Lung, in Borman, G.A., Eustis, S.L.,<br />

Elwell, M.R., Montgomery, C.A. and MacKenzie, W.F. (Eds), Pathology <strong>of</strong> the<br />

Fischer Rat pp. 339–68, New York: Academic Press.<br />

CHAMBERLAIN, M., BROWN, R.C., DAVIES, R. and GRIFFITHS, D.M., 1979,<br />

In vitro prediction <strong>of</strong> the pathogenicity <strong>of</strong> mineral dusts, Br. J. Exp. Pathol.,<br />

60, 320–7.<br />

CHIAZZE, L., WATKINS, D.K. and FRYER, C., 1992, A case-control study <strong>of</strong><br />

malignant and non malignant respiratory disease among employees <strong>of</strong> a<br />

fiberglass manufacturing facility, Br. J. Ind. Med., 49, 326–31.<br />

CHOLAK, J. and SCHAFER, L., 1971, Erosion <strong>of</strong> fibers from installed fibrous<br />

glass ducts, Arch. Environm. Hlth, 22, 220–9.<br />

DAVIS, J.M.G., ADDISON, J., BOLTON, R.E., DONALDSON, K., JONES, A.D.<br />

and WRIGHT, A., 1984, The pathogenic effects <strong>of</strong> fibrous ceramic aluminum<br />

sili cate glass administered to rats by inhalation or peritoneal injection, in<br />

Biological Effects <strong>of</strong> Man-made Mineral Fibres (Proceedings <strong>of</strong> a WHO/IARC<br />

Conference). Vol. 2, pp. 303–22, Copenhagen: World Health Organization.<br />

DEMENT, J.M., NETTESHEIM, P. and BRODY, A.R. (Eds), 1990, Workshop on<br />

fiber toxicology research needs. Environ. Hlth Perspect., 88, 261–8.<br />

ENGHOLM, G., ENGLUND, A., FLETCHER, T. and HALLIN, N., 1987,<br />

Respiratory cancer incidence in Swedish construction workers exposed to manmade<br />

mineral fibres, Ann. Occup. Hyg., 31, 665–77.<br />

ENTERLINE, P.E., MARSH, G.M., HENDERSON, V. and CALLAHAN, C.,<br />

1987, Mortality update <strong>of</strong> a cohort <strong>of</strong> US man-made mineral fibre workers,<br />

Ann. Occup. Hyg., 31, 625–56.<br />

ESMEN, N., et al., 1980, Estimation <strong>of</strong> Employee Exposures to Total Suspended<br />

Particulate Matter and Airborne Fibers in Insulation Installation Operations,<br />

University <strong>of</strong> Pittsburgh, March 19.<br />

GROSS, P., DE TREVILLE, R.T.P., CRALLEY, L.J., GRANQUIST, W.T. and<br />

PUNDSACK, F.L., 1970, The pulmonary response to fibrous dusts <strong>of</strong> diverse<br />

compositions, Am. Ind. Hyg. Assoc. J., 31, 125–32.<br />

GUTHRIE, G.D.JR. and MOSSMAN, B.T., (Eds) 1993, Health effects <strong>of</strong> mineral<br />

dusts, Rev. Mineral. Vol. 28, Washington, DC: Mineralogical Society <strong>of</strong><br />

America.<br />

HART G.A., KATHMAN, L.M. and HESTERBERG, T.W. 1994, In vitro<br />

cytotoxicity <strong>of</strong> asbestos and man-made vitreous fibers: roles <strong>of</strong> fiber length,<br />

diameter and composition, Carcinogenesis, 15, 971–7.<br />

HESTERBERG, T.W. and BARRETT, J.C., 1984, Dependence <strong>of</strong> asbestos- and<br />

mineral dust-induced transformation <strong>of</strong> mammalian cells in culture on fiber<br />

dimension, Cancer Res., 44, 2170–80.<br />

HESTERBERG, T.W. and HART, G.A., 1994, A comparison <strong>of</strong> human exposures<br />

to fiber glass with those used in a recent rat chronic inhalation study, Regulat.<br />

Toxicol. Pharmacol., 20, 535–46.


T.W.HESTERBERG ET AL. 113<br />

HESTERBERG, T.W., Vu, V., CHASE, G.R., MCCONNELL, E.E., BUNN, W.B.<br />

and ANDERSON, R., 1991, Use <strong>of</strong> animal models to study man-made fiber<br />

carcinogenesis, in Brinkly, B., Lechmer, J. and Harris, C. (Eds) Cellular and<br />

Molecular Aspects <strong>of</strong> Fiber Carcinogenesis, pp. 183–205, Cold Spring Harbor<br />

New York: Cold Spring Harbor Laboratory.<br />

HESTERBERG, T.W., HART, G.A. and BUNN, W.B., 1993a, In vitro toxicology<br />

<strong>of</strong> fibers: mechanistic studies and possible use for screening assays, in Warheit,<br />

D. (Ed) Fiber <strong>Toxicology</strong> New York: Academic Press.<br />

HESTERBERG, T.W., MIILLER, W.C., MCCONNELL, E.E., CHEVALIER, J.,<br />

HADLEY, J., BERNSTEIN, D.M., THEVENAZ, P. and ANDERSON, R.,<br />

1993b, Chronic inhalation toxicity <strong>of</strong> size-separated glass fibers in Fischer 344<br />

rats, Fundam. Appl. Toxicol, 20, 464–76.<br />

HESTERBERG, T.W., MCCONNELL, E.E., MIILLER, W.C., BERNSTEIN,<br />

D.M., MAST, R. and ANDERSON, R., 1994, Relationship between lung<br />

biopersistence and biological effects <strong>of</strong> man-made vitreous fibres after chronic<br />

inhalation in rodents, in Bignon, J., Saracci, R. and Touray, J.-C. (Eds)<br />

Biopersistence <strong>of</strong> Respir-able Synthetic Fibres and Minerals (in press).<br />

IARC, 1988, Monographs on the Evaluation <strong>of</strong> Carcinogenic Risks to Humans, Vol.<br />

43, World Health Organization International Agency for Research on Cancer,<br />

Man-Made Mineral Fibres, Environmental Health Criteria 77, Geneva.<br />

IPCS, 1988, International Programme on Chemical Safety, Man-made Mineral<br />

Fibers, Environmental Health Criteria Document No. 77, Geneva: World<br />

Health Organization.<br />

JACOB, T.R., HADLEY, J.G., BENDER, J.R. and EASTES, W., 1992, Airborne<br />

glass fiber concentrations during installation <strong>of</strong> residential insulation, Am.<br />

Ind., Hyg. Assoc. J., 53, 519–23.<br />

JACOB, T.R., HADLEY, J.G., BENDER, J.R. and EASTES, W., 1993, Airborne<br />

glass fiber concentrations during manufacturing operations involving glass<br />

wool insulation, Am. Ind., Hyg. Assoc. J., 54, 320–6.<br />

KODAMA, Y., MANESS, S.C., IGLEHART, J.D., BOREIKO, C.J. and<br />

HESTERBERG, T.W., 1993, Cytotoxic and cytogenetic effects <strong>of</strong> asbestos to<br />

human bronchial epithelial cells in culture, Carcinogenesis, 14, 691–7.<br />

LEBOUFFANT, L., DANIEL, H., HENIN, J.P., MARTIN, J.C., NORMAND, C.,<br />

THICHOUX, G. and TROLARD, F., 1987, Experimental study on long-term<br />

effects <strong>of</strong> inhaled MMMF on the lung <strong>of</strong> rats, Ann. Occup. Hyg., 31, 765–90.<br />

LEE, K.P., BARRAS, C.E., GRIFFITH, F.D., WARITZ, R.S. & LAPIN, C.A.,<br />

1981, Comparative pulmonary responses to inhaled inorganic fibers with<br />

asbestos and fiberglass, Environm. Res., 24, 167–91.<br />

LEES, P., BREYSSE, P., MCARTHUR, B., MILLER, M., ROONEY, B.,<br />

ROBBINS, C. and CORN, M., 1993a, End user exposures to man-made<br />

vitreous fibers: I. Installation <strong>of</strong> residential insulation products. Appl. Occup.<br />

Environ. Hyg. 8(12), 1022–30.<br />

LEES, P., BREYSSE, P., MCARTHUR, B., MILLER, M., ROONEY, B.,<br />

ROBBINS, C. and CORN, M., 1993b, End user exposures to refractory<br />

ceramic fiber insulation products, unpublished report.<br />

MARSH, G.M., ENTERLINE, P.E., STONE, R.A. and HENDERSON, V.L., 1990,<br />

Mortality among a cohort <strong>of</strong> U.S. man-made mineral fiber workers: 1985<br />

followup, J. Occup. Med., 32, 594–604.


114 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

MAST, R.W., MCCONNELL, E.E., ANDERSON, R., CHEVALIER, J., KOTIN, P.,<br />

BERNSTEIN, D.M., THEVENAZ, P. GLASS, L.R. MILLER, W.C. and<br />

HESTERBERG, T.W., 1995, Studies on the chronic toxicity (inhalation) <strong>of</strong><br />

refractory ceramic fiber in male Fischer 344 rats, Inhalat. Toxicol., 7, 425–67.<br />

McCLELLAN, R.O., MILLER, F.J., HESTERBERG, T.W., WARHEIT, D.B.,<br />

BUNN, W.B. et al., 1992, Approaches to evaluating the toxicity and<br />

carcinogenicity <strong>of</strong> man-made fibers, Regulat. Toxicol. Pharmacol., 16, 321–<br />

64.<br />

MCCONNELL, E.E., WAGNER, J.C., SKIDMORE, J.W. and MOORE, J.A.,<br />

1984, A comparative study <strong>of</strong> the fibrogenic and carcinogenic effects <strong>of</strong> UICC<br />

Canadian chrysotile B asbestos and glass micr<strong>of</strong>ibre (JM 100), in Biological<br />

Effects <strong>of</strong> Man-made Mineral Fibres (Proceedings <strong>of</strong> a WHO/IARC<br />

Conference), Vol. 2. pp. 234–52, Copenhagen: World Health Organization.<br />

MCCONNELL, E.E., KAMSTRUP, O., MUSSELMAN, R., HESTERBERG, T.W.,<br />

CHEVALIER, J., MIILLER, W.C. and THEVENAZ, P., 1994, Chronic<br />

inhalation study <strong>of</strong> size-separated rock and slag wool insulation fibers in<br />

Fischer 344/N rats, Inhalat. Toxicol., 6, 571–614.<br />

MCDONALD, J.C., CASE, B.W., ENTERLINE, P.E., HENDERSON, V.,<br />

MCDONALD, A.D., PLOURDE, M. and SEBASTIEN, P., 1990, Lung dust<br />

analysis in the assessment <strong>of</strong> past exposure <strong>of</strong> man-made mineral fibre<br />

workers, Ann. Occup. Hyg., 34(5), 427–41.<br />

MITCHELL, R.I., DONOFRIO, D.J. and MOORMAN, W.J., 1986, Chronic<br />

inhala-tion toxicity <strong>of</strong> fibrous glass in rats and monkeys, J. Am. Coll. Tox., 5,<br />

545–75.<br />

MUHLE, H., POTT, F., BELLMANN, B., TAKENAKA, S. and ZIEM, U., 1987,<br />

Inhalation and injection experiments in rats to test the carcinogenicity <strong>of</strong><br />

MMMF, Am. Occup. Hyg., 31, 755–64.<br />

NIOSH Manual <strong>of</strong> Analytical Methods, 3rd. ed, US Dept <strong>of</strong> Health & Human<br />

Services, Public Health Service, Centers for Disease Control.<br />

OSHA (Occupational Safety and Health Administration), 1992, Proposed Rules for<br />

Fibrous Glass, Including Refractory Ceramic Fibers, Federal Register, Vol. 57,<br />

No. 114, p. 26195.<br />

OSHIMURA, M., HESTERBERG, T.W., TSUTSUI, T. and BARRETT, C.J., 1984,<br />

Correlation <strong>of</strong> asbestos-induced cytogenetic effects with cell transformation <strong>of</strong><br />

Syrian hamster embryo cells in culture, Cancer Res., 44, 5017–22.<br />

POOLE, A., BROWN, R.C. and ROOD, A.P., 1986, The in vitro activities <strong>of</strong> a<br />

highly carcinogenic mineral fibre—potassium octatitanate, Br. J. Exp. Pathol,<br />

67, 289–96.<br />

POTT, F., ZIEM, U., REIFFER, F.J., HUTH, F., ERNST, H. and MOHR, U., 1987,<br />

Carcinogenicity studies <strong>of</strong> fibres, metal compounds and some other dusts in<br />

rats., Exp. Pathol, 32, 129–52.<br />

RIRIE, D.G., HESTERBERG, T.W., BARRETT, J.C. and NETTESHEIM, P.,<br />

1985, Toxicity <strong>of</strong> asbestos and glass fibers for rat tracheal epithelial cells in<br />

culture, in Beck, E.G. and Bignon, J. (Eds), In vitro Effects <strong>of</strong> Mineral Dusts,<br />

NATO ASI Series, Vol. G3, pp. 177–184, Berlin (West): Springer.<br />

SARACCI, R., SIMONATO, L., ACHESON, E.D., ANDERSEN, A., BERTAZZI,<br />

P. A., CLAUDE, J., CHARNAY, N., ESTEVE, J., FRENTZEL-BEYME, R.R.,<br />

GARDNER, M.J., JENSEN, O.M., MAASING, R., OLSEN, J.H., TEPPO, L.,


T.W.HESTERBERG ET AL. 115<br />

WESTERHOLM, P. and ZOCCHETTI, C., 1984, Mortality and incidence <strong>of</strong><br />

cancer <strong>of</strong> workers in the man-made vitreous fibres producing industry: an<br />

international investigation at 13 European plants, Brit. J. Indust. Med. 41,<br />

425–36.<br />

Schuller International, Inc., 1985–1988, Unpublished data from industrial hygiene<br />

surveys.<br />

Schuller International, Inc., 1987, Unpublished data from industrial hygiene<br />

surveys.<br />

SHANNON, H.S., HAYES, M., JULIAN, J.A. and MUIR, D.C.F., 1987, Mortality<br />

experience <strong>of</strong> Ontario glass fibre workers—extended follow-up, Ann. Occup.<br />

Hyg., 31, 657–62.<br />

SIMONATO, L., FLETCHER, A.C., CHERRIE, J., ANDERSEN, A., VERTAZI,<br />

P., CHARNAY, N., CLAUDE, J., DODGSON, J., ESTEVE, J., FRENTZEL-<br />

BEYME, R., GARDNER, M.J., JENSEN, O., OLSEN, J., TEPPO, L.,<br />

WINKELMANN, R., WESTERHOLM, P., WINTER, P.D., ZOCCHETTI, C.<br />

and SARACCI, R., 1987, The International Agency for Research on Cancer<br />

historical cohort study <strong>of</strong> MMMF production workers in seven European<br />

countries, extension <strong>of</strong> the followup, Ann. Occup. Hyg., 31, 603–23.<br />

SINCOCK, A. and SEABRIGHT, M., 1975, Induction <strong>of</strong> chromosome changes in<br />

Chinese hamster cells by exposure to asbestos fibres, Nature, 257, 56–8.<br />

SMITH, D.M., ORTIZ, L.W., ARCHULETA, R.F. and JOHNSON, N.F., 1987,<br />

Long-term health effects in hamsters and rats exposed chronically to manmade<br />

vitreous fibers, Ann. Occup. Hyg., 31, 731–54.<br />

SOLLEVELD, H.A., HASEMAN, J.K. and McCoNNELL, E.E., 1984, Natural<br />

history <strong>of</strong> body weight gain, survival and neoplasia in the F344 rat, J. Nat.<br />

Cancer Inst., 72(4), 929–40.<br />

STANTON, J.F., LAYARD, M., TEGERIS, A., MILLER, E., MAY, M., MORGAN,<br />

E. and SMITH, A. 1981, Relation <strong>of</strong> particle dimension to carcinogenicity in<br />

amphibole and other fibrous minerals, J. Nat. Cancer Inst., 67, 965–75.<br />

TIESLER, H. and DRAEGER, U., 1994, Measurement and identification <strong>of</strong><br />

insulation product-related fibres in constrast to ubiquitous fibres, in<br />

Proceedings from the Symposium on Indoor Air, Helsinki, January, 1993, in<br />

press.<br />

TILKES, F. and BECK, E.G., 1980, Comparison <strong>of</strong> length-dependent cytotoxicity<br />

<strong>of</strong> inhalable asbestos and man-made mineral fibres, in Wagner, J.C., (Ed.)<br />

Biological Effects <strong>of</strong> Mineral Fibres (IARC Scientific Publications No. 30), pp.<br />

475–83, Lyon: International Agency for Research on Cancer.<br />

TIMA (Thermal Insulation Manufacturer’s Association), 1990, Health and Safety<br />

Aspects <strong>of</strong> Man-made Vitreous Fibres: Information, Data, Comments and<br />

Recommendations Regarding Occupational Exposure to Man-made Vitreous<br />

Fibers, Part 3 Glass Fibers, Sections III, IV, submitted 7/10/90 to the National<br />

Institute for Occupational Safety and Health in response to 55 Fed. Reg. 5073.<br />

Vu, V. (US Environmental Protection Agency, EPA), 1988, Health Hazard<br />

Assessment <strong>of</strong> Nonasbestos Fibers, Final Draft, Health and Environmental<br />

Review Division, Office <strong>of</strong> Toxic Substances.<br />

WAGNER, J.C., BERRY, G.B., HILL, R.J., MUNDAY, D.E. and SKIDMORE, J.<br />

W., 1984, Animal experiments with MMM(V)F—effects <strong>of</strong> inhalation and<br />

intrapleural inoculation in rats, in Biological Effects <strong>of</strong> Man-made Mineral


116 CARCINOGENIC POTENTIAL OF MAN-MADE VITREOUS FIBERS<br />

Fibres (Proceedings <strong>of</strong> a WHO/IARC Conference), Vol. 2, pp. 209–33,<br />

Copenhagen: World Health Organization.<br />

WHO, 1984, Biological effects <strong>of</strong> man-made mineral fibers: Proceedings <strong>of</strong> a WHO/<br />

IARC conference, Vol. 2, Copenhagen: WHO Regional <strong>of</strong>fice.<br />

WHO, 1985, Reference methods for measuring man-made mineral fibers<br />

(MMMF). Prepared by WHO/EURO Technical Committee for evaluating<br />

MMMF, Copenhagen: WHO Regional Office.<br />

WHO, 1992, Validity <strong>of</strong> methods for assessment <strong>of</strong> carcinogenicity <strong>of</strong> fibres,<br />

Copenhagen: WHO Regional Office.<br />

WONG, O., FOLIART, D. and TRENT, L.S., 1991, A case-control study <strong>of</strong> lung<br />

cancer in a cohort <strong>of</strong> workers potentially exposed to slag wool fibres, Brit. J.<br />

Indust. Med., 418, 818–24.


8<br />

Pulmonary Toxicity Studies with Man-made<br />

Organic Fibres: Preparation and Comparisons <strong>of</strong><br />

Size-separated Para-aramid with Chrysotile<br />

Asbestos Fibres<br />

DAVID B.WARHEIT, 1 MARK A.HARTSKY, 1 CHARLES<br />

J.BUTTERICK 2 and STEVEN R.FRAME 1<br />

1 DuPont Haskell Laboratory, Newark, DE, 2 Texas Tech<br />

Health Sciences Center Lubbock, TX<br />

Introduction<br />

This study was designed to compare the pulmonary toxic effects <strong>of</strong><br />

inhaled, size-separated preparations <strong>of</strong> chrysotile asbestos fibres with paraaramid<br />

fibrils at similar aerosol fibre concentrations. Chrysotile samples<br />

are known to have an abundance <strong>of</strong> short fibres, with mean lengths<br />

generally in the range <strong>of</strong> 2 µm. This is important to note because one <strong>of</strong> the<br />

critical factors influencing the pathogenesis <strong>of</strong> fibre-related lung disease is<br />

fibre dimension (Davis et al., 1986). As a consequence, attempts were made<br />

to selectively enhance the mean lengths <strong>of</strong> chrysotile asbestos fibres used in<br />

this inhalation toxicity study, in order to make reasonable comparisons<br />

between the two fibre-types.<br />

Methods<br />

General experimental design<br />

Groups <strong>of</strong> male Crl: CDBR rats (7–8 weeks old, Charles River Breeding<br />

Laboratories, Kingston, New York) were used to assess the pulmonary<br />

effects <strong>of</strong> 2-week inhalation exposures to size-separated preparations <strong>of</strong><br />

Kevlar ® para-aramid fibrils or chrysotile asbestos fibres. Animals were<br />

exposed 6 hr day −1 , 5 days week −1 for 2 weeks. For this study, Kevlar ® was<br />

utilized as a representative para-aramid fibril. The two commercial types <strong>of</strong><br />

para-aramid fibres are Twaron ® , made by Akzo, and Kevlar ® , made by<br />

DuPont. Following exposure, the lungs <strong>of</strong> p-aramid or chrysotile-exposed<br />

animals and age-matched sham controls were subsequently evaluated by<br />

bronchoalveolar lavage fluid analysis at 0 h, 5 days, 1 and 3 months<br />

postexposure. The lungs <strong>of</strong> additional animals were evaluated for<br />

biodurability, pulmonary clearance, pulmonary histopathologic lesions and


118 PULMONARY TOXICITY STUDIES WITH MAN-MADE ORGANIC FIBRES<br />

lung and mesothelial cell proliferation at 0 hrs, 5 days 1, 3, 6 and 12<br />

months postexposure.<br />

Fibre preparation and inhalation exposure<br />

Ultrafine Kevlar ® p-aramid fibrils were supplied by DuPont Fibres. A<br />

special preparation <strong>of</strong> respirable p-aramid fibrils which had been prepared<br />

for the 2-year inhalation study (Lee et al., 1988) was utilized for this study.<br />

Bulk Canadian chrysotile asbestos fibres were obtained from Mr John<br />

Addison <strong>of</strong> the Institute <strong>of</strong> Occupational Medicine in Edinburgh, Scotland.<br />

Attempts were made to size-separate the bulk fibre preparation (i.e.<br />

selectively enhance the percentages <strong>of</strong> long fibres while removing the short<br />

fibres) by placing the fibres in a rotating sieve shaker and sieving through a<br />

series <strong>of</strong> metal mesh screens. The fraction containing the longer fibres (and<br />

a number <strong>of</strong> short fibres) was collected and generated for inhalation<br />

studies; fibres were collected on a filter and dimensional analysis (i.e. length<br />

and diameter assessments) was performed using scanning electron<br />

microscopy. The results showed that this technique was partially successful<br />

as the median and mean lengths <strong>of</strong> fibres were increased from 3 and 5 µm,<br />

respectively, in the original bulk sample to 6 and 9 µm in the generated<br />

sample preparation. The median lengths and diameters <strong>of</strong> p-aramid fibrils<br />

used in the study were 9 µm and 0.3 µm, respectively.<br />

The methods for aerosol generation <strong>of</strong> p-aramid fibrils have previously<br />

been reported (Warheit et al., 1992). Final mean fibre concentrations for<br />

the p-aramid exposures were 772 and 419 f cm −3 .<br />

Aerosols <strong>of</strong> chrysotile asbestos fibres were generated in a similar<br />

manner, i.e. with a binfeeder and baffles, but without the microjet<br />

apparatus. Final mean fibre concentrations for the chrysotile asbestos<br />

exposures were 782 and 458 f cm −3 . Fibre lung burdens were quantified<br />

from digested lung tissue <strong>of</strong> animals sacrificed immediately after the end <strong>of</strong><br />

the 2-week exposure.<br />

Pulmonary lavage and biochemical assays on lavaged<br />

fluids<br />

Bronchoalveolar lavage procedures, cell quantification, and biochemical<br />

assays were conducted according to methods previously described (Warheit<br />

et al., 1984a, 1992). In addition, the methods for measuring lactate<br />

dehydrogenase (LDH), N-acetyl-β-glucosaminidase (NAG), and alkaline<br />

phosphatase (ALP) and protein in BAL fluids have been reported (Warheit<br />

et al., 1992).


Lung dissection, tissue preparation and pulmonary cell<br />

proliferation<br />

The lungs <strong>of</strong> rats exposed to p-aramid and chrysotile asbestos fibres for 2<br />

weeks were prepared for light microscopy by airway infusion using<br />

methods previously reported (Warheit et al., 1984b, 1991).<br />

Pulmonary cell proliferation experiments were designed to measure the<br />

effects <strong>of</strong> fibre inhalation exposure on terminal bronchiolar, proximal lung<br />

parenchymal (i.e. alveolar duct bifurcations and adjacent areas), subpleural<br />

and visceral pleural, and mesothelial cell turnover in rats following 2-week<br />

exposures. Groups <strong>of</strong> sham and fibre-exposed rats were given a 2-h pulse<br />

immediately after exposures, as well as 5 days, 1, 3, 6 and 12 months (still<br />

in progress) postexposure with an intraperitoneal injection <strong>of</strong> 5-bromo-2′deoxy-uridine<br />

(BrdU) dissolved in a 0.5N sodium bicarbonate buffer<br />

solution at a dose <strong>of</strong> 100 mg kg −1 body weight as previously described<br />

(Warheit et al., 1992). In addition, sections <strong>of</strong> duodenum served as a<br />

positive control. For each treatment group, there were immunostained<br />

nuclei in airways (i.e. terminal bronchiolar epithelial cells), lung parenchyma<br />

(i.e. epithelial, interstitial cells or macrophages), subpleura and visceral<br />

pleura, and mesothelial cells. All regions were counted by light microscopy<br />

at ×1000 magnification. Statistics were carried out using a two-tailed<br />

Students t test on a Micros<strong>of</strong>t Excel s<strong>of</strong>tware program.<br />

Fibre recovery from lung tissue<br />

Para-aramid fibrils were recovered from the lungs <strong>of</strong> exposed rats using a<br />

diluted 1.3% hypochlorite (Clorox bleach) solution. The results <strong>of</strong><br />

validation studies in our laboratory demonstrated that the dilute Clorox<br />

solution (10 min digestion) was more effective in digesting lung tissue than<br />

the KOH method that we had previously reported (Warheit et al., 1992).<br />

Chrysotile asbestos fibres were recovered from the lungs <strong>of</strong> exposed rats<br />

by incubating the lung tissue with a 5.25% hypochlorite solution for 3 h.<br />

Subsequently, the filters containing fibres recovered from lung tissue were<br />

mounted and prepared for phase-contrast light microscopy (for counting)<br />

and for scanning electron microscopy (for fibre dimensional analysis),<br />

according to methods previously described (Warheit et al., 1992).<br />

Results<br />

D.B.WARHEIT ET AL. 119<br />

Size-separation methods for chrysotile asbestos fibres<br />

The results from size-separation attempts showed that there was a shift in<br />

the distribution <strong>of</strong> fibre lengths from shorter fibres to longer fibres<br />

(Figures 8.1(A)– (C)). Count median lengths <strong>of</strong> chrysotile asbestos fibres


120 PULMONARY TOXICITY STUDIES WITH MAN-MADE ORGANIC FIBRES<br />

were increased from 3 µm in the original generated sample to 6 µm in the<br />

size-separated sample. In comparison to the chrysotile asbestos sample,<br />

there was a significantly greater proportion <strong>of</strong> long p-aramid fibrils which<br />

were used in the inhalation study with median lengths >9 µm.<br />

Lung burden analysis<br />

Although the aerosol fibre concentrations were similar throughout the<br />

study (p-aramid high conc.=772 f cm −3 , chrysotile high conc.=782 f cm −3 ;<br />

p-aramid low conc.=419 f cm −3 , chrysotile low conc.=458 f cm −3 ),<br />

measurement <strong>of</strong> lung fibre burdens from digested lung tissue at time 0 (i.e.<br />

immediately after exposure) demonstrated a substantial difference in lung<br />

burden between the two fibre-types as measured by phase contrast optical<br />

microscopy (PCOM). The mean lung fibre (>5 µm) burden from 3 rats/<br />

dose group exposed to chrysotile asbestos was 3.7×10 7 (±7.4×10 6 ) fibres/<br />

lung for the high dose group and 1.3×10 7 (±4×10 6 ) fibres/lung for the low<br />

dose group. In contrast, the mean lung fibre burden from 3 rats/dose group<br />

exposed to para-aramid fibres was 7.6×10 7 (±1.9×10 7 ) fibres per lung for<br />

the high dose group and 4.8×10 7 ( ±2.1×10 7 ) fibres/lung for the low dose<br />

group. In addition, the count median length <strong>of</strong> chrysotile fibres recovered<br />

from the lungs <strong>of</strong> exposed animals immediately after 2-week exposure was<br />

3.5 µm, while the count median diameter was 0.15 µm. In contrast, the<br />

count median length <strong>of</strong> para-aramid fibres recovered from the lungs <strong>of</strong><br />

exposed animals immediately after 2-week exposure was 8.6 µm, while the<br />

count median diameter was 0.3 µm (Figure 8.2(A) and (B); numerical data<br />

not shown). These data indicate that our attempts to size-separate<br />

Canadian chrysotile fibres were only partially successful. The lung burden<br />

data also suggest that comparisons <strong>of</strong> the effects <strong>of</strong> chrysotile vs paraaramid<br />

at high and low doses are difficult to make since the doses were not<br />

equivalent.<br />

Bronchoalveolar lavage data<br />

Two-week exposures to p-aramid fibrils or chrysotile asbestos fibres<br />

produced transient pulmonary inflammatory responses as measured by<br />

bronchoalveolar lavage fluid analysis (see Table 8.1).<br />

Light microscopic histopathology<br />

Exposures to p-aramid and chrysotile were associated with minimal to mild<br />

centriacinar inflammation and fibrosis (increased trichrome staining)<br />

immediately after and 5 days after 2-week exposures. Lesions were slightly<br />

more prominent in p-aramid-exposed rats due to increased inflammation.<br />

Lesions were less severe at 1 month and essentially resolved at 6 months


D.B.WARHEIT ET AL. 121<br />

Figure 8.1 (A) Chrysotile asbestos lengths—original generated sample for 4 different<br />

experiments. The graph depicts the fibre length distributions as assessed by<br />

scanning electron microscopy from four aerosol exposures prior to attempts to size<br />

separate the fibres. Fifty percent <strong>of</strong> the fibres from all four groups are less than 3–4<br />

µm. (B) Distributions <strong>of</strong> size-separated chrysotile asbestos lengths used in the<br />

inhalation study from the high-dose


122 PULMONARY TOXICITY STUDIES WITH MAN-MADE ORGANIC FIBRES<br />

Figure 8.1 Continued<br />

exposures and (C) the low-dose exposure groups. A casual glance at the two graphs<br />

B and C indicates that some success was attained in increasing the mean lengths in<br />

the aerosol <strong>of</strong> the generated chrysotile asbestos sample.<br />

with only occasional centriacinar regions having slight, fibril-associated<br />

thickening <strong>of</strong> alveolar duct bifurcations. At 1 year postexposure, the lungs<br />

in p-aramid exposed rats were similar to controls. The 1-year chrysotileexposed<br />

animals are still in recovery.<br />

Pulmonary cell proliferation<br />

In chrysotile asbestos-exposed rats, substantial increases compared to<br />

controls in pulmonary cell proliferation indices were measured on terminal<br />

bronchiolar, parenchymal, visceral pleural/subpleural and mesothelial<br />

surfaces, and many <strong>of</strong> these effects were sustained through 3 months<br />

postexposure. These data demonstrate that 2-week chrysotile exposures<br />

produced a prolonged proliferative response in airway, alveolar and<br />

subpleural cells, as evidenced by the sustained effect through 3 months<br />

postexposure (Table 8.2).<br />

Pulmonary cell proliferation studies demonstrated that 2-week exposures<br />

to the high dose <strong>of</strong> p-aramid fibrils produced a transient increase in<br />

terminal bronchiolar and visceral pleural/subpleural cell labeling responses.<br />

No increases in lung parenchymal, or subpleural cell labeling indices were<br />

mea sured at any time period relative to sham controls. In addition, no


D.B.WARHEIT ET AL. 123<br />

Figure 8.2 (A) Scanning electron microscopy (SEM) micrograph <strong>of</strong> an aerosol filter<br />

containing a mixture <strong>of</strong> long and short chrysotile asbestos fibres (arrows). (B) An<br />

SEM micrograph <strong>of</strong> fibres recovered from the lung <strong>of</strong> a rat 3 months after 2-week<br />

chrysotile exposures. Note that most <strong>of</strong> the fibres are long (arrows), indicating that<br />

the long chrysotile asbestos fibres were retained in the lung while the shorter fibres<br />

were cleared from the respiratory tract.<br />

increases in cell labeling indices were measured in animals exposed to a<br />

lower dose <strong>of</strong> p-aramid fibrils at any postexposure time period (Table 8.2).


124 PULMONARY TOXICITY STUDIES WITH MAN-MADE ORGANIC FIBRES<br />

Table 8.1 Pulmonary inflammation and fibre biodurability in the lungs <strong>of</strong><br />

chrysotile asbestos and p-aramid-exposed rats<br />

0 h=immediately after exposure; 5 D=5 days; 1 M=1 month; 3 M=3 months; 6<br />

M=6 months;<br />

ND=not determined.<br />

Lung digestion/biodurability studies<br />

Preliminary dimensional analysis studies demonstrated that median lengths<br />

<strong>of</strong> fibres recovered from digested asbestos-exposed lung tissue were<br />

increased over time suggesting that short asbestos fibres were selectively<br />

cleared from the lungs, with apparent insignificant or pulmonary clearance<br />

and greater durability/retention <strong>of</strong> long fibres (Table 8.1).<br />

Preliminary studies with p-aramid fibrils recovered from the lungs <strong>of</strong><br />

exposed rats are consistent with earlier data suggesting biodegradability <strong>of</strong><br />

inhaled p-aramid fibrils (Warheit et al., 1992; Kelly et al., 1993)<br />

(Table 8.1). These data also are in agreement with the results <strong>of</strong> a current<br />

interim report authored by the Institute <strong>of</strong> Occupational Medicine in<br />

Edinburgh, Scotland. In addition, as previously reported (Warheit et al.,<br />

1992), a transient increase in fibre numbers at early postexposure time<br />

periods was measured following cessation <strong>of</strong> exposure. These results<br />

indicate that the increase in p-aramid fibres is due to fibre shortening and as<br />

a consequence, increased numbers <strong>of</strong> shorter fibres. This is accounted for<br />

by a substantial reduction in the median lengths <strong>of</strong> recovered fibres<br />

concomitant with only a slight decrease in fibre diameter.


Table 8.2 Cell proliferation effects in chrysotile asbestos and p-Aramid-exposed<br />

rats<br />

a p


126 PULMONARY TOXICITY STUDIES WITH MAN-MADE ORGANIC FIBRES<br />

The BrdU pulmonary cell labeling results demonstrating sustained<br />

proliferative effects in chrysotile-exposed rats presented here are consistent<br />

with findings from several other investigators (Brody and Overby, 1989;<br />

McGavran et al., 1990; Coin et al., 1992a). In studies by Brody and<br />

Overby (1989), acute inhalation exposures to chrysotile asbestos fibres<br />

produced a biphasic cell labeling response in the lungs <strong>of</strong> exposed rats and<br />

mice. This was characterized by dramatic increases in epithelial cell DNA<br />

synthesis, followed several days later by enhanced labeling <strong>of</strong> interstitial<br />

cells. In follow-up studies, a 3 day exposure prolonged the duration <strong>of</strong><br />

increased cell labeling (Coin et al., 1992b). In another study, Coin et al.,<br />

(1991) reported that a 5-h exposure to chrysotile fibres in mice produced<br />

substantial increases in mesothelial and subpleural cell labeling indices at 2<br />

and 8 days postexposure.<br />

The finding <strong>of</strong> sustained subpleural and mesothelial cell proliferation in<br />

chrysotile-exposed rats was unexpected and raises the issue regarding the<br />

association <strong>of</strong> chrysotile with the development <strong>of</strong> mesothelioma. In this<br />

regard, inhalation <strong>of</strong> chrysotile asbestos fibres produced mesotheliomas in<br />

exposed rats (Wagner et al., 1974; Davis and Jones, 1988).<br />

The biodurability data reported here demonstrating retention or reduced<br />

clearance <strong>of</strong> long chrysotile fibres are consistent with the results <strong>of</strong><br />

previous studies by Roggli and Brody (1984) and Bellmann et al., (1986,<br />

1987). In contrast to the enhanced biodurability <strong>of</strong> chrysotile asbestos<br />

fibres, the results with p-aramid fibres suggest that the fibrils undergo<br />

biodegradability in the lungs <strong>of</strong> exposed rats. These findings confirm our<br />

earlier studies (Warheit et al., 1992) and are in concordance with the<br />

results <strong>of</strong> Kelly et al. (1993) and the recent findings <strong>of</strong> the IOM.<br />

In conclusion, size separation techniques for chrysotile asbestos fibres<br />

were partially successful in increasing median lengths from 3 µm to 6 µm.<br />

Histopathological studies demonstrated that both p-aramid and chrysotile<br />

produced a minimal to mild inflammatory response which produced<br />

thickening <strong>of</strong> the alveolar duct bifurcations. These effects peaked at 1<br />

month postexposure and were essentially reversible by 6 months<br />

postexposure.<br />

Pulmonary cell labeling studies demonstrated substantial increases in<br />

lung parenchymal, airway, pleural/subpleural, and mesothelial cell<br />

proliferation effects following chrysotile exposures, suggesting that<br />

chrysotile produces a potent proliferative response in the airways, lung<br />

parenchyma, and subpleural/ pleural regions. In contrast, p-aramid<br />

exposures produced only transient effects in airway and subpleural regions.<br />

Fibre biopersistence/durability results thus far indicate that the long<br />

chrysotile fibres are retained in the lung or cleared at a slow rate. In<br />

contrast, p-aramid fibres have low biodurability in the lungs <strong>of</strong> exposed<br />

animals. In this regard, median lengths <strong>of</strong> chrysotile fibres recovered from


exposed lung tissue were increased over time, while median lengths <strong>of</strong> paramid<br />

fibrils were decreased over time.<br />

It is concluded that the proliferative effects and enhanced biodurability<br />

<strong>of</strong> chrysotile that are associated with the induction <strong>of</strong> chronic disease do<br />

not occur with p-aramid fibrils. Therefore, inhalation <strong>of</strong> chrysotile asbestos<br />

fibres is likely to produce greater long-term pulmonary toxic effects in<br />

comparison to para-aramid fibrils.<br />

Acknowledgments<br />

This study was sponsored by the DuPont Co. and Akzo Nobel Corp.<br />

References<br />

D.B.WARHEIT ET AL. 127<br />

BELLMANN, B., KONIG, H., MUHLE, H. and POTT, F., 1986, Chemical<br />

durability <strong>of</strong> asbestos and <strong>of</strong> man-made mineral fibres in vivo, J. Aerosol Sci.,<br />

17, 341–5.<br />

BELLMANN, B., MUHLE, H., POTT, F., KONIG, H., KLOPPEL, H. and<br />

SPURNY, K., 1987, Persistence <strong>of</strong> man-made mineral fibers (MMMF) and<br />

asbestos in rat lungs, Ann. Occup. Hyg., 31(4B), 693–709.<br />

BRODY, A.R. and OVERBY, L.H., 1989, Incorporation <strong>of</strong> tritiated thymidine by<br />

epithelial and interstitial cells in bronchiolar-alveolar regions <strong>of</strong> asbestosexposed<br />

rats, Am. J. Pathol., 134, 133–40.<br />

COIN, P.G., MOORE, L.B., ROGGLI, V. and BRODY, A.R., 1991, Pleural<br />

incorporation <strong>of</strong> 3 H-TdR after inhalation <strong>of</strong> chrysotile asbestos in the mouse,<br />

Am. Rev. Respir. Dis., 143, A604.<br />

COIN, P.G., ROGGLI, V.L. and BRODY, A.R., 1992a, Deposition, clearance and<br />

translocation <strong>of</strong> chrysotile asbestos from peripheral and central regions <strong>of</strong> the<br />

rat lung, Environ, Res., 58, 97–116.<br />

COIN, P.G., ROGGLI, V. and BRODY, A.R., 1992b, Pulmonary fibrogenesis and<br />

BRDU incorporation after three consecutive inhalation exposures to chrysotile<br />

asbestos, Am. Rev. Respir. Dis., 145, A328.<br />

DAVIS, J.M.G. and JONES, A.D., 1988, Comparisons <strong>of</strong> the pathogenicity <strong>of</strong> long<br />

and short fibres <strong>of</strong> chrysotile asbestos in rats, Br. J. Exp. Pathol., 69, 717–37.<br />

DAVIS, J.M.G., ADDISON, J., BOLTON, R.E., DONALDSON, K. et al., 1986,<br />

The pathogenicity <strong>of</strong> long versus short fibre samples <strong>of</strong> amosite asbestos<br />

administered to rats by inhalation or intraperitoneal injection, Br. J. Exp.<br />

Pathol, 67, 415–30.<br />

KELLY, D.P., MERRIMAN, E.A., KENNEDY, G.L.JR. and LEE, K.P., 1993,<br />

Deposition, clearance, and shortening <strong>of</strong> Kevlar para-aramid fibrils in acute,<br />

subchronic, and chronic inhalation studies in rats, Fundam. Appl. Toxicol, 21,<br />

345–54.<br />

LEE, K.P., KELLY, D.P., O’NEAL, F.O., STADLER, J.C. and KENNEDY, G.<br />

L.JR, 1988, Lung response to ultrafine Kevlar aramid synthetic fibrils<br />

following 2-year inhalation exposure in rats, Fundam. Appl. Toxicol., 11, 1–<br />

20.


128 PULMONARY TOXICITY STUDIES WITH MAN-MADE ORGANIC FIBRES<br />

McGAVRAN, P.D., BUTTERICK, C.J. and BRODY, A.R., 1990, Tritiated<br />

thymidine incorporation and the development <strong>of</strong> an interstitial lesion in the<br />

bronchiolar alveolar regions <strong>of</strong> the lungs <strong>of</strong> normal and complement deficient<br />

mice after inhalation <strong>of</strong> chrysotile asbestos, J. Environ. Pathol. Toxicol.<br />

Oncol, 9, 377–92.<br />

ROGGLI, V.L. and BRODY, A.R., 1984, Changes in numbers and dimensions <strong>of</strong><br />

chrysotile asbestos fibers in lungs <strong>of</strong> rats following short-term exposure, Exp.<br />

Lung Res., 7, 133–47.<br />

WAGNER, J.C., BERRY, G., SKIDMORE, J.W. and TIMBRELL, V., 1974, The<br />

effects <strong>of</strong> the inhalation <strong>of</strong> asbestos in rats, Br. J. Cancer, 29, 252–70.<br />

WARHEIT, D.B., HILL, L.H. and BRODY, A.R., 1984a, Surface morphology and<br />

correlated phagocytic capacity <strong>of</strong> pulmonary macrophages lavaged from the<br />

lungs <strong>of</strong> rats, Exp. Lung Res., 6, 71–82.<br />

WARHEIT, D.B., CHANG, L.Y., HILL, L.H., HOOK, G.E.R., CRAPO, J.D. and<br />

BRODY, A.R., 1984b, Pulmonary macrophage accumulation and<br />

asbestosinduced lesions at sites <strong>of</strong> fiber deposition, Am. Rev. Respir. Dis., 129,<br />

301.<br />

WARHEIT, D.B., CARAKOSTAS, M.C., HARTSKY, M.A. and HANSEN, J.F.,<br />

1991, Development <strong>of</strong> a short-term inhalation bioassay to assess pulmonary<br />

toxicity <strong>of</strong> inhaled particles: Comparisons <strong>of</strong> pulmonary responses to carbonyl<br />

iron and silica, Toxicol Appl. Pharmacol., 107, 350–68.<br />

WARHEIT, D.B., KELLAR, K.A. and HARTSKY, M.A., 1992, Pulmonary cellular<br />

effects in rats following aerosol exposures to ultrafine Kevlar® aramid fibrils:<br />

evidence for biodegradability <strong>of</strong> inhaled fibrils, Toxicol. Appl. Pharmacol,<br />

116, 225– 39.


9<br />

Pulmonary Hyperreactivity to <strong>Industrial</strong><br />

Pollutants<br />

JÜRGEN PAULUHN<br />

Bayer AG, Wuppertal<br />

Introduction<br />

Environmental agents, such as ozone, nitrogen dioxide, formaldehyde, and<br />

sulfur dioxide; occupational pollutants, including natural dusts (grain, red<br />

cedar, animal dander), irritant fumes or vapors, and organic acid<br />

anhydrides, reactive dyes, or (di)isocyanates can cause increases in airway<br />

reactivity. Airway hyperreactivity is defined as an exaggerated acute<br />

obstructive response <strong>of</strong> the airways to one or more nonspecific stimuli. The<br />

incriminated etiologic low-molecular-weight agents all share a common<br />

toxicological characteristic <strong>of</strong> being irritant in nature. In some cases, the<br />

agents are present as a gas, in others the inciting agent is an aerosol. As yet<br />

it is not clear, for instance, whether induced airway hyperreactivity is a<br />

dose-effect phenomenon and whether a brief high level exposure causes<br />

more prolonged or intense airways response. While the illness clinically<br />

simulates bronchial asthma and is associated with airway hyperreactivity,<br />

it is considered to be different from typical occupational asthma because <strong>of</strong><br />

its rapid onset, specific relationship to a single environmental exposure,<br />

and no apparent preexisting period <strong>of</strong> sensitization to occur with the<br />

apparent lack <strong>of</strong> an allergic or immunologic etiology. Hence, this illness is<br />

termed reactive airways dysfunction syndrome, or RADS, because the<br />

characteristic finding is hyperreactivity <strong>of</strong> the airways (Brooks et al.,<br />

1985). Mechanisms to explain the development <strong>of</strong> RADS focus on the<br />

toxic effects <strong>of</strong> the irritant exposure on the airways. How this increased<br />

bronchial responsiveness is precisely triggered, amplified, sustained and<br />

how it relates to inflammatory events remains, to a certain extent,<br />

incompletely elucidated (Kay, 1991).<br />

A common pathologie accompaniment or cause <strong>of</strong> increased airway<br />

hyper-responsiveness is pulmonary inflammation. It is suggested that this<br />

inflammation is responsible for the change in histamine or cholinergic<br />

agonist responsiveness. Because subepithelial irritant receptors are<br />

superficial in location, they could be affected by an extensive bronchial<br />

inflammatory response which might occur after heavy irritant exposure.


130 PULMONARY HYPERREACTIVITY TO INDUSTRIAL POLLUTANTS<br />

Subsequent re-epithelialization and probable reinervation <strong>of</strong> bronchial<br />

mucosa might drastically alter the threshold <strong>of</strong> the receptors and cause<br />

airways hyperreactivity. It has been hypothesized that damage to airway<br />

epithelium by irritant chemicals could decrease the threshold <strong>of</strong> sensory<br />

endings within the mucosa, resulting in increased afferent and efferent vagal<br />

activity. Airway mucosal inflammation, activation <strong>of</strong> airway afferent<br />

nerves, and the release <strong>of</strong> low-molecular-weight neuropeptides as<br />

mediators <strong>of</strong> inflammation are known to affect the tonus <strong>of</strong> the airway<br />

smooth muscle and may play a crucial role in the acute increase in airway<br />

hyperresponsiveness occurring after exposure to irritant or inflammatory<br />

stimuli. Additionally, inflammatory mediators may further attract and<br />

activate inflammatory cells, which themselves release a whole array <strong>of</strong><br />

chemotactic and cytotoxic mediators that serve to perpetuate and amplify<br />

the inflammatory response. This complex interaction <strong>of</strong> different factors<br />

may result in epithelial desquamation, mucus gland hyperplasia, smooth<br />

muscle hypertrophy, and eventually render the airways hyperreactive to<br />

specific as well as nonspecific stimuli.<br />

Increased bronchial irritability, or hyperresponsiveness, to a wide variety<br />

<strong>of</strong> chemical agents and physical stimuli is also a major characteristic<br />

feature <strong>of</strong> bronchial asthma and the reactive airways dysfunction syndrome<br />

might clinically be indistinguishable from the asthma syndrome. Also for<br />

the latter, particular attention has been placed on the role <strong>of</strong> inflammation<br />

mediated influx <strong>of</strong> cells, mediator release and the interaction <strong>of</strong> irritant<br />

induced neurogenic and inflammatory factors. Neural control <strong>of</strong> airway<br />

caliber is far from being simple and it is likely to contribute to airway<br />

narrowing and bronchial hyper-responsiveness. Myelinated and<br />

nonmyelinated nerve fibers (C fibers) are involved in the sensory irritation<br />

response and their stimulation may result in release <strong>of</strong> specific<br />

neuropeptides, known to be potent releasers <strong>of</strong> mediators from airway<br />

mast cells (Barnes et al., 1991a, b; Nielsen, 1991). Specific neuropeptides<br />

are also known to attract eosinophils which can be stimulated to release<br />

cytotoxic mediators that may exacerbate these pseudoallergic-like<br />

responses even further. Experimental and clinical studies have intimated<br />

that there is reason to suspect that acute exposure to brief high-level<br />

concentrations <strong>of</strong> asthmagenic chemicals and the development <strong>of</strong> increased<br />

airway hyperresponsiveness are associated. Thus, it could be assumed that<br />

specific mast cell sensitization—in combination with neurogenic stimuli—<br />

amplify the inflammatory process and airway hyperresponsiveness. The<br />

corresponding increase in vagal activity would increase reflex release <strong>of</strong><br />

acetylcholine and, correspondingly, may enhance airway responsiveness<br />

following the exogenous administration <strong>of</strong> cholinergic agents.<br />

Animal models <strong>of</strong> airway inflammation might allow us to investigate this<br />

relationship further. Models <strong>of</strong> allergic pulmonary inflammation have been<br />

developed in various animal species (Kips et al., 1992), using different


method- ological approaches. In toxicology, the guinea-pig has been used<br />

for decades in order to evaluate the skin sensitizing properties <strong>of</strong> chemicals<br />

and proteins and has also been able to reproduce immediate-onset<br />

pulmonary hypersensitivity responses following inhalation <strong>of</strong> chemical<br />

haptens, their protein-conjugates or antigens. This animal model has<br />

therefore been used to disclose principles governing both the development<br />

<strong>of</strong> pulmonary hypersensitivity and airway hyperreactivity. Due to the<br />

guinea-pig’s abundant amount <strong>of</strong> smooth bronchial musculature, it is used<br />

as a physiologic elicitation model that reproduces bronchospasm upon<br />

challenge to specific or nonspecific stimuli. Other animal models designed<br />

to display many <strong>of</strong> the chronic features <strong>of</strong> hypersensitivity lung diseases<br />

characteristic <strong>of</strong> occupational asthma focus more on the induction <strong>of</strong><br />

airway inflammation, the basic prerequisite for airway hyperreactivity. It<br />

should be noted, however, that the induction <strong>of</strong> asthma in the rat model,<br />

for example, commonly requires more aggressive protocols and more<br />

elaborate techniques to classify responses when compared with the guineapig<br />

elicitation model (vide infra).<br />

The guinea-pig model<br />

J.PAULUHN 131<br />

To date, practically all such models have relied upon the use <strong>of</strong> the guineapig,<br />

a species known to be sensitive for agents inducing<br />

bronchoconstriction and in which respiratory function and respiratory<br />

hypersensitivity can be measured readily. In addition, guinea-pigs are easy<br />

to handle, relatively inexpensive, and produce consistent<br />

bronchoconstrictive reactions. The models have utilized various modes <strong>of</strong><br />

hapten or antigen administration and methods for detecting sensitization,<br />

It has been shown that guinea-pigs sensitized by inhalation exposure to<br />

either a free or a protein-bound chemical can be induced to exhibit changes<br />

in respiratory patterns following inhalation challenge with the same<br />

chemical in the free or in the form <strong>of</strong> its hapten-protein conjugate. In the<br />

guinea-pig no adjuvant is needed for successful lung sensitization. More<br />

recently it has been found that changes in sensitive respiratory parameters<br />

can also be provoked in dermally sensitized guinea-pigs by inhalation<br />

challenge with the free chemical or the hapten-protein conjugate (Botham et<br />

al., 1988; Pauluhn and Eben, 1991; Hayes et al., 1992). In attempting to<br />

derive an animal model that permits the identification <strong>of</strong> asthmagenic lowmolecularweight<br />

chemicals without the presence <strong>of</strong> overriding effects<br />

caused by toxic (irritant) airway inflammation the intradermal route <strong>of</strong><br />

induction appears to be preferable. This route <strong>of</strong> induction also minimizes<br />

the risk <strong>of</strong> potential confounding effects attributable to irritant-induced<br />

nonspecific reactive airways dysfunction as a result <strong>of</strong> previous inhalation<br />

exposures (Briatico-Vangosa et al., 1993).


132 PULMONARY HYPERREACTIVITY TO INDUSTRIAL POLLUTANTS<br />

For challenge exposures it appears to be advantageous to use the free<br />

chemical in slightly irritant concentrations rather than the proteinconjugate<br />

<strong>of</strong> the hapten. It is believed that the in vitro synthesis <strong>of</strong> the<br />

hapten-protein conjugate may not necessarily result in immunologically<br />

identical conjugates when compared with those produced under in vivo<br />

conditions. Also standardized procedures to synthesize and characterize<br />

hapten-protein conjugates <strong>of</strong> multifunctional, highly reactive chemicals are<br />

not yet established. On the other hand, an essential prerequisite for<br />

challenge exposures with the free chemical is the evaluation <strong>of</strong> the irritant<br />

threshold concentration <strong>of</strong> the hapten under investigation. The importance<br />

<strong>of</strong> concentration in distinguishing irritation from sensitization cannot be<br />

overstated and is one <strong>of</strong> the most critical determinants <strong>of</strong> this animal<br />

model.<br />

For volatile, irritant haptens the characteristic feature <strong>of</strong> upper<br />

respiratory tract irritation is the reflexively induced decrease in respiratory<br />

rate which is a common finding in laboratory rodents (Figure 9.1).<br />

Consistent with this approach, naive mice, rats and guinea-pigs were<br />

exposed for 45 min to slightly irritant concentrations <strong>of</strong> phenyl isocyanate<br />

(PI). As evident from Figure 9.1, the exposure to ca. 5 mg PI m −3 air<br />

provoked a decrease in respiratory rate <strong>of</strong> approximately 25–45%. The<br />

observation that remarkable differences in response patterns between mice,<br />

rats and guinea-pigs did not occur demon strate that irritant threshold<br />

concentrations obtained in mice may also be valid for guinea-pigs. Mainly<br />

for volatile chemicals attempts have been made to establish methods for the<br />

measurement and analysis <strong>of</strong> the irritant-induced changes in respiratory<br />

pattern in mice (Vijayaraghavan et al., 1993) and to understand the<br />

mechanisms <strong>of</strong> the irritant receptor stimulation (Nielsen, 1991). For<br />

volatile irritant haptens, such as PI, an unequivocal respiratory<br />

hypersensitivity response is characterized by a shallow rapid breathing<br />

pattern, i.e. a response opposite to that occurring as a result <strong>of</strong> upper<br />

respiratory tract irritation. For volatile irritant haptens this type <strong>of</strong><br />

breathing pattern, however, can only be obtained when using the proteinconjugate<br />

<strong>of</strong> the hapten.<br />

The interpretation <strong>of</strong> changes in respiratory pattern induced by irritant<br />

particulates is less predictable because <strong>of</strong> the size-dependent deposition <strong>of</strong><br />

particles within the respiratory tract. Irritant aerosols that evoke bronchial<br />

or pulmonary irritation may produce changes similar to those occurring<br />

following immediate-onset responses. Therefore, the selection <strong>of</strong> adequate<br />

haptenchallenge concentrations as well as the measurement <strong>of</strong> several<br />

breathing parameters is <strong>of</strong> primary importance. For such chemicals,<br />

currently the relative effectiveness <strong>of</strong> the acute high-concentration<br />

inhalation (single inhalation exposure <strong>of</strong> 15 min) and the high-dose<br />

intradermal route for sensitization <strong>of</strong> guinea-pigs had been investigated<br />

(Pauluhn and Eben, 1991; Pauluhn and Mohr, 1994). The airway function


J.PAULUHN 133<br />

Figure 9.1 Time-response curves for respiratory rate from mice, rats and guineapigs<br />

during single 45-min exposures to appoximately 5 mg m −3 phenyl isocyanate.<br />

Data were normalized on pre-exposure values during 15-min air exposure. Data<br />

points for each concentration are the mean <strong>of</strong> four animals and were averaged for<br />

45 s.<br />

<strong>of</strong> conscious guinea-pigs that were sensitized to and challenged with 4,4′diphenylmethane-diisocyanate<br />

(MDI) aerosol or trimellitic anhydride<br />

(TMA) dust as well as their corresponding proteinconjugates was<br />

monitored plethysmographically. The airway hyper-responsiveness to<br />

subsequently increased inhaled acetylcholine (ACh) concentrations was<br />

assessed 1 day after the hapten challenge (Pauluhn, 1994). In most<br />

instances, selected morphological features <strong>of</strong> the airways (increased<br />

number <strong>of</strong> eosinophils in the bronchial mucosa and lung associated lymph<br />

nodes) were also taken into account.<br />

Collectively, it was noticed that elicitation <strong>of</strong> respiratory hypersensitivity<br />

is concentration-dependent and that challenge concentrations should<br />

slightly exceed the threshold concentration for irritation. The evaluation <strong>of</strong><br />

eosinophils in subepithelial tissues and lung associated lymph nodes<br />

appears to provide an important independent adjunct to measurements <strong>of</strong><br />

respiratory function. The combined assessment <strong>of</strong> specific pathologic<br />

features such as eosinophilic infiltration and the evaluations <strong>of</strong> several


134 PULMONARY HYPERREACTIVITY TO INDUSTRIAL POLLUTANTS<br />

breathing parameters upon acetylcholine and hapten or conjugate challenge<br />

significantly enhance the diagnostic sensitivity <strong>of</strong> the guinea-pig model.<br />

From studies using single, brief high level aerosol or dust exposures for the<br />

induction <strong>of</strong> animals it can be concluded that previous high level exposures<br />

evoke bronchial hyperresponsiveness upon challenge at lower hapten<br />

concentrations when compared with intradermally sensitized animals.<br />

However, guinea-pigs sensitized intradermally to the volatile PI<br />

demonstrated remarkable immediate-type respiratory reactions only upon<br />

challenge with the conjugate and not with slightly irritant concentrations<br />

<strong>of</strong> the free PI. To study if phenyl isocyanate is capable <strong>of</strong> inducing a<br />

reactive airway or an asthma like syndrome, the subsequently described rat<br />

model was used.<br />

The rat model<br />

This animal model focuses on the induction <strong>of</strong> airway inflammation which<br />

comprises most <strong>of</strong> the characteristic features <strong>of</strong> asthma. It has been stated<br />

that respiratory hypersensitivity should depend on two separate factors:<br />

first, the degree <strong>of</strong> allergic airways, and second, the sensitivity to<br />

bronchoconstrictive mediators. Increasing evidence suggests that the<br />

eosinophils play a critical role in the pathogenesis <strong>of</strong> asthma and <strong>of</strong> other<br />

non-allergic hyperresponsive airway diseases. For the induction <strong>of</strong> the<br />

asthmatic state male rats were exposed for 2 consecutive weeks by<br />

inhalation (5 h day −1 , 5 days week −1 ). The target concentrations <strong>of</strong> phenyl<br />

isocyanate were chosen on the basis <strong>of</strong> a single 45-min exposure study<br />

which suggested that approximately 1 mg m −3 air is the irritant threshold<br />

concentration for ‘any’ duration <strong>of</strong> exposure. The 2 week repeated<br />

inhalation study was designed to assess the functional, bio chemical and<br />

morphological signs <strong>of</strong> phenyl isocyanate induced lung disease and their<br />

regression during an observation period <strong>of</strong> approximately 2 months.<br />

The most characteristic features <strong>of</strong> asthma comprise an increased influx<br />

<strong>of</strong> eosinophilic granulocytes into the tissue <strong>of</strong> the airways, secretory cell<br />

hyper-plasia and metaplasia, smooth muscle hypertrophy and hyperplasia,<br />

epithelial desquamation, airway hyperresponsiveness, and eventually partial<br />

occlusion <strong>of</strong> the airway lumen with mucus and cellular debris. The<br />

formation <strong>of</strong> mucus plugs is a regular feature <strong>of</strong> asthma and accounts for<br />

most <strong>of</strong> the clinical, biochemical and physiological abnormalities.<br />

Histopathological evaluation <strong>of</strong> the respiratory tract indicated a<br />

bronchiolitis obliterans and smooth muscle hypertrophy in rats exposed to<br />

approximately 7 mg m −3 air, whereas only minimal effects were found<br />

following 4 mg m −3 air. Lung function measurements revealed that some rats<br />

were hyperresponsive to an ACh-stimulus. As shown in Figure 9.2, also the<br />

increase in shunt blood (Q s/Q t) anddecrease in forced expiratory flow rates<br />

(MMEF) as well as mucus products (sialomucins), polymorphonuclear


cells, including eosinophils, in the bronchoalveolar lavage fluid (BALF)<br />

were consistent with an asthma like syndrome. As evident from Figure 9.2,<br />

the changes observed in rats exposed to 7 mg m −3 air did not fully regress<br />

during an observation period <strong>of</strong> approximately 2 months.<br />

Conclusion<br />

J.PAULUHN 135<br />

Figure 9.2 Relative comparison <strong>of</strong> sensitive diagnostic parameters in rats exposed to<br />

either 0 (air), 1, 4 and 7 mg PI m −3 air for 2 consecutive weeks (6 h day −1 , 5 days<br />

week −1 ). The measurements were performed in weeks 3 and 9. Abbreviations:<br />

MMEF: Maximal mid-expiratory flow rate, Q s/Q t: venous admixture, PMN:<br />

polymorphonuclear cells, Eos: eosinophilic granulocytes, Sialomucins: total sialic<br />

acid (after hydrolysis).<br />

Experimental evidence suggests that changes within the respiratory tract<br />

leading to the reactive airway dysfunction syndrome and/or asthma are<br />

fully consistent with an inflammatory response involving tissue <strong>of</strong> direct<br />

contact. The toxicity <strong>of</strong> irritant chemicals known to induce such illness is<br />

highly focal, and the variability <strong>of</strong> response in different regions <strong>of</strong> the<br />

respiratory tract could be a result <strong>of</strong> the actual concentration <strong>of</strong> the<br />

toxicant reaching various airway levels. Determination <strong>of</strong> immunologic<br />

etiology is particularly important for chemical allergy since all recognized<br />

low-molecular-weight chemical sensitizers are also respiratory irritants and<br />

in sufficient concentrations can cause airway constriction by<br />

nonimmunological mechanisms. As shown by studies using phenyl


136 PULMONARY HYPERREACTIVITY TO INDUSTRIAL POLLUTANTS<br />

isocyanate, damage <strong>of</strong> the airways is characterized by a steep concentrationresponse<br />

curve. Based on acute 45-min exposure <strong>of</strong> rats the threshold for<br />

respiratory tract irritation is approximately 1 mg m −3 . Exposures equaling<br />

this concentration were tolerated without exposure-related effects, whether<br />

exposure occurred singly for 45-min or repeatedly for 2 weeks. Marginal<br />

effects were observed at 4 mg m −3 , all effects, including mortality, were<br />

produced at 7 mg m −3 . This demonstrates that selection <strong>of</strong> appropriate<br />

exposure concentrations appears to be most critical in the rat model. The<br />

assessment <strong>of</strong> diagnostic sensitivity <strong>of</strong> the methods used to probe damage<br />

to the respiratory tract demonstrated that respiratory function data, blood<br />

gas measurements, and BALF analysis facilitate a meaningful interpretation<br />

<strong>of</strong> the effects observed and are important adjuncts to common inhalation<br />

toxicological studies on rats to describe quantitatively the diseased state <strong>of</strong><br />

the lung.<br />

The guinea-pig model is experimentally less demanding and therefore can<br />

suitably be used as a screening test for respiratory sensitization, as far as<br />

the limitations <strong>of</strong> this model are taken into account. Studies on guinea-pigs<br />

demonstrate that elicitation <strong>of</strong> respiratory hypersensitivity is<br />

challengeconcentration dependent and that the concentrations used should<br />

slightly exceed the threshold concentration for irritation to maximize the<br />

magnitude <strong>of</strong> the response. However, sensitization by inhalation may<br />

increase the susceptibility to irritant stimuli and thus confounds the<br />

selection <strong>of</strong> the most appropriate concentration for challenge. The<br />

combined approach <strong>of</strong> evaluating several breathing parameters, e.g.<br />

respiratory rate, flow- and volume-derived parameters, during both the<br />

hapten (free or conjugated) and the ACh challenge provides a promising<br />

method to distinguish specific and nonspecific hypersensitivity responses.<br />

Furthermore, it is critically important to assess the respiratory irritant<br />

potency <strong>of</strong> the compound under investigation. For potent irritant<br />

substances such as volatile isocyanates, challenge with the haptenprotein<br />

conjugate minimizes the likelihood to confound specific hypersensitivity<br />

responses with those evoked merely by irritation. Taking all imponderable<br />

factors into consideration, it appears that the guinea-pig intradermalinduction<br />

inhalation-challenge protocol is adequately susceptible to identify<br />

potent respiratory tract sensitizers. However, if the airway inflammation<br />

related features <strong>of</strong> asthma are the endpoints <strong>of</strong> primary interest other<br />

animal models appear to be more appropriate.<br />

References<br />

BARNES, P.J., BARANIUK, J.N. and BELVISI, M.G., 1991a, Neuropeptides in the<br />

respiratory tract (Part II). Am. Rev. Respir. Dis., 144, 1391–9.


J.PAULUHN 137<br />

BARNES, P.J., CHUNG, K.F., PAGE, C.P., 1991b, Pharmacology <strong>of</strong> asthma,<br />

Chapter 3, Inflammatory Mediators in Page, C.P. and Barnes, P.J. (Eds.), pp<br />

54–106. Handbook <strong>of</strong> Experimental Pharmacology, Berlin, Heidelberg, New<br />

York: Springer-Verlag.<br />

BOTHAM, P.A., HEXT, P.M., RATTRAY, N.J., WALSH, S.T. and WOOD-<br />

COCK, D.R., 1988, Sensitisation <strong>of</strong> guinea-pigs by inhalation exposure to lowmolecular-weight<br />

chemicals, Toxicol. Lett., 41, 159–73.<br />

BRIATICO-VANGOSA, G, BRAUN, C.J.L., COOKMAN, G., HOFMANN, T.,<br />

KIMBER, I., LOVELESS, S.E., MORROW, T., PAULUHN, J., SØRENSEN, T.<br />

and NIESSEN, H.J., 1993, Respiratory Allergy. ECETOC Monograph No. 19.<br />

BROOKS, S.M., WEISS, M.A. and BERNSTEIN, I.L., 1985, Reactive airway dysfunction<br />

syndrome (RADS). Persistent asthma syndrome after high level irritant<br />

exposure, Chest, 88, 376–84.<br />

HAYES, J.P., DANIEL, H.R., TEE, R.D., BARNES, P.J., NEWMAN-TAYLOR,<br />

A.J. and CHUNG, K.F., 1992, Bronchial hyperreactivity after inhalation <strong>of</strong><br />

trimellitic anhydride dust in guinea-pigs after intradermal sensitization to the<br />

free hapten, Am. Rev. Respir. Dis., 146, 1311–14.<br />

KAY, A.B., 1991, Asthma and inflammation, J. Allergy Clin. Immunol., 87, 893–<br />

910.<br />

KIPS, J.C., CUVELIER, C.A. and PAUWELS, R.A., 1992, Effect <strong>of</strong> acute and<br />

chronic antigen inhalation on airway morphology and responsiveness in<br />

actively sensitized rats, Am. Rev. Respir. Dis., 145, 1306–10.<br />

NIELSEN, G.D., 1991, Mechanisms <strong>of</strong> activation <strong>of</strong> the sensory irritant receptor by<br />

airborne chemicals, Crit. Rev. Toxicol, 21, 183–208.<br />

PAULUHN, J., 1994, Test methods for respiratory sensitization in use <strong>of</strong><br />

mechanistic information in risk assessment, EUROTOX Proceedings, Arch.<br />

Toxicol., suppl. 16, 77–86.<br />

PAULUHN, J. and EBEN, A., 1991, Validation <strong>of</strong> a non-invasive technique to<br />

assess immediate or delayed onset <strong>of</strong> airway hypersensitivity in guinea-pigs, J.<br />

Appl. Toxicol, 11, 423–31.<br />

PAULUHN, J. and MOHR, U., 1994, Assessment <strong>of</strong> respiratory hypersensitivity in<br />

guinea-pigs sensitized to diphenylmethane-4,4'-diisocyanate (MDI) and<br />

challenged with MDI, acetylcholine or MDI-albumin conjugate, <strong>Toxicology</strong> (in<br />

press).<br />

VIJAYARAGHAVAN, R., SCHAPER, M., THOMPSON, R., STOCK, M.F. and<br />

ALARIE, Y., 1993, Characteristic modifications <strong>of</strong> the breathing pattern <strong>of</strong><br />

mice to evaluate the effects <strong>of</strong> airborne chemicals on the respiratory tract, Arch.<br />

Toxicol, 67, 478–90.


10<br />

Mechanisms <strong>of</strong> Pulmonary Sensitization<br />

IAN KIMBER<br />

Zeneca Central <strong>Toxicology</strong> Laboratory, Macclesfield<br />

Introduction<br />

A wide range <strong>of</strong> chemicals is known to cause allergic contact dermatitis. It<br />

is apparent, however, that chemicals also have the potential to provoke other<br />

forms <strong>of</strong> allergy and <strong>of</strong> growing concern is pulmonary sensitization.<br />

Examples <strong>of</strong> chemicals identified as human respiratory allergens are listed<br />

in Table 10.1. Respiratory allergic hypersensitivity is characterized by<br />

pulmonary reactions which occur normally in only a proportion, and<br />

frequently in only a small proportion, <strong>of</strong> exposed individuals. In those who<br />

are sensitized, respiratory reactions can be provoked by atmospheric<br />

concentrations <strong>of</strong> the causative chemical allergen which were tolerated<br />

previously and which are without effect in the non-sensitized population<br />

(Newman Taylor, 1988). Almost invariably there is a latent period between<br />

the onset <strong>of</strong> exposure and the development <strong>of</strong> respiratory symptoms such<br />

as asthma and rhinitis.<br />

By definition, allergy, including sensitization <strong>of</strong> the respiratory tract,<br />

results from the stimulation <strong>of</strong> specific immune responses by the causative<br />

agent. Although it is assumed frequently that effective allergic sensitization<br />

<strong>of</strong> the respiratory tract results largely or wholly from inhalation exposure,<br />

this is not necessarily the case. Allergic reactions manifest in a particular<br />

organ commonly result from the local provocation by the inducing agent <strong>of</strong><br />

a systemically sensitized individual. There is no reason to suppose that the<br />

quality <strong>of</strong> immune response necessary for sensitization <strong>of</strong> the respiratory<br />

tract may not result from exposure to the chemical allergen at a different<br />

site. Consistent with this is evidence that occupational respiratory allergy<br />

may be caused by dermal contact with the chemical (Karol, 1986; Nemery<br />

and Lenaerts, 1993). Furthermore, it has been reported that respiratory<br />

rate changes can be provoked by inhalation exposure <strong>of</strong> guinea pigs<br />

sensitized previously by either topical or subcutaneous treatment with the<br />

same chemical (Karol et al., 1981; Rattray et al., 1994). Despite the fact<br />

that, in practice, pulmonary sensitization may not be caused exclusively by


Table 10.1 Chemicals identified as human respiratory allergens<br />

I.KIMBER 139<br />

inhalation <strong>of</strong> the chemical allergen, it is likely that this is an important<br />

route <strong>of</strong> exposure in the occupational setting.<br />

It is well established that respiratory sensitization caused by protein<br />

aeroallergens is effected by IgE antibody. This class <strong>of</strong> antibody in man is<br />

homocytotropic and is able to associate, via specific membrane receptors,<br />

with mast cells, including mast cells in the respiratory tract. Following<br />

subsequent exposure <strong>of</strong> the sensitized individual to the same allergen, mast<br />

cell-bound IgE is cross-linked and this, in turn, results in mast cell<br />

degranulation and the release <strong>of</strong> both preformed and newly-synthesized<br />

mediators which provoke acute inflammatory reactions. In the case <strong>of</strong><br />

sensitization <strong>of</strong> the respiratory tract caused by chemicals, however, an<br />

invariable association with the presence <strong>of</strong> specific IgE antibody has failed<br />

to emerge. Although IgE antibody specific for all recognized chemical<br />

respiratory allergens has been demonstrated, it is not uncommonly the case<br />

that individuals displaying symptoms <strong>of</strong> pulmonary hypersensitivity have<br />

been reported to lack demonstrable IgE. This may suggest that<br />

immunological processes independent <strong>of</strong> IgE antibody may play a decisive<br />

role in the induction <strong>of</strong> respiratory sensitization. An alternative explanation<br />

is that inappropriate or insensitive techniques have been employed for<br />

serological analysis and that IgE antibody may be associated more<br />

commonly than suspected previously with chemical respiratory allergy. In<br />

this context it is relevant that it has been found that positive skin prick<br />

tests can be provoked in patients sensitized to acid anhydrides who, on the<br />

basis <strong>of</strong> radioallergosorbent tests (RAST), were found to lack measurable<br />

levels <strong>of</strong> serum IgE antibody (Drexler et al., 1993). Despite the absence <strong>of</strong><br />

formal confirmation that there exists a universal causal relationship<br />

between specific IgE and pulmonary hypersensitivity induced by chemicals,


140 MECHANISMS OF PULMONARY SENSITIZATION<br />

it remains likely that this class <strong>of</strong> antibody is responsible, in at least the<br />

majority <strong>of</strong> cases, for the acute onset symptoms associated with respiratory<br />

allergy (Karol et al., 1994).<br />

The induction and regulation <strong>of</strong> IgE responses<br />

IgE antibody responses are subject to a variety <strong>of</strong> immunoregulatory<br />

control mechanisms. Chief among these are the stimulatory and inhibitory<br />

actions <strong>of</strong> cytokines which serve to influence the induction and duration <strong>of</strong><br />

IgE responses. It has been found in mice that interleukin 4 (IL-4) is<br />

necessary for the initiation and maintenance <strong>of</strong> IgE antibody production<br />

(Finkelman et al., 1988b). The essential role for this cytokine in IgE<br />

responses has been emphasized further by studies <strong>of</strong> mice homozygous for<br />

a mutation that inactivates the gene for IL-4. These animals lack detectable<br />

serum IgE and fail to mount IgE responses (Kuhn et al., 1991). Importantly,<br />

in mice which produce constitutively high levels <strong>of</strong> IL-4, significantly<br />

elevated concentrations <strong>of</strong> serum IgE are evident (Burstein et al., 1991). A<br />

balance to the promotional influence <strong>of</strong> IL-4 is provided by interferon<br />

(IFN- ), a cytokine which exerts an inhibitory affect on IgE responses<br />

(Finkelman et al., 1988a). The reciprocal antagonistic activity <strong>of</strong> these<br />

cytokines is not restricted to the mouse, IL-4 and IFN- have been found to<br />

regulate human IgE production (Del Prete et al., 1988; Pene et al., 1988).<br />

The cytokines which influence the integrity <strong>of</strong> IgE responses are the<br />

products <strong>of</strong> discrete subpopulations <strong>of</strong> T helper (Th) cells, lymphocytes<br />

characterized by possession <strong>of</strong> the CD4 membrane determinant. It has been<br />

found in both mouse and man that there exists a functional heterogeneity<br />

among Th cells. Two major populations, designated Th 1 and Th 2, have<br />

been described (Mosmann and C<strong>of</strong>fman, 1989; Romagnani, 1991). It is<br />

believed currently that these subsets represent the most differentiated forms<br />

<strong>of</strong> Th cells and develop from less mature precursors as the immune<br />

response evolves (Mosmann et al., 1991). The major functional distinction<br />

between Th 1 and Th 2 cells resides in the spectrum <strong>of</strong> cytokines which they<br />

elaborate (Mosmann and C<strong>of</strong>fman, 1989). The cytokine products <strong>of</strong><br />

murine Th 1 and Th 2 cells are displayed in Table 10.2.<br />

It has been reported previously that chemicals known to cause<br />

respiratory hypersensitivity in man induce in mice immune responses<br />

characteristic <strong>of</strong> Th 2 cell activation, stimulate the production <strong>of</strong> specific IgE<br />

antibody and cause an increase in the serum concentration <strong>of</strong> IgE.<br />

Conversely, chemical allergens considered not to cause respiratory<br />

sensitivity, but which are nevertheless able to induce skin sensitization,<br />

elicit instead Th 1-type responses. In the latter case, immune responses are<br />

characterized by comparatively high levels <strong>of</strong> IgG2a antibody (an isotype<br />

known to be upregulated by IFN- ) and the absence <strong>of</strong> specific IgE<br />

(Dearman and Kimber, 1991, 1992; Dearman et al., 1991, 1992a,c,d,


Table 10.2 The cytokine products <strong>of</strong> murine Th 1 and Th 2 cells<br />

From: Mosmann and C<strong>of</strong>fman (1989).<br />

I.KIMBER 141<br />

1994). The implication is that certain chemicals favour the development <strong>of</strong><br />

Th 2 cells which will then synthesize and secrete IL-4 and thereby encourage<br />

IgE antibody responses and mast cell sensitization. The converse is that<br />

other classes <strong>of</strong> chemical allergen preferentially stimulate Th1 cells and IFNproduction.<br />

Such conditions will be nonpermissive for IgE antibody<br />

production and cell-mediated immune responses, including contact<br />

sensitization, will be favoured instead. A selective stimulation by different<br />

classes <strong>of</strong> chemical sensitizers <strong>of</strong> divergent Th responses may provide an<br />

explanation at the cellular level for the observation that chemicals vary<br />

with respect to the nature <strong>of</strong> allergic reactions that they will elicit<br />

preferentially in man. The stimulation by chemical allergens <strong>of</strong><br />

differentiated Th cell responses may have implications for allergic disease<br />

other than the regulation <strong>of</strong> IgE antibody. It is known for instance that<br />

IL-3, IL-4 and IL-10, all <strong>of</strong> which are products <strong>of</strong> murine Th 2 cells<br />

(Table 10.2), act as mast cell growth factors or c<strong>of</strong>actors (Smith and<br />

Rennick, 1986; Thompson-Snipes et al., 1991). Moreover, IL-5 is a growth<br />

and differentiation factor for eosinophils (Yokota et al., 1987) and serves<br />

to regulate the accumulation <strong>of</strong> these cells at the site <strong>of</strong> allergeninduced<br />

hypersensitivity reactions in the respiratory tract (Gulbenkian et al., 1992).<br />

It has been found recently that the cytokines IL-3 and IL-4 also enhance the<br />

secretory activity <strong>of</strong> mast cells following activation (Coleman et al., 1993).<br />

Antagonistic and inhibitory influences <strong>of</strong> Th cell products may also affect<br />

the elicitation <strong>of</strong> allergic reactions. It has been found that IFN- not only<br />

suppresses the secretory function <strong>of</strong> mast cells (Holliday et al., 1994), but<br />

also antagonizes the antigen-induced infiltration <strong>of</strong> eosinophils into the<br />

respiratory tract <strong>of</strong> sensitized mice (Iwamoto et al., 1993). Contact allergic<br />

reactions may in theory be regulated by Th 2 cytokines. It has been shown<br />

that IL-4 and IL-10 act in concert to inhibit Th 1 cell function and to


142 MECHANISMS OF PULMONARY SENSITIZATION<br />

depress cell-mediated immunity (Powrie et al., 1993) and that Il–4 is able<br />

to reduce significantly the severity <strong>of</strong> contact allergic reactions in mice<br />

(Gautam et al., 1992).<br />

Taken together the available data suggest that the selective stimulation<br />

<strong>of</strong> Th cell responses and the consequent balance created between Th 1- and<br />

Th 2-derived cytokines will have an important impact on both the induction<br />

and elicitation stages <strong>of</strong> allergy. It is perhaps not surprising, therefore, that<br />

there is increasing evidence for selective Th responses in human allergic<br />

disease. Clones <strong>of</strong> T lymphocytes specific for aeroallergens such as house<br />

dust mite and grass pollen, which cause IgE-mediated respiratory allergic<br />

reactions in susceptible individuals, have been shown to elaborate Th 2<br />

cytokines, but not IFN- (Parronchi et al., 1991). A predominance <strong>of</strong> the<br />

Th 2-type cells has been found at sites <strong>of</strong> skin reactions in atopic individuals<br />

(Kay et al., 1991) and increased numbers <strong>of</strong> IL-4 + T lymphocytes have been<br />

identified in the nasal mucosa in allergen-induced rhinitis (Ying et al., 1994).<br />

By contrast, human immune responses to nickel, a common cause <strong>of</strong><br />

allergic contact dermatitis, are characterized by the selective activation <strong>of</strong><br />

Th 1-type cells. Allergen-specific T lymphocyte clones isolated from the<br />

peripheral blood <strong>of</strong> patients sensitized to nickel have been found to secrete<br />

only low or undetectable amounts <strong>of</strong> IL-4 and IL-5, but high levels <strong>of</strong> IFN-<br />

(Kapsenberg et al., 1991).<br />

Although the relative contribution <strong>of</strong> Th 1 and Th 2 cells during immune<br />

responses, and in particular the relative availability <strong>of</strong> IL-4 and IFN- , is<br />

likely to play a predominant role in the regulation <strong>of</strong> IgE antibody, other<br />

factors may be relevant. Not least, the priming <strong>of</strong> Th 1 cells for the<br />

production <strong>of</strong> IFN- may in turn be dependent upon the action <strong>of</strong> another<br />

cytokine, interleukin 12 (IL-12) (Manetti et al., 1994; Morris et al., 1994;<br />

Schmitt et al., 1994). It has been demonstrated also that CD8 + T<br />

lymphocytes exert an important immunoregulatory influence on IgE<br />

responses (Kemeny et al., 1994; Renz et al., 1994), possibly via downregulation<br />

<strong>of</strong> CD4 + Th 2 cell development (Noble et al., 1993).<br />

It is clear that conditions outwith the immune system also influence the<br />

magnitude <strong>of</strong> IgE responses. Certainly genetic predisposition is an<br />

important, although poorly understood factor. In addition, there have been<br />

suggestions that cigarette smoking and exposure to certain environmental<br />

pollutants may result in increased IgE levels and may also serve to<br />

aggravate asthma (Zetterstrom et al., 1981; Muranka et al., 1986;<br />

Wardlaw, 1993).<br />

Cell-mediated immune responses in chemical respiratory<br />

allergy<br />

The elicitation <strong>of</strong> chemical respiratory hypersensitivity may be associated<br />

with both immediate-onset and late phase reactions. While IgE antibody


and local degranulation <strong>of</strong> mast cells may be necessary for acute<br />

symptoms, late asthmatic responses appearing some hours following<br />

exposure are characterized by an infiltration <strong>of</strong> mononuclear cells and<br />

increased numbers <strong>of</strong> leucocytes in bronchoalveolar lavage fluid. Chronic<br />

inflammation is an important component <strong>of</strong> asthma and, in addition to<br />

mononuclear cell accumulation, is characterized by mucus production, the<br />

destruction and sloughing <strong>of</strong> airway epithelial cells and subepithelial<br />

fibrosis secondary to collagen deposition. Eosinophils, acting together with<br />

infiltrating T lymphocytes, play a pivotal role in chronic bronchial<br />

inflammation (Corrigan and Kay, 1992). It is apparent also that the<br />

generation <strong>of</strong> eosinophilia in the respiratory tract is influenced markedly by<br />

Th cell products. As described previously, IL-5 effects the accumulation <strong>of</strong><br />

eosinophils at the site <strong>of</strong> hypersensitivity reactions in respiratory tissues,<br />

while IFN- , secondary to an inhibition <strong>of</strong> CD4 + cell infiltration,<br />

antagonizes this process (Gulbenkian et al., 1992; Iwamoto et al., 1993). It<br />

may prove that the cell-mediated immune processes relevant to the<br />

development <strong>of</strong> respiratory hypersensitivity and asthma are also a function<br />

<strong>of</strong> Th cell heterogeneity. Certainly the stimulation <strong>of</strong> Th 2 cell activation<br />

will have pr<strong>of</strong>ound effects on all stages <strong>of</strong> respiratory allergy. The<br />

infiltration <strong>of</strong> such cells into sites <strong>of</strong> encounter with inducing allergen, a<br />

process perhaps facilitated by vasodilation resulting from mast cell<br />

degranulation, will provide a local source <strong>of</strong> cytokines such as IL-4 and<br />

IL-5. Mast cell secretory activity will be potentiated by the former and<br />

eosinophil accumulation triggered by the latter. That Th 2 cells do in fact<br />

accumulate in the area <strong>of</strong> immediate-type hypersensitivity reactions is<br />

supported by the studies <strong>of</strong> Kay et al. (1991) who demonstrated that the<br />

cells infiltrating lesional skin at the sites <strong>of</strong> late phase cutaneous reactions<br />

in atopic patients produce IL-3, IL-4, IL-5 and GM-CSF, but not IFN- .<br />

Practical applications<br />

I.KIMBER 143<br />

In the course <strong>of</strong> investigations designed to examine the characteristics <strong>of</strong><br />

immune responses induced in mice by chemical sensitizers it was found<br />

that only those materials known to cause respiratory hypersensitivity in<br />

man provoked in mice a substantial increase in the serum concentration <strong>of</strong><br />

IgE; a phenomenon thought to reflect the selective stimulation <strong>of</strong> Th 2 celltype<br />

responses by this class <strong>of</strong> allergen. It was observed also that contact<br />

allergens known or suspected not to cause occupational respiratory<br />

hypersensitivity failed to result in similar changes in serum IgE levels<br />

(Dearman and Kimber, 1991, 1992; Dearman et al., 1992a,d). The<br />

differential ability <strong>of</strong> chemical respiratory and contact allergens to<br />

stimulate changes in the concentration <strong>of</strong> serum IgE in mice forms the basis<br />

<strong>of</strong> a novel approach to the identification <strong>of</strong> chemicals which have the<br />

potential to cause sensitization <strong>of</strong> the respiratory tract. This method, the


144 MECHANISMS OF PULMONARY SENSITIZATION<br />

mouse IgE test (Dearman et al., 1992b, Kimber and Dearman, 1993) is<br />

being evaluated currently in the context <strong>of</strong> internal and inter-laboratory<br />

validation studies.<br />

References<br />

BERNSTEIN, D.I., PATTERSON, R. and ZEISS, C.R., 1982, Clinical and<br />

immunological evaluation <strong>of</strong> trimellitic anhydride- and phthalic anhydrideexposed<br />

workers using a questionnaire and comparative analysis <strong>of</strong> enzymelinked<br />

immunosorbent and radioimmunoassay studies, Journal <strong>of</strong> Allergy and<br />

Clinical Immunology, 69, 311–18.<br />

BURSTEIN, H.J., TEPPER, R.I., LEDER, P. and ABBAS, A.K., 1991, Humoral<br />

immune function in IL-4 transgenic mice, Journal <strong>of</strong> Immunology, 147, 2950–<br />

6.<br />

CARTIER, A., CHAN H., MALO, J-L., PINEAU, L., TSE, K.S. and<br />

CHANYEUNG, M., 1986, Occupational asthma caused by eastern white<br />

cedar (Thuja occidentalis) with demonstration that plicatic acid is present in this<br />

wood dust and is the causal agent, Journal <strong>of</strong> Allergy and Clinical<br />

Immunology, 77, 639–45.<br />

COLEMAN, J.W., HOLLIDAY, M.R., KIMBER, I., ZSEBO, K.M. and GALLI, S.<br />

J., 1993, Regulation <strong>of</strong> mouse peritoneal mast cell secretory function by stem<br />

cell factor, IL-3 or IL-4, Journal <strong>of</strong> Immunology, 150, 556–62.<br />

CORRIGAN, C.J. and KAY, A.B., 1992, T cells and eosinophils in the<br />

pathogenesis <strong>of</strong> asthma, Immunology Today, 13, 501–7.<br />

DEARMAN, R.J. and KIMBER, I., 1991, Differential stimulation <strong>of</strong> immune<br />

function by respiratory and contact chemical allergens, Immunology, 72, 563–<br />

70.<br />

DEARMAN, R.J. and KIMBER, I., 1992, Divergent immune responses to<br />

respiratory and contact chemical allergens: antibody elicited by phthalic<br />

anhydride and oxazolone, Clinical and Experimental Allergy, 22, 241–50.<br />

DEARMAN, R.J., HEGARTY, J.M. and KIMBER, I., 1991, Inhalation exposure<br />

<strong>of</strong> mice to trimellitic anhydride induces both IgG and IgE anti-hapten antibody,<br />

International Archives <strong>of</strong> Allergy and Applied Immunology, 95, 70–6.<br />

DEARMAN, R.J., BASKETTER, D.A., COLEMAN, J.W. and KIMBER, I., 1992a,<br />

The cellular and molecular basis for divergent allergic responses to chemicals,<br />

Chemical-Biological Interactions, 84, 1–10.<br />

DEARMAN, R.J., BASKETTER, D.A. and KIMBER, I., 1992b, Variable effects <strong>of</strong><br />

chemical allergens on serum IgE concentrations in mice. Preliminary evaluation<br />

<strong>of</strong> a novel approach to the identification <strong>of</strong> respiratory sensitizers, Journal <strong>of</strong><br />

Applied <strong>Toxicology</strong>, 12, 317–23.<br />

DEARMAN, R.J., MITCHELL, J.A., BASKETTER, D.A. and KIMBER, I., 1992c,<br />

Differential ability <strong>of</strong> occupational chemical contact and respiratory allergens<br />

to cause immediate and delayed dermal hypersensitivity reactions in mice,<br />

International Archives <strong>of</strong> Allergy and Immunology, 97, 315–21.


I.KIMBER 145<br />

DEARMAN, R.J., SPENCE, L.M. and KIMBER, I., 1992d, Characterization <strong>of</strong><br />

murine immune responses to allergenic diisocyanates, <strong>Toxicology</strong> and Applied<br />

Pharmacology, 112, 190–7.<br />

DEARMAN, R.J., RAMDIN, L.S.P., BASKETTER, D.A. and KIMBER, I. 1994,<br />

Inducible interleukin-4-secreting cells provoked in mice during chemical<br />

sensitization, Immunology, 81, 551–7.<br />

DEL PRETE, G., MAGGI, E., PARRONCHI, P., CHRETIEN, I., TIRI, D.,<br />

MACCHIA, D., RICI, M., ANSARI, A.A. and ROMAGNANI, S., 1988, IL-4<br />

is an essential factor for the IgE synthesis induced in vitro by human T cell<br />

clones and their supernatants, Journal <strong>of</strong> Immunology, 140, 4193–8.<br />

DOCKER, A., WATTIE, J.M., TOPPING, M.D., LUCZYNSKA, C.M., NEWMAN<br />

TAYLOR, A.J., PICKERING, C.A.C., THOMAS, P. and GOMPERTZ, D.,<br />

1987, Clinical and immunological investigations <strong>of</strong> respiratory disease in<br />

workers using reactive dyes, British Journal <strong>of</strong> lndustrial Medicine, 44, 534–41.<br />

DREXLER, H., SCHALLER, K-H., WEBER, A., LETZEL, S. and LEHNERT, G.,<br />

1993, Skin prick tests with solutions <strong>of</strong> acid anhydrides in acetone,<br />

International Archives <strong>of</strong> Allergy and Immunology, 100, 251–5.<br />

FINKELMAN, F.D., KATONA, I.M., MOSMANN, T.R. and COFFMAN, R.L.,<br />

1988a, IFN- regulates the isotypes <strong>of</strong> Ig secreted during in vivo humoral<br />

immune responses, Journal <strong>of</strong> Immunology, 140, 1022–7.<br />

FINKELMAN, F.D., KATONA, I.M., URBAN, J.F.JR, HOLMES, J., OHARA. J.,<br />

TUNG, A.S., SAMPLE, J.G. and PAUL, W.E., 1988b, IL-4 is required to<br />

generate and sustain in vivo IgE responses, Journal <strong>of</strong> Immunology, 141, 2335–<br />

41.<br />

GAUTAM, S.C., CHIKKALA, N.F. and HAMILTON, T.A., 1992,<br />

Antiinflammatory action <strong>of</strong> IL-4. Negative regulation <strong>of</strong> contact sensitivity to<br />

trinitrochlorobenzene, Journal <strong>of</strong> Immunology, 148, 1411–15.<br />

GULBENKIAN, A.R., EGAN, R.W., FERNANDEZ, X., JONES, H., KREUTNER,<br />

W., KUNG, T., PAYVANDI, F., SULLIVAN, L., ZURCHER, J.A. and<br />

WATNIK, A.S., 1992, Interleukin-5 modulates eosinophil accumulation in<br />

allergic guinea pig lung, American Review <strong>of</strong> Respiratory Diseases, 146, 263–9.<br />

HOLLIDAY, M.R., BANKS, E.M. S., DEARMAN, R.J., KIMBER, I. and<br />

COLEMAN, J.W., 1994. Interactions <strong>of</strong> IFN- with IL-3 and IL-4 in the<br />

regulation <strong>of</strong> serotonin and arachidonate release from peritoneal mast cells,<br />

Immunology, 82, 70–4.<br />

HOWE, W., VENABLES, K.M., TOPPING, M.D., DALLY, M.B., HAWKINS, R.,<br />

LAW, J.S. and NEWMAN TAYLOR, A.J., 1983, Tetrachlorophthalic<br />

anhydride asthma: evidence for specific IgE antibody, Journal <strong>of</strong> Allergy and<br />

Clinical Immunology, 7l, 5–11.<br />

IWAMOTO, I., NAKAJIMA, H., ENDO, H. and YOSHIDA, S., 1993, Interferon<br />

regulates antigen-induced eosinophil recruitment into the mouse airways by<br />

inhibiting infiltration <strong>of</strong> CD4 + T cells, Journal <strong>of</strong> Experimental Medicine, 177,<br />

573–6.<br />

KAPSENBERG, M.L., WIERENGA, E.A., Bos, J.D. and JANSEN, H.M., 1991,<br />

Functional subsets <strong>of</strong> allergen-reactive human CD4 + T cells, Immunology<br />

Today, 12, 392–5.<br />

KAROL, M.H., 1986, Respiratory effects <strong>of</strong> inhaled isocyanates, CRC Critical<br />

Reviews in <strong>Toxicology</strong>, 16, 349–79.


146 MECHANISMS OF PULMONARY SENSITIZATION<br />

KAROL, M.H., HAUTH, B.A., RILEY, E.J. and MAGRENI, C.M., 1981, Dermal<br />

contact with toluene diisocyanate (TDI) produces respiratory tract<br />

hypersensitivity in guinea pigs, <strong>Toxicology</strong> and Applied Pharmacology, 58,<br />

221–30.<br />

KAROL, M.H., TOLLERUD, D.J., CAMPBELL, T.P., FABBRI, L., MAESTRELLI,<br />

P., SAETTA, M. and MAPP, C.E., 1994, Predictive value <strong>of</strong> airways hyperresponsiveness<br />

and circulating IgE for identifying types <strong>of</strong> responses to toluene<br />

diisocyanate inhalation challenge, American Journal <strong>of</strong> Respiratory and<br />

Critical Care Medicine, 149, 611–15.<br />

KAY, A.B., YING, S., VARNEY, V., GAGA, M., DURHAM, S.R., MOQBEL, R.,<br />

WARDLAW, A.J. and HAMID, Q., 1991, Messenger RNA expression <strong>of</strong> the<br />

cytokine gene cluster, interleukin 3 (IL-3), IL-4, IL-5 and granulocyte/<br />

macrophage colony stimulating factor, in allergen-induced last phase reactions<br />

in atopic subjects, Journal <strong>of</strong> Experimental Medicine, 173, 775–8.<br />

KEMENY, D.M., NOBLE, A., HOLMES, B.J. and DIAZ-SANCHEZ, D., 1994,<br />

Immune regulation: a new role for the CD8 + T cell, Immunology Today, 15,<br />

107–10.<br />

KIMBER, I. and DEARMAN, R.J., 1993, Approaches to the identification and<br />

classification <strong>of</strong> chemical allergens in mice, Journal <strong>of</strong> Pharmacological and<br />

Toxicological Methods, 29, 11–16.<br />

KUHN, R., RAJEWSKY, K. and MULLER, W., 1991, Generation and analysis <strong>of</strong><br />

interleukin-4 deficient mice, Science, 254, 707–10.<br />

MACCIA, C.A. BERNSTEIN, I.L., EMMETT, E.A. and BROOKS, S.M. 1976, In<br />

vitro demonstration <strong>of</strong> specific IgE in phthalic anhydride sensitivity, American<br />

Review <strong>of</strong> Respiratory Disease, 113, 701–4.<br />

MANETTI, R., GEROSA, F., GIUDIZI, M.G., BIAGIOTTI, R., PARRONCHI, P.,<br />

PICCINNI, M-P., SAMPOGNARO, S., MAGGI, E., ROMAGNANI, S. and<br />

TRI NCHIERI, G., 1994, Interleukin 12 induces stable priming for interferon<br />

(IFN- ) production during differentiation <strong>of</strong> human T helper (Th) cells and<br />

transient IFN- production in established Th 2 cell clones, Journal <strong>of</strong><br />

Experimental Medicine, 179, 1273–83.<br />

MOLLER, D.R., GALLAGHER, J.S., BERNSTEIN, D.I., WILCOX, T.G.,<br />

BURROUGHS, H.E. and BERNSTEIN, I.L., 1985, Detection <strong>of</strong> IgE-mediated<br />

respiratory sensitization in workers exposed to hexahydrophthalic anhydride,<br />

Journal <strong>of</strong> Allergy and Clinical Immunology, 15, 663–72.<br />

MORRIS, S.C., MADDEN, K.B., ADAMOVICZ, J.J., GAUSE, W.C., HUBBARD,<br />

B.R., GATELY, M.K. and FINKELMAN, F.D., 1994, Effects <strong>of</strong> IL-12 on in<br />

vivo cytokine gene expression and Ig isotype selection, Journal <strong>of</strong><br />

Immunology, 152, 1047–56.<br />

MOSMAN, T.R. and COFFMAN, R.L., 1989, Heterogeneity <strong>of</strong> cytokine secretion<br />

patterns and functions <strong>of</strong> helper T cells, Advances in Immunology, 46, 111–<br />

47.<br />

MOSMANN, T.R., SCHUMACHER, J.H., STREET, N.F., BUDD, R., O’GARRA,<br />

A., FONG, T.A.T., BOND, M.W., MOORE, K.W.M., SHER, A. and<br />

FIORENTINO, D.F., 1991, Diversity <strong>of</strong> cytokine synthesis and function <strong>of</strong><br />

mouse CD4 + T cells, Immunological Reviews, 123, 209–29.<br />

MURANKA, M., SUZUKI, S., KOIZUMI, K., TAKAFUJI, S., MIYAMOTO, T.,<br />

IKEMURI, R. and TOKIWA, H., 1986, Adjuvant activity <strong>of</strong> diesel-exhaust


I.KIMBER 147<br />

particulates for the production <strong>of</strong> IgE antibody in mice, Journal <strong>of</strong> Allergy and<br />

Clinical Immunology, 77, 616–23.<br />

MURDOCH, R.D., PEPYS, J. and HUGHES, E.G., 1986. IgE antibody responses<br />

to platinum group metals: a large scale refinery survey, British Journal <strong>of</strong><br />

<strong>Industrial</strong> Medicine, 43, 37–43.<br />

NEMERY, B. and LENAERTS, L., 1993, Exposure to methylene diphenyl<br />

diisocyanate in coal mines, Lancet, 341, 318.<br />

NEWMAN TAYLOR, A.J., 1988, Occupational asthma, Postgraduate Medical<br />

Journal, 64, 505–10.<br />

NOBLE, A., STAYNOV, D.Z., DIAZ-SANCHEZ, D., LEE, T.J. and KEMENY, D.<br />

M., 1993, Elimination <strong>of</strong> IgE regulatory rat CD8 + T cells in vivo increases the<br />

co-ordinate expression <strong>of</strong> Th 2 cytokines IL-4, IL-5 and IL-10, Immunology, 80,<br />

326–9.<br />

O’BRIEN, I.M., HARRIES, M.G., BURGE, P.S. and PEPYS, J., 1979, Toulene<br />

diisocyanate-induced asthma. 1. Reactions to TDI, MDI, HDI and histamine,<br />

Clinical Allergy, 9, 1–6.<br />

PARRONCHI, P., MACCHIA, D., PICCINI, M-P., BISWAS, P., SIMONELLI, C.,<br />

MAGGI, E., RICCI, M., ANSARI, A.A. and ROMAGNANI, S., 1991,<br />

Allergen and bacterial antigen-specific T cell clones established from atopic<br />

donors show a different pr<strong>of</strong>ile <strong>of</strong> cytokine production, Proceedings <strong>of</strong> the<br />

National Academy <strong>of</strong> Sciences USA, 88, 4538–42.<br />

PENE, J., ROUSSET, F., BRIERE, F., CHRETIEN, I., PALIARD, X.,<br />

BANCHEREAU, J., SPITS, H. and DE VRIES, J.E., 1988, IgE production by<br />

normal human B cells induced by alloreactive T cell clones is mediated by 11–4<br />

and suppressed by IFN- , Journal <strong>of</strong> Immunology, 141, 1218–24.<br />

POWRIE, F., MENON, S. and COFFMAN, R.L., 1993, Interleukin-4 and<br />

interleukin-10 synergize to inhibit cell-mediated immunity in vivo, European<br />

Journal <strong>of</strong> Immunology, 23, 3043–9.<br />

QUIRCE, S., CUEVAS, M., OLAGUIBEL, J.M. and TABAR, A.I., 1994,<br />

Occupa tional asthma and immunological responses induced by inhaled<br />

carmine among employees at a factory making natural dyes, Journal <strong>of</strong> Allergy<br />

and Clinical Immunology, 93, 44–52.<br />

RATTRAY, N.J., BOTHAM, P.A., HEXT, P.M., WOODCOCK, D.R.,<br />

FIELDING, I., DEARMAN, R.J. and KIMBER, I., 1994, Induction <strong>of</strong><br />

respiratory hypersensitivity to diphenylmethane-4,4′-diisocyanate (MDI) in<br />

guinea pigs. Influence <strong>of</strong> route <strong>of</strong> exposure, <strong>Toxicology</strong>, 88, 15–30.<br />

RENZ, H., LACK, G., SALOGA, J., SCHWINZER, R., BRADLEY, K., LOADER,<br />

J., KUPFER, A., LARSEN, G.L. and GELFAND, E.W., 1994, Inhibition <strong>of</strong> IgE<br />

production and normalization <strong>of</strong> airways responsiveness by sensitized CD8 T<br />

cells in a mouse model <strong>of</strong> allergen-induced sensitization, Journal <strong>of</strong><br />

Immunology, 152, 351–60.<br />

ROMAGNANI, S., 1991, Human TH1 and T H2 subsets: doubt no more,<br />

Immunology Today, 12, 256–7.<br />

SCHMITT, E., HOEHN, P., HUELS, C., GOEDERT, S., PALM, N., RUDE, E. and<br />

GERMANN, T., 1994, T helper type 1 development <strong>of</strong> naive CD4 + T cells<br />

requires the coordinate action <strong>of</strong> interleukin-12 and interferon- and is<br />

inhibited by transforming growth factor-β, European Journal <strong>of</strong> Immunology,<br />

24, 793–8.


148 MECHANISMS OF PULMONARY SENSITIZATION<br />

SMITH, C.A. and RENNICK, D.M., 1986, Characterization <strong>of</strong> a murine<br />

lymphokine distinct from interleukin 2 and interleukin 3 (IL-3) possessing a Tcell<br />

growth factor and mast cell growth factor activity that synergizes with<br />

IL-3, Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences USA, 83, 1857–61.<br />

THOMPSON-SNIPES, L., DHAR, V., BOND, M.W., MOSMANN, T.R.,<br />

MOORE, K.W. and RENNICK, D.M., 1991, Interleukin-10: a novel<br />

stimulatory factory for mast cells and their progenitors, Journal <strong>of</strong><br />

Experimental Medicine, 173, 507–10.<br />

TOPPING, M.D., VENABLES, K.M., LUCZYNSKA, C.M., HOWE, W. and<br />

NEWMAN TAYLOR, A.J., 1986, Specificity <strong>of</strong> the human IgE response to<br />

inhaled acid anhydrides, Journal <strong>of</strong> Allergy and Clinical Immunology, 77, 834–<br />

42.<br />

VANDENPLAS, O., CARTIER, A., LESAGE, J., CLOUTIER, Y., PERREAULT,<br />

G., GRAMMER, L.C., SHAUGHNESSY, M.A. and MALO, J-L., 1993, Prepolymers<br />

<strong>of</strong> hexamethylene diisocyanate as a cause <strong>of</strong> occupational asthma,<br />

Journal <strong>of</strong> Allergy and Clinical Immunology, 91, 850–61.<br />

WARDLAW, A.J., 1993, The role <strong>of</strong> air pollution in asthma, Clinical and<br />

Experimental Allergy, 23, 81–96.<br />

YING, S., DURHAM, S.R., JACOBSON, M.R., RAK, S., MASUYAMA, K.,<br />

LOWHAGEN, O., KAY, A.B. and HAMID, Q.A., 1994, T lymphocytes and<br />

mast cells express messenger RNA for interleukin-4 in the nasal mucosa in<br />

allergen-induced rhinitis, Immunology, 82, 200–6.<br />

YOKOTA, T., COFFMAN, R.L., HAGIWARA, H., RENNICK, D.M., TAKEBE,<br />

Y., YOKOTA, K., GEMMELL, L., SCHRADER, B., YANG, G., MEYERSON,<br />

P., LUH, H., HOY, P., PENE, J., BRIERE, F., SPITS, H., BANCHEREAU, J.,<br />

DE VRIES, J., LEE, F.D., ARAI, N. and ARAI, K.I., 1987, Isolation and<br />

characterization <strong>of</strong> lymphokine cDNA clones encoding mouse and human IgAenhancing<br />

and eosinophil colony-stimulating factor activities. Relationship to<br />

interleukin 5. Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences USA, 84,<br />

7388–92.<br />

ZEISS, C.R., KANELLAKES, T.M., BELLONE, J.D., LEVITZ, D., PRUZANSKY,<br />

J.J. and PATTERSON, R., 1980, Immunoglobulin E-mediated asthma and<br />

hypersensitivity pneumonitis with precipitating anti-hapten antibodies due<br />

to diphenylmethane diisocyanate (MDI) exposure, Journal <strong>of</strong> Allergy and<br />

Clinical Immunology, 65, 346–52.<br />

ZETTERSTROM, O., OSTERMAN, K., MACHADO, L. and JOHANSSON, S.G.,<br />

1981, Another smoking hazard: raised serum IgE concentration and increased<br />

risk <strong>of</strong> occupational allergy, British Medical Journal, 283, 1215–17.


11<br />

Occupational Asthma Induced by Chemical<br />

Agents<br />

C.A.C.PICKERING<br />

Wythenshawe Hospital Manchester<br />

Introduction<br />

Occupational asthma may be defined as variable airways narrowing<br />

causally related to exposure in the working environment to airborne dust,<br />

gases, vapours or fumes (Newman Taylor, 1980). This definition includes,<br />

therefore, both immunological and nonimmunological causes <strong>of</strong> asthma in<br />

the workplace. Immunological causes <strong>of</strong> asthma in general demonstrate a<br />

latent period between exposure and the development <strong>of</strong> symptoms. Once<br />

sensitisation has occurred airway responses may be seen at very low levels<br />

<strong>of</strong> exposure. Both high and low molecular weight agents may cause<br />

sensitisation. Irritantinduced occupational asthma characteristically follows<br />

within 24 h <strong>of</strong> a usually, single, high level exposure to an irritant substance<br />

and has been named reactive airways dysfunction syndrome (Brooks et al.,<br />

1985).<br />

The number <strong>of</strong> chemical agents causing occupational asthma is<br />

extensive. As new, highly reactive, chemicals are developed these numbers<br />

are likely to grow. Low molecular weight chemicals may act as haptens,<br />

reacting with body protein to form a complete antigen to which specific<br />

antibodies are formed.<br />

The incidence <strong>of</strong> occupational asthma in most countries is not known<br />

with any great accuracy, there are considerable variations in reporting<br />

systems between countries. Since many individuals with occupational<br />

asthma change jobs without a specific diagnosis being established, the<br />

published figures <strong>of</strong> incidence will be significant underestimates <strong>of</strong> the true<br />

incidences. In Japan (Kobayashi et al., 1973), the prevalence <strong>of</strong><br />

occupational asthma amongst adult male asthmatics is said to be about<br />

15%. In the UK a new reporting system has recently been established—<br />

Surveillance <strong>of</strong> Work-related and Occupational Respiratory Disease Project<br />

(SWORD). Newly diagnosed cases <strong>of</strong> workrelated respiratory disease are<br />

reported monthly by consultant chest and occupational physicians.<br />

Between 1989 and 1991, 631 cases <strong>of</strong> chemically induced occupational<br />

asthma were reported, <strong>of</strong> these 53% were associated with exposure to


150 OCCUPATIONAL ASTHMA INDUCED BY CHEMICAL AGENTS<br />

isocyanates. A similar system (SHIELD) has been established in the UK in<br />

the West Midlands (Gannon et al., 1993). They reported an incidence <strong>of</strong> 43<br />

new cases per million workers per year. Specific occupational incidences<br />

varied from 1833 per million paint sprayers to 8 per million clerks. Again<br />

more than half the cases <strong>of</strong> asthma were attributed to isocyanates.<br />

The initial diagnosis <strong>of</strong> occupational asthma is based on a workers<br />

history <strong>of</strong> respiratory symptoms improving on days away from work and<br />

when on holiday. At the onset <strong>of</strong> occupational asthma this pattern is<br />

usually present but continued exposure to the allergen leads to increasing<br />

airway reactivity. Their symptoms may then persist over weekends and be<br />

triggered by nonspecific factors outside the workplace such as exhaust<br />

fumes, aerosol sprays and perfumes. The difficulties <strong>of</strong> relying on the<br />

history alone in the diagnosis <strong>of</strong> occupational asthma has been well<br />

documented (Malo et al., 1991). A series <strong>of</strong> 162 hospital referrals to two<br />

expert physicians were initially categorised, on the basis <strong>of</strong> their histories,<br />

into highly probable, probable, uncertain, unlikely or absent occupational<br />

asthma. The diagnosis was then established by bronchial provocation<br />

testing and or serial measurements <strong>of</strong> lung function. The predictive value <strong>of</strong><br />

a physician’s assessment <strong>of</strong> occupational asthma being highly probable or<br />

probable was only 63%. This improved to 83% in the groups in whom<br />

occupational asthma was assessed as being unlikely or absent.<br />

The early identification <strong>of</strong> work-related symptoms and their subsequent<br />

investigation in the workplace is important. Only rarely, when very acute<br />

episodes <strong>of</strong> workplace asthma are described, should lung function<br />

measurements at work be avoided. While pre- and post-shift measurements<br />

<strong>of</strong> lung function may identify a work-related effect, late asthmatic<br />

responses occurring in the evening after leaving work are frequently seen in<br />

chemically induced forms <strong>of</strong> occupational asthma. Serial measurements <strong>of</strong><br />

lung function made every 2 h from waking to sleeping, both on working<br />

days and on days away from work, using a peak flow meter, will identify<br />

these late responders. The sensitivity <strong>of</strong> this type <strong>of</strong> investigation in<br />

establishing a diagnosis <strong>of</strong> occupational asthma is about 80% (Burge, 1982),<br />

this falls to 46% once the worker is started on specific treatment for his<br />

asthma, again emphasising the importance <strong>of</strong> early identification and<br />

investigation <strong>of</strong> work-related respiratory symptoms.<br />

Currently more than 140 low molecular weight chemicals have been<br />

reported to induce occupational asthma (Butcher and Salvaggio, 1986).<br />

The majority <strong>of</strong> these chemicals induce asthma by mechanisms which have<br />

yet to be identified. In a minority <strong>of</strong> instances specific IgE antibodies to the<br />

implicated chemical have been identified.<br />

Bronchial challenge tests with chemicals which are non-IgE dependent<br />

usually induce either an isolated late asthmatic response or a biphasic or<br />

dual asthmatic response. The IgE dependent responses induce immediate or<br />

dual asthmatic responses.


The most common chemical causes <strong>of</strong> occupational asthma include the<br />

iso- cyanates and the acid anhydrides. This chapter will examine these two<br />

groups in more detail.<br />

Isocyanates<br />

C.A.C.PICKERING 151<br />

The polyisocyanates and their oligomers are the most important cause <strong>of</strong><br />

chemically induced asthma. These organic compounds are synthesised by<br />

the reaction <strong>of</strong> amines with phosgene. There are a number <strong>of</strong> related<br />

compounds the most important <strong>of</strong> which are 2,4- and 2,6-toluene<br />

diisocyanate (TDI), methylene diphenyldiisocyanate (MDI), hexamethylene<br />

diisocyante (HDI), napthalene diisocyanate (NDI), isophorone diisocyanate<br />

(IPDI), and polyisocyanates derived from HDI and MDI.<br />

The incidence <strong>of</strong> occupational asthma due to diisocyanates varies widely.<br />

It is influenced by the type <strong>of</strong> compound and its vapour pressure. TDI and<br />

HDI are highly volatile at room temperature, whereas MDI has to be<br />

heated to above 60°C to volatilise. It is thought that approximately 5% <strong>of</strong><br />

an exposed working population will develop occupational asthma after<br />

exposure to TDI (Diem et al., 1982). Because <strong>of</strong> the known respiratory<br />

problems associated with exposure to isocyanates with high vapour<br />

pressure properties, new isocyanate compounds with low vapour pressure<br />

properties have been developed particularly for use in the paint spraying<br />

industry. Recent studies however continue to demonstrate significant levels<br />

<strong>of</strong> occupational asthma despite the use <strong>of</strong> recommended respiratory<br />

protection (Seguin et al., 1987, Welinder et al., 1988). Bronchial<br />

provocation studies with HDI- and MDI-derived polyisocyanates have<br />

confirmed their ability to cause occupational asthma. Airborne iso-cyanate<br />

prepolymers appear to be able to induce asthma to the same or greater<br />

frequency as isocyanate monomers.<br />

High exposures to isocyanate vapours, such as occur in a major<br />

industrial spillage, cause acute rhinitis, lacrymation, cough and wheezing<br />

leading to subsequent sensitisation. In some individuals this type <strong>of</strong><br />

exposure induces persistent asthma—reactive airways dysfunction<br />

syndrome (RADS). Respiratory sensitisation may occur at very low levels<br />

<strong>of</strong> exposure. Pepys et al. (1972) described a boat builder who became<br />

sensitised to TDI at exposure levels <strong>of</strong> between 0.00173 and 0.0018 ppm.<br />

Similarly White et al. (1980) reported respiratory symptoms and the<br />

development <strong>of</strong> IgE antibodies to TDI, in machinists manufacturing carseat<br />

covers exposed to levels <strong>of</strong> TDI <strong>of</strong> between 0.0003 and 0.003 ppm. It is<br />

more usual, in the author’s experience, for the sensitised individual to<br />

provide a history <strong>of</strong> short lived peak exposures to isocyanates which have<br />

clearly been above the current threshold limit value. These intermittent<br />

relatively high level exposures may be important in the sensitisation


152 OCCUPATIONAL ASTHMA INDUCED BY CHEMICAL AGENTS<br />

process. Once sensitised, a worker may have his symptoms initiated by very<br />

low exposure levels <strong>of</strong> isocyanates.<br />

Diisocyanate asthma is usually but not always associated with the<br />

presence <strong>of</strong> nonspecific bronchial hyperreactivity. The majority <strong>of</strong> workers<br />

who develop occupational asthma remain symptomatic requiring regular<br />

treatment permanently after cessation <strong>of</strong> exposure (Allard et al., 1989).<br />

The duration <strong>of</strong> exposure with symptoms before diagnosis has a major<br />

influence on recovery patterns. In a group <strong>of</strong> 43 isocyanate workers with<br />

occupational asthma, those who had fully recovered were exposed with<br />

symptoms for 1.6 years, those who had improved, 2.8 years and those who<br />

had not improved, 5.4 years (Pisati et al., 1993). The resolution or<br />

improvement in occupational asthma takes place over a 2 year period after<br />

cessation <strong>of</strong> exposure, symptoms still present at 2 years should be regarded<br />

as permanent. Most epidemiological studies have not identified any specific<br />

risk factors including atopic status, smoking or nonspecific bronchial<br />

hyperreactivity.<br />

The laboratory identification <strong>of</strong> specific antibodies to diisocyanates has<br />

proved <strong>of</strong> very limited value. Diisocyanate specific IgE is demonstrable in<br />

only 10–20% <strong>of</strong> sensitised individuals and have also been identified in<br />

individuals with no history <strong>of</strong> asthma (Butcher et al., 1983). Similarly<br />

specific IgG antibodies to diisocyanates have been described in workers<br />

both with and without evidence <strong>of</strong> disease.<br />

At the present time the recommended long-term exposure limit (8 h<br />

TWA reference period) for diisocyanates is 0.02 mg m −3 and the short-term<br />

exposure limit (10 min reference period) is 0.07 mg m −3 in the UK. There is<br />

discussion at the present time as to whether levels should be lower in order<br />

to prevent the development <strong>of</strong> diisocyanate asthma. However since most<br />

workers with diisocyanate airways disease describe exposures in excess <strong>of</strong><br />

the current recommended exposure levels the prevalence <strong>of</strong> occupational<br />

asthma in a workforce without such exposures is not known. There need to<br />

be improvements in hygiene control to prevent these peak exposures to<br />

isocyanates.<br />

Acid anhydrides<br />

The acid anhydrides are a group <strong>of</strong> low molecular weight chemicals used as<br />

curing agents in the production <strong>of</strong> epoxy and alkyd resins and in the<br />

production <strong>of</strong> plasticisers such as dioctyl phthalate. Acid anhydrides exert<br />

diverse effects on man both as sensitisers, irritants or both. The most<br />

frequently used anhydrides, all <strong>of</strong> which have been described causing<br />

occupational asthma, are phthalic anhydride (PA), trimellitic anhydride<br />

(TMA), tetrachlorophthalic anhydride (TCPA) and maleic anhydride<br />

(MA). In addition himic anhydride and pyromellitic dianhydride (PMDA)<br />

have been described as causing asthma.


The direct toxicity <strong>of</strong> anhydrides involves irritation <strong>of</strong> the mucus<br />

membranes and skin which may result in eye lesions, epistaxis, pulmonary<br />

congestion, haemoptysis and skin burns (Venables, 1989).<br />

Occupational asthma is most frequently reported due to PA, less<br />

commonly to TMA, TCPA and MA and finally there are single case reports<br />

<strong>of</strong> asthma due to HA, HHPA and PMDA (Venables, 1989). A second type<br />

<strong>of</strong> response to acid anhydrides has also been described and is termed the<br />

‘late respiratory systemic syndrome’ (LRRS). This is characterised by the<br />

development <strong>of</strong> influenzal type symptoms, fever, generalised acheing and<br />

malaise, late in the working shift or in the evening after work. These<br />

symptoms may occur in isolation or in association with asthma. It is not<br />

clear whether this response is immunologically mediated or a nonspecific<br />

response to high levels <strong>of</strong> anhydride exposure. Lastly, exposure to TMA,<br />

probably at high exposure levels, has been described as causing severe<br />

pulmonary haemorrhage requiring both blood transfusion and mechanical<br />

ventilation (Rivera et al., 1989).<br />

Serum IgE and IgG antibodies to acid anhydrides have been identified.<br />

IgE antibodies appear to be more specifically associated with occupational<br />

asthma. Howe et al. (1983) reported seven cases <strong>of</strong> TCPA asthma all <strong>of</strong><br />

whom had IgE antibody to TCPA, compared with 8% <strong>of</strong> 300 exposed<br />

workers without TCPA asthma; 29% <strong>of</strong> this exposed nonasthmatic<br />

population had IgG antibodies to TCPA.<br />

The exposure levels <strong>of</strong> acid anhydrides that initiate sensitisation are<br />

poorly understood. TMA at levels <strong>of</strong> 1.7–4.7 mg m −3 (Zeiss et al., 1977)<br />

and 0.007–2.1 mg m −3 (Bernstein et al., 1983; McGrath et al., 1984) have<br />

been described causing occupational asthma. PA at 0.03–15 mg m −3 has<br />

also been reported as causing asthma (Wernfors et al., 1986). As in other<br />

forms <strong>of</strong> occupational asthma, the early identification <strong>of</strong> cases <strong>of</strong> acid<br />

anhydride induced asthma and their removal from exposure is <strong>of</strong> prime<br />

importance.<br />

Reactive airways dysfunction syndrome<br />

C.A.C.PICKERING 153<br />

Reactive airways dysfunction syndrome (RADS) or irritant-induced asthma<br />

was first described in 1985 (Brooks et al., 1985). The criteria used in<br />

diagnosis include a high level exposure to an irritant fume, vapour or smoke,<br />

the development <strong>of</strong> respiratory symptoms within minutes or hours <strong>of</strong><br />

exposure, in an individual with no previous history <strong>of</strong> respiratory symptoms,<br />

with persistence <strong>of</strong> symptoms and physiological abnormalities for more<br />

than 1 year. A variety <strong>of</strong> different chemical exposures have been described<br />

inducing this syndrome including: chlorine (Moore and Sherman, 1991),<br />

glacial acetic acid (Kern, 1991), hydrochloric acid (Promisl<strong>of</strong>f et al., 1990)<br />

and miscellaneous chemical exposures (Brooks et al., 1985). A comparison<br />

between cases <strong>of</strong> occupational asthma and RADS (Gautrin et al., 1994)


154 OCCUPATIONAL ASTHMA INDUCED BY CHEMICAL AGENTS<br />

suggest that cases with RADS are left with less airway reversibility than<br />

occupational asthmatics. This would be consistent with the pathological<br />

findings (Boutet et al., 1993) in RADS, with more severe basement<br />

membrane thickening and bronchial wall fibrosis than is present in<br />

occupational asthma.<br />

The development <strong>of</strong> occupational asthma in any individual has<br />

potentially serious consequences both in terms <strong>of</strong> persisting disability,<br />

possible unemployment and loss <strong>of</strong> income (Gannon et al., 1993). It is<br />

incumbent on management to ensure safe working conditions with<br />

adequate control and regular monitoring <strong>of</strong> atmospheric levels <strong>of</strong> chemical<br />

agents.<br />

References<br />

ALLARD, C., CARTIER, A., GHEZZO, H. and MALO, J-L., 1989, Occupational<br />

asthma due to various agents. Absence <strong>of</strong> clinical and functional improvement<br />

at an interval <strong>of</strong> four or more years after cessation <strong>of</strong> exposure, Chest, 96,<br />

1046–9.<br />

BERNSTEIN, D.I., ROACH, D.E., MCGRATH, K.G., LARSEN, R.S., ZEISS, C. R.<br />

and PATTERSON, R., 1983, The relationship <strong>of</strong> airborne trimellitic<br />

anhydrideinduced symptoms and immune responses, J. Allergy Clin.<br />

Immunol., 72, 709–13.<br />

BOUTET, M., BOULET, L.-P., MALO, J.L., CARTIER, A., CÔTÉ, J., LEBLANC,<br />

C., MILOT, J. and LAVIOLETTE, M., 1993, Morphological evidence <strong>of</strong><br />

modified contractile properties <strong>of</strong> airways in occupational asthma and reactive<br />

airways dys-function syndrome, Am. Rev. Respir. Dis., 147, A113.<br />

BROOKS, S.M., WEISS, M.A. and BERNSTEIN, I.L., 1985, Reactive airways<br />

dysfunction syndrome. Case reports <strong>of</strong> persistent airways hyperreactivity<br />

following high-level irritant exposures, J. Occup. Med., 27, 473–6.<br />

BURGE, P.S., 1982, Single and serial measurements <strong>of</strong> lung function in the<br />

diagnosis <strong>of</strong> occupational asthma, Eur. J. Resp. Dis., 63 (suppl. 123), 47–9.<br />

BUTCHER, B.T. and SALVAGGIO, J.E., 1986, Continuing medical education—<br />

occupational asthma, J. Allergy Clin. Immunol. 78, 547–9.<br />

BUTCHER, B.T., O’NEIL, C.E., REED, M.A. and SALVAGGIO, J.E., 1983, Radioallergosorbent<br />

testing with p-tolyl monoisocyanate in toluene diisocyanate<br />

workers, Clin. Allergy., 13, 31–4.<br />

DIEM, J.E., JONES, R.N., HENDRICK, D.J., GLINDMEYER, H.W.,<br />

DHARMARAJAN, V., BUTCHER, B.T., SALVAGGIO.J.E., and WEILL, H.,<br />

1982, Five year longitudinal study <strong>of</strong> workers employed in a new toluene<br />

diisocyanate manufacturing plant, Am. Rev. Respir. Dis., 126, 420–8.<br />

GANNON, P.F.G. and BURGE, P.S., 1993, The SHIELD scheme in the West<br />

Midlands region, United Kingdom, Brit. J. Ind. Med., 50, 791–6.<br />

GANNON, P.F.G., WEIR, D.C., ROBERTSON, A.S. and BURGE, P.S., 1993,<br />

Health, employment, and flnancial outcomes in workers with occupational<br />

asthma, Brit. J. Ind. Med., 50, 491–6.


C.A.C.PICKERING 155<br />

GAUTRIN, D., BOULET, L.-P., BOUTET, M., DUGAS, M., BHÉRER, L.,<br />

L’ARCHEVÊQUE, J., LAVIOLETTE, M., CÔTÉ, J. and MALO, J.-L., 1994,<br />

Is reactive airways dysfunction syndrome a variant <strong>of</strong> occupational asthma? J.<br />

Allergy Clin. Immunol, 93, 12–22.<br />

HOWE, W., VENABLES, K.M., TOPPING, M.D., DALLY M.B., HAWKINS, R.,<br />

LAW, J.S. and NEWMAN TAYLOR, A.J., 1983, Tetrachlorophthalic<br />

anyhdride asthma: evidence for specific IgE antibody, J. Allergy Clin.<br />

Immunol., 71, 5–11.<br />

KERN, D.G., 1991. Outbreak <strong>of</strong> the reactive airways dysfunction syndrome after a<br />

spill <strong>of</strong> glacial acetic acid, Am. Rev. Respir. Dis. 144, 1058–64.<br />

KOBAYASHI, S.Y., YAMAMORA, Y., FRICK, O.L., HORIUCHI, S.<br />

KISHIMOTO, T. and MIYAMOTO, T., 1973, Occupational asthma due to the<br />

inhalation <strong>of</strong> pharmaceutical dusts and other chemical agents with some<br />

reference to other occupational asthma in Japan, Proc. VIII Int. Congr.<br />

Allergology, Tokyo, October 1973, pp. 124–32. Amsterdam: Excerpta<br />

Medica.<br />

MALO, J.L., GHEZZO, H., L’ARCHEVÊQUE, LAGIER, F. and CARTIER, A.,<br />

1991, Is the clinical history a satisfactory means for diagnosing occupational<br />

asthma? Am. Rev. Respir. Dis., 143 528–32.<br />

MCGRATH, K.G., ROACH, D., ZEISS, C.R. and PATTERSON, R., 1984,<br />

Four year evaluation <strong>of</strong> workers exposed to trimellitic anhydride: a brief<br />

report, J. Occup. Med., 26, 671–5.<br />

MOORE, B.B. and SHERMAN, M., 1991, Chronic reactive airway disease<br />

following acute chlorine gas exposure in an asymptomatic atopic patient,<br />

Chest, 100, 855–6.<br />

NEWMAN TAYLOR, A.J., 1980, Occupational asthma, Thorax, 35, 241–5.<br />

PEPYS, J., PICKERING, C.A .C., BRESLIN, A.B.X. and TERRY D.J., 1972,<br />

Asthma due to inhaled chemical agents—tolylene diisocyanate, Clin. Allergy,<br />

2. 225–36.<br />

PISATI, G., BARUFFINI, A. and ZEDDA, S., 1993, Toluene diisocyanate induced<br />

asthma: outcome according to persistence or cessation <strong>of</strong> exposure, Brit. J.<br />

Ind. Med., 50, 60–4.<br />

PROMISLOFF, R.A., PHAN, A., LENCHNER, G.S. and CICHELLI, A.V., 1990,<br />

Reactive airway dysfunction syndrome in three police <strong>of</strong>ficers following a<br />

roadside chemical spill, Chest, 98, 928–9.<br />

RIVERA, M., NICOTRA, M.B., BYRON, G.E., PATTERSON, R., YAWN, D.H.,<br />

FRANCO, M., ZEISS, C.R. and GREENBERG, S.D., 1981, Trimellitic<br />

anhydride toxicity: a cause <strong>of</strong> acute multisystem failure, Arch. Intern. Med.,<br />

141, 1071– 4.<br />

SEGUIN, P., ALLARD, A., CARTIER, A. and MALO, J.-L., 1987, Prevalence <strong>of</strong><br />

occupational asthma in spray painters exposed to several types <strong>of</strong> isocyanates,<br />

including polymethylene polyphenylisocyanate, J. Occup. Med., 29, 340–4.<br />

VENABLES, K.M., 1989, Low molecular weight chemicals, hypersensitivity, and<br />

direct toxicity: the acid anhydrides, Brit. J. Ind. Med., 46, 222–32.<br />

WELINDER, H., NIELSEN, J., BENSRYD, I. and SKERFVING, S., 1988, IgG<br />

antibodies against polyisocyanates in car painters, Clin. Allergy, 18, 85–93.


156 OCCUPATIONAL ASTHMA INDUCED BY CHEMICAL AGENTS<br />

WERNFORS, M., NIELSEN, J. and SKERFVING, S., 1986, Phthalic<br />

anhydrideinduced occupational asthma, Int. Arch. Allergy Appl. Immunol. 79,<br />

77–82.<br />

WHITE, W.G., SUGDEN, E., MORRIS, M.J. and ZAPATA, E, 1980, Isocyanateinduced<br />

asthma in a car factory, Lancet, 1, 756–60.<br />

ZEISS, C.R., PATTERSON, R., PRUZANSKY, J.J., MILLER, M.M.,<br />

ROSENBERG, M. and LEVITZ, D., 1977, Trimellitic anhydride-induced<br />

airway syndromes: clinical and immunological studies, J. Allergy Clin.<br />

Immunol., 60, 96–103.


PART FOUR<br />

Biomarkers and risk assessment <strong>of</strong><br />

industrial chemicals


12<br />

Biomarkers and Risk Assessment<br />

KARI HEMMINKI<br />

Karolinska Institute, Huddinge<br />

Introduction<br />

Many chemical carcinogens cause covalent DNA-binding products,<br />

adducts, which may induce mutations or other types <strong>of</strong> DNA damage in<br />

important growth-controlling genes or loci resulting in aberrant cellular<br />

growth and cancer (Harris, 1991; IARC 1992; Hemminki, 1993). Human<br />

exposure to compounds such as polycyclic aromatic hydrocarbons (PAH)<br />

can be determined, for example, by ambient air, biological or DNA adduct<br />

monitoring. The usefulness <strong>of</strong> a method for the determination <strong>of</strong> DNA<br />

adducts in human biomonitoring requires high sensitivity because the levels<br />

<strong>of</strong> adducts are low. Here the primary focus is on the assessment <strong>of</strong><br />

exposure using the above indicators in industries where high exposure to<br />

PAHs occur, such as iron founding, coke production, aluminium<br />

production, garage work and engine overhauling with exposure to used<br />

lubricating oils.<br />

Biomonitoring <strong>of</strong> PAH exposure<br />

Literature on the application <strong>of</strong> DNA adduct studies in humans is extensive<br />

(Beach and Gupta, 1992; IARC, 1993, 1994; Hemminki et al., 1993a;<br />

Hemminki, 1994). A large majority <strong>of</strong> the 32 P-postlabelling studies on<br />

human samples focus on tobacco smoking, occupational exposures and<br />

cancer chemotherapy patients. Most occupational exposures studied relate<br />

to complex mixtures, including polycyclic aromatic hydrocarbons (PAHs).<br />

In exposure to complex mixtures multiple radioactive spots (called diagonal<br />

radioactive zones, DRZ) are detected. The adduct spots cannot be<br />

definitively identified nor quantitated. As it has turned out that for many<br />

adducts labelling is not completed, even among structural analogues such<br />

as PAHs, the adduct levels measured are likely to be underestimates<br />

(Segerbäck and Vodicka, 1993).


Table 12.1 Exposure and aromatic adducts in occupational populations, presented in simplified tabulated form<br />

K.HEMMINKI 159<br />

Notes:<br />

a Total white blood cells.<br />

b Lymphocytes.<br />

c Not given.<br />

d No data.<br />

The types <strong>of</strong> occupational groups studied by postlabelling include<br />

foundry, coke oven and aluminium workers, ro<strong>of</strong>ers, garage and terminal<br />

workers, car mechanics and chimney sweeps. All these groups have had an


160 BIOMARKERS AND RISK ASSESSMENT<br />

increased risk <strong>of</strong> lung cancer. As, however, epidemiological studies relate to<br />

exposure a few decades earlier the risks <strong>of</strong> present exposures can only be<br />

predicted. The levels <strong>of</strong> aromatic adducts are elevated in white blood cells<br />

or lymphocytes in many <strong>of</strong> these groups. The reported total aromatic<br />

adduct levels usually range between 1 and 10 adducts per 10 8 nucleotides.<br />

Long-lived lymphocytes tend to have higher levels <strong>of</strong> adducts than shortlived<br />

granulocytes (Savela and Hemminki, 1991; Grzybowska et al., 1993).<br />

As a rule <strong>of</strong> thumb, it can be assumed that in a steady-state (i.e. long term<br />

exposure) lymphocytes contribute to the level <strong>of</strong> adducts overwhelmingly.<br />

Because they represent about 25 percent <strong>of</strong> the DNA in blood, the<br />

relationship between total white blood cell (WBC) and lymphocyte DNA<br />

adducts should be about 1:4, granulocytes only contributing to the amount<br />

<strong>of</strong> DNA denominator. Yet one has to be cautious in the comparison <strong>of</strong><br />

results between various assays even within a laboratory as the results may<br />

‘drift’ with time.<br />

The levels <strong>of</strong> white blood cell/lymphocyte aromatic adducts from<br />

workers in several industries, as measured by postlabelling, and as<br />

compared to ambient air concentrations <strong>of</strong> benzo (a) pyrene (BP) and 1hydroxypyrene<br />

levels are presented in Table 12.1. A boxplot presentation<br />

<strong>of</strong> the adduct levels <strong>of</strong> bus maintenance and truck terminal workers is<br />

shown in Figure 12.1 (Hemminki et al., 1994). The differences that were<br />

statistically significant from the controls were, in addition to the groups <strong>of</strong><br />

maintenance and terminal workers, garage workers and diesel forklift<br />

drivers.<br />

There does not seem to be a direct relationship between exposure and<br />

adduct levels. Electrode, coke and aluminium workers, exposed up to<br />

several 100 ng m −3 concentrations <strong>of</strong> BP, do not differ from the control<br />

more than foundry workers, exposed to less than 1/10 <strong>of</strong> the cited levels.<br />

The apparently higher level <strong>of</strong> adducts in the aluminium and electrode<br />

former workers (and controls) as compared to the other measurements, is<br />

due a method applied earlier with higher amounts <strong>of</strong> radioactive ATP. The<br />

later assays were carried out in small volumes but high concentrations <strong>of</strong><br />

ATP (Hemminki et al., 1993b; Szyfter et al., 1994). An increased level <strong>of</strong><br />

lymphocyte adducts has also been found in garage and truck terminal<br />

workers, with estimated exposures <strong>of</strong> about 10 ng m −3 (Hemminki et al.,<br />

1994). This would imply that the detection limit <strong>of</strong> the postlabelling<br />

method in humans exposed to PAHs lies somewhere between 1 and 10 ng<br />

m −3 BP. Whether diesel exhaust is a particularly potent inducer <strong>of</strong> adducts<br />

remains to be demonstrated. The differences between the exposed and the<br />

controls are statistically significant among foundry workers, all bus<br />

maintenance personnel and garage workers as a subgroup, all truck<br />

terminal workers and the diesel forklift drivers in particular. Coke<br />

workers differed significantly from the local controls in summer when


environmental pollution was low and the adduct levels in the controls were<br />

about 1/10 <strong>of</strong> their level in the winter (Grzybowska et al., 1993).<br />

Adducts and other endpoints<br />

K.HEMMINKI 161<br />

Figure 12.1 A boxplot <strong>of</strong> the white blood cell DNA adduct levels (per 10 8<br />

nucleotides) among bus maintenance and truck terminal workers and controls<br />

(Hemminki et al., 1994).<br />

It has become customary to include many types <strong>of</strong> endpoints to<br />

biomonitoring studies. The foundry study cited in Table 12.1 belongs to<br />

the most versatile <strong>of</strong> them. Exposure is measured by ambient air and 1hydroxypyrene<br />

monitoring (Santella et al., 1993). DNA adducts are<br />

assayed for by postlabelling and immunoassay. Plasma albumin PAH<br />

adducts are measured. Hypoxanthin guanine phosphoribosyl transferase<br />

(HPRT) and glycophorin A mutations are assayed for in lymphocytes and<br />

erythrocytes, respectively (Perera et al., 1993, 1994). Single-stand breaks in<br />

DNA and three types <strong>of</strong> cytogenetic parameters, chromosomal aberrations,<br />

sister chromatid exchanges and micronuclei, are analysed, in addition to<br />

genotyping <strong>of</strong> drug metabolising enzyme genes. Sampling <strong>of</strong> workers was<br />

repeated in four consecutive years, each at the same time <strong>of</strong> the year. As the<br />

last sampling was in the end <strong>of</strong> 1993, it will take some time before the<br />

complete data set will be available for analysis.<br />

In some published work from this data set an increase in DNA adducts<br />

and mutation frequency in the HPRT and glycophorin A genes was<br />

reported (Figure 12.2). Yet unreported results appear to show an increase


162 BIOMARKERS AND RISK ASSESSMENT<br />

Figure 12.2 Total white blood cell DNA adducts, determined by immunoassay,<br />

HPRT and glycophorin A mutations in foundry workers, exposed to various levels<br />

<strong>of</strong> BP (Perera et al., 1993).<br />

in singlestrand breaks while none <strong>of</strong> the cytogenetic parameters are<br />

elevated in the foundry workers.<br />

Adducts and metabolic genotypes<br />

The modulation <strong>of</strong> environmental carcinogenesis by host polymorphism in<br />

genes for xenobiotics metabolising enzymes is currently under extensive<br />

investigation. It was initially sparked by findings linking certain<br />

phenotypes <strong>of</strong> drug metabolism to cancer risk (Seidegård et al., 1986;<br />

Nebert, 1991). The enzymes <strong>of</strong> interest in the context <strong>of</strong> exposure to PAHs<br />

include cytochrome P450 CYP1A1 and glutathione transferase GST,<br />

involved in the activation and inactivation, respectively, <strong>of</strong> PAHs. By<br />

restriction enzyme mapping two allelic forms, ml and m2, and two other<br />

closely linked codons for isoleucine (Ile, linked to ml) and valine (Val,<br />

linked to m2) can be defined, where m2 and valine represent the rare<br />

mutant genotypes, associated with the inducibility <strong>of</strong> the enzyme activity<br />

(Hayashi et al., 1991). Polymorphism in GST1 involves the presence or the<br />

absence <strong>of</strong> the gene (Nakachi et al., 1992). The null genotype lacks the<br />

enzyme completely.<br />

Among chimney sweeps there was an association <strong>of</strong> the rare, inducible<br />

CYP1A1 genotype ml/m2 with low adduct levels in white blood cell DNA<br />

(Ichiba et al., 1994). In the same study an increased level <strong>of</strong> adducts was<br />

noted in the GST1-individuals. The level <strong>of</strong> DNA adducts appeared to be<br />

related to both the GST and CYP1A1 genotype (Figure 12.3). Analysis <strong>of</strong><br />

micronuclei in chimney sweeps resulted in no differences between<br />

individuals <strong>of</strong> either CYP1A1 ml/m2, m2/m2 or Ile/Val genotypes nor <strong>of</strong><br />

GST1 + or − genotypes (Carstensen et al., 1993). There was however a


correlation between white blood cell DNA adducts and micronuclei and it<br />

was stronger among the GST-individuals (Ichiba et al., 1994).<br />

How important is the role <strong>of</strong> metabolic phenotype or genotype as a<br />

predictor <strong>of</strong> cancer risk remains to be established. However it would seem<br />

prudent to assume some role as long as there is significant exposure to a<br />

carcinogen, metabolism <strong>of</strong> which is regulated by polymorphic genes. It<br />

would be important to note that the question can only be addressed if both<br />

<strong>of</strong> these conditions are met. In much <strong>of</strong> the published literature there are<br />

uncertainties regarding the active agents and their metabolic routes in the<br />

tissues studied. Adjustment for a metabolic phenotype or genotype, when<br />

justified, may increase the precision in the measurement.<br />

Risk assessment<br />

K.HEMMINKI 163<br />

Figure 12.3 Total white blood cell DNA adducts, measured by postlabelling,<br />

according to CYP1A1 and GST1 genotype (Ichiba et al., 1994). Controls,<br />

Sweeps.<br />

Monitoring <strong>of</strong> DNA adducts in occupational setting has mainly been<br />

applied to workers exposed to PAHs. In the case <strong>of</strong> 32 P-postlabelling<br />

increases in the level <strong>of</strong> adducts has been noted at exposures around 10 ng<br />

BP m −3 or slightly below. This is close to the detection limit that can<br />

conveniently be attained with personal monitoring or by measuring urinary<br />

1-hydroxypyrene. As the adduct measurements also reflect some aspects <strong>of</strong><br />

metabolism and DNA repair, they extend the scope <strong>of</strong> exposure


164 BIOMARKERS AND RISK ASSESSMENT<br />

measurements to host factors that may underly individual susceptibility to<br />

cancer.<br />

It has become increasingly common to try and incorporate other<br />

endpoints to DNA adduct studies. These include metabolic parameters,<br />

discussed above, protein adducts, cytogenetic parameters and point<br />

mutations. Examples include ethylene oxide exposed workers (Tates et al.,<br />

1991) and foundry workers (Perera et al., 1993, 1994; Santella et al.,<br />

1993). In both studies several parameters were elevated. The study on<br />

chimney sweeps illustrated how the intermediary endpoint may increase<br />

precision in the measurements (cf. Figure 12.3). The initial study showed<br />

no correlation between sweeping and micronuclei even though an<br />

adjustment was made for CYP1A1 and GST genotypes (Carstensen et al.,<br />

1993). There was a moderate correlation between sweeping and white<br />

blood cell DNA adducts, and adducts and micronuclei. Both <strong>of</strong> these<br />

correlations were strengthened once GST genotype was considered (Ichiba<br />

et al., 1994).<br />

Increasing circumstantial evidence associates DNA adducts within<br />

groups to an increased risk <strong>of</strong> cancer (IARC, 1992; Hemminki, 1993).<br />

Many <strong>of</strong> the adduct studies have been carried out in occupational groups<br />

which have been at a risk <strong>of</strong> cancer based on epidemiological results. These<br />

studies may be old and relate to exposures decades ago. Even new<br />

epidemiological publications on cancer cannot accurately address<br />

exposures after about 1970. Simultaneously there have been large changes<br />

in technology and industrial hygiene, undermining the quantitative and<br />

sometimes even the qualitative findings <strong>of</strong> the old epidemiological studies.<br />

This is one justification for the biomonitoring studies.<br />

Another justification is on exposures where epidemiological studies have<br />

not been conducted or have provided inadequate results, in spite <strong>of</strong><br />

suspicions raised by short-term or animal experiments. The International<br />

Agency for Research on Cancer has pointed out this as one <strong>of</strong> the criteria<br />

to be used in the evaluation <strong>of</strong> carcinogenicity <strong>of</strong> chemicals (IARC, 1992).<br />

Styrene belongs to this group <strong>of</strong> industrial exposures, where<br />

epidemiological findings are equivocal but adduct data are available on<br />

workers (Vodicka et al., 1993).<br />

Acknowledgements<br />

The research was supported by the Swedish Medical Research Council and<br />

Work Environment Fund.


References<br />

K.HEMMINKI 165<br />

BEACH, A. and GUPTA, R., 1992, Human biomonitoring and the 32 P-postlabeling<br />

assay, Carcinogenesis, 13, 1053–74.<br />

CARSTENSEN, U., ALEXANDRIE, A.-K., HÖGSTEDT,B., RANNUG, A.,<br />

BRATT, I. and HAGMAR, L., 1993, B- and T-lymphocyte micronuclei in<br />

chimney sweeps with respect to genetic polymorphism for CYP1A1 and GST1<br />

(class Mu), Mutat. Res., 289, 187–95.<br />

GRZYBOWSKA, E., HEMMINKI, K., SZELINGA, J. and CHORAZY, M., 1993,<br />

Sea-sonal variation <strong>of</strong> aromatic DNA adducts in human lymphocytes and<br />

granulocytes, Carcinogenesis, 14, 2523–6.<br />

HARRIS, C., 1991, Chemical and physical carcinogenesis: advances and<br />

perspectives for the 1990s, Cancer Res., 51, 5023–44.<br />

HAYASHI, S., WATANABE, J., NAKACHI, K. and KAWAJIRI, K., 1991, Genetic<br />

linkage <strong>of</strong> lung cancer-associated Msp I polymorphisms with amino acid<br />

replacement in the heme binding region <strong>of</strong> the human cytochrome P4501A1<br />

gene, J. Biochem., 110, 407–11.<br />

HEMMINKI, K., 1993, DNA adducts, mutations and cancer, Carcinogenesis, 14,<br />

2007– 12.<br />

HEMMINKI, K., 1995, DNA adducts in biomonitoring, J. Occup. Environ. Med.,<br />

37, 44–51.<br />

HEMMINKI, K., AUTRUP, H. and HAUGEN, A., 1993a, Environmental<br />

carcinogens: Assessment <strong>of</strong> Exposure and Effect, pp. 89–102, Heidelberg:<br />

Springer Verlag.<br />

HEMMINKI, K., FÖRSTI, A., LÖFGREN, M., SÄGERBÄCK, D., VACA, C. and<br />

VODICKA, P., 1993b, Testing <strong>of</strong> quantitative parameters in the 32 Ppostlabelling<br />

method, in Phillips, D.H., Castegnaro, M. and Bartsch, H. (Eds),<br />

Postlabelling Methods for Detection <strong>of</strong> DNA Adducts, IARC Sci. Publ., No.<br />

124, pp, 51–63, Lyon: IARC.<br />

HEMMINKI, K., SÖDERLING, J., ERICSON, P., NORBECK, H.E. and<br />

SEGERBÄCK, D., 1994, DNA adducts among personnel servicing and loading<br />

diesel vehicles, Carcinogenesis, 15, 767–9.<br />

IARC, 1992, Mechanisms <strong>of</strong> Carcinogenesis in Risk Identification, IARC Sci. Publ.<br />

No. 116, Lyon: IARC.<br />

IARC, 1993, Postlabelling Methods for Detection <strong>of</strong> DNA Adducts, IARC Sci.<br />

Publ. No. 124, Lyon: IARC.<br />

IARC, 1994, DNA Adducts: Identification and Biological Significance, IARC Sci.<br />

Publ. No. 125, Lyon: IARC.<br />

ICHIBA, M., HAGMAR, L., RANNUG, A.O., HÖGSTEDT, B., ALEXANDRIE, A.-<br />

K. and HEMMINKI, K., 1994, Aromatic DNA adducts, micronuclei and<br />

genetic polymorphism for CYP1A1 and GST1 in chimney sweeps,<br />

Carcinogenesis, 15, 1347–52.<br />

NAKACHI, K., IMAI, K., HAYASHI, S.I., WATANABE, J., KAWAJIRI, K.,<br />

HAYASHI, S.-I., WATANABE, J. and KAWAJIRI, K., 1992, High<br />

susceptibility to lung cancer analyzed in terms <strong>of</strong> combined genotypes <strong>of</strong><br />

P450IA1 and mu-class glutathione S-transferase genes, Jpn J. Cancer Res., 83,<br />

866–70.


166 BIOMARKERS AND RISK ASSESSMENT<br />

NEBERT, D.W., 1991, Role <strong>of</strong> genetics and drug metabolism in human cancer<br />

risk, Mutat. Res., 247, 267–81.<br />

ØVREBø, S., HAUGEN, A., FJELDSTAD, P.E., HEMMINKI, K. and SZYFTER, K.,<br />

1994, Biological monitoring <strong>of</strong> exposure to PAH in an electrode paste plant, J.<br />

Occup. Med., 36, 303–10.<br />

ØVREBø, S., HAUGEN, A., HEMMINKI, K., SZYFTER, K., DRABLÖS, P.A. and<br />

SKOGLAND, M., 1995, Studies <strong>of</strong> biomarkers in aluminium workers<br />

occupationally exposed to polycyclic aromatic hydrocarbons, Cancer<br />

Detection Prev., 19, 258.<br />

PERERA, F.P., TANG, D.L., O’NEILL, J.P., BIGBEE, W.L., ALBERTINI, R.J.,<br />

SANTELLA, R., OTTMAN, R., TSAI, W.Y., DICKEY, C., MOONEY, L.A.,<br />

SAVELA, K. and HEMMINKI, K., 1993, HPRT and glycophorin A mutations<br />

in foundry worker: relationship to PAH exposure and PAH-DNA adducts,<br />

Carcinogenesis, 14, 969–73.<br />

PERERA, F.P., DICKEY, C., SANTELLA, R., O’NEILL, J.P., ALBERTINI, R.J.,<br />

OTTMAN, R., TSAI, W.Y., MOONEY, L.A., SAVELA, K. and HEMMINKI,<br />

K., 1994, Carcinogen-DNA adducts and gene mutations in foundry workers<br />

with changing exposure to PAH, Carcinogenesis, 15, 2905–10.<br />

SANTELLA, R.M., HEMMINKI, K., TANG, D.-L., PAIK, M., OTTMAN, R.,<br />

YOUNG, T.L., SAVELA, K., VODICKOVA, L., DICKEY, C., WHYATT, R.<br />

and PERERA, P.P., 1993, Polycyclic aromatic hydrocarbon-DNA adducts in<br />

white blood cells and urinary 1-hydroxypyrene in foundry workers, Cancer<br />

Epidemiol. Biomarkers Prevent., 2, 59–62.<br />

SAVELA, K. and HEMMINKI, K., 1991, DNA adducts in lymphocytes and<br />

granulocytes <strong>of</strong> smokers and non-smokers detected by the 32 P-postlabelling<br />

assay, Carcinogenesis, 12, 503–8.<br />

SEGERBÄCK, D. and VODICKA, P., 1993, Recoveries <strong>of</strong> DNA adducts <strong>of</strong><br />

polycyclic aromatic hydrocarbons in the 32 P-postlabelling assay,<br />

Carcinogenesis, 14, 2463–9.<br />

SEIDEGǺRD, J., PERO, R.W., MILLER, D.G. and BEATTIE, E.J., 1986, A glutathione<br />

transferase in human leukocytes as a marker for the susceptibility to<br />

lung cancer, Carcinogenesis, 7, 751–3.<br />

SZYFTER, K., KRUGER, J., ERICSON, P., VACA, C., FÖRSTI, A. and<br />

HEMMINKI, K., 1994, 32 P-postlabelling analysis <strong>of</strong> DNA adducts in humans:<br />

adducts distribution and method improvement, Mut. Res., 313, 269–76.<br />

TATES, A.D., GRUMMT, T., TÖRNQVIST, M., FARMER, P.B., VAN DAM,<br />

F.J., VAN MOSSEL, H., SCHOEMAKER, H.M., OSTERMAN-GOLKAR, S.,<br />

UEBEL, C., TANG, Y.S., ZWINDERMAN, A.H., NATARAJAN, A.T. and<br />

EHRENBERG, L., 1991, Biological and chemical monitoring <strong>of</strong> occupational<br />

exposure to ethylene oxide, Mut. Res., 250, 483–97.<br />

VODICKA, P., VODICKOVA, L. and HEMMINKI, K., 1993, 32 P-postlabelling <strong>of</strong><br />

DNA adduct <strong>of</strong> styrene-exposed lamination workers, Carcinogenesis, 14,<br />

2007–12.


13<br />

Extrapolation <strong>of</strong> Toxicity Data and Assessment<br />

<strong>of</strong> Risk<br />

NORBERT FEDTKE<br />

Hüls AG, Marl<br />

Introduction<br />

Risk assessment provides a link between scientific research and risk<br />

management, or in other words, it is ‘a method for reaching public policy<br />

decisions’ (Silbergeld, 1993). Risk assessment includes the key elements<br />

hazard identification, dose-response assessment, and exposure assessment.<br />

These elements are integrated in a risk characterization step to predict<br />

adverse effects that may occur in a given population in a particular<br />

exposure situation, <strong>of</strong>ten based on the quantification <strong>of</strong> the likelihood <strong>of</strong> this<br />

occurrence. Risk management determines whether the particular exposure<br />

situation presents an acceptable or unacceptable risk and whether it is<br />

necessary to reduce the risk by reducing the exposure. Whereas risk<br />

management has to account for public health, socio-economical factors,<br />

technical feasibility, social perceptions, governmental policy and political<br />

consequences, risk assessment should be based on scientific principles.<br />

Since for the majority <strong>of</strong> industrial chemicals no or only limited human<br />

data exist, the question <strong>of</strong> how to extrapolate the data obtained from<br />

laboratory studies in experimental animals in order to predict the effects in<br />

humans has become one important aspect in risk assessment. The final<br />

goal is either to determine a level <strong>of</strong> exposure at which there is no reasonable<br />

doubt that an adverse effect will not occur in man or to define the risk<br />

associated with this exposure level. The use <strong>of</strong> mechanistic information to<br />

provide linkages between exposure, dose to tissue, and biological responses<br />

may assist in some <strong>of</strong> the steps necessary in the process <strong>of</strong> species<br />

extrapolation. Especially the use <strong>of</strong> physiologically based pharmacokinetic<br />

models (PBPK) for some aspects <strong>of</strong> risk assessment has been promoted to<br />

reduce the uncertainty associated with the current default methods. PBPK<br />

modeling is explained in general terms and a recently developed PBPK model<br />

for 2-butoxyethanol is provided as an example to illustrate the use <strong>of</strong><br />

kinetic and mechanistic data in risk assessment.


168 EXTRAPOLATION OF TOXICITY DATA AND ASSESSMENT OF RISK<br />

Legislative background in the European Union<br />

Although the process <strong>of</strong> risk assessment is not new for the individual<br />

member states <strong>of</strong> the European Union (EU) and representatives <strong>of</strong> industry<br />

and government have been assessing risk for human health and the<br />

environment for decades, EU legislation for existing 1 and new 2 substances<br />

did not formally require a systematic risk assessment up to 1992. The<br />

situation has changed with the Seventh Amendment <strong>of</strong> the Directive on the<br />

Classification, Packaging and Labelling <strong>of</strong> Dangerous Substances (EEC,<br />

1992) and the existing substances regulation (EEC, 1993a), which address<br />

risk assessment <strong>of</strong> new and existing substances, respectively.<br />

For new chemicals, the general principles <strong>of</strong> risk assessment are defined<br />

in Commission Directive 93/67/EEC (EEC, 1993b). In addition, the<br />

Directorate General XI <strong>of</strong> the European Commission has issued a series <strong>of</strong><br />

draft guidance documents for use by the competent authorities appointed<br />

by the member states. These documents provide the technical details for the<br />

risk assessment <strong>of</strong> new substances mainly by defining the testing strategies<br />

for individual toxic endpoints. For existing chemicals, a guidance<br />

document has been drafted. However, these technical guidance documents<br />

provide only little information on how to extrapolate laboratory data to<br />

humans. It has to be assumed that the extrapolation principles in the EU<br />

member states will be based on historical approaches used by authorities in<br />

other countries.<br />

Approaches to risk assessment<br />

The final goal <strong>of</strong> species extrapolation is to define a dose or dose rate<br />

which produces no adverse effects in humans. The estimation <strong>of</strong> a human<br />

no-effectlevel may include:<br />

– determination <strong>of</strong> the appropriate animal species for extrapolation to<br />

man,<br />

– determination <strong>of</strong> the most critical effect(s) and the target organ(s),<br />

– determination <strong>of</strong> the no-observed-adverse-effect level(s) (NOAEL) <strong>of</strong><br />

this effect(s), <strong>of</strong>ten followed by<br />

– extrapolation <strong>of</strong> the NOAEL(s) from subacute or subchronic to chronic<br />

exposure (time extrapolation),<br />

– extrapolation <strong>of</strong> effects observed in a high-dose region <strong>of</strong> a dose<br />

response curve to a low-dose region,<br />

1 Listed in the European Inventory <strong>of</strong> Existing Commercial Substances<br />

(EINECS).<br />

2 Not listed in EINECS.


N.FEDTKE 169<br />

– extrapolation <strong>of</strong> effects from one route <strong>of</strong> exposure to another route,<br />

and<br />

– extrapolation <strong>of</strong> effects observed in a rather homogeneous animal<br />

population to a heterogeneous human population (interspecies<br />

extrapolation), which also has to take into account the existence <strong>of</strong><br />

subgroups regarded as more sensitive as the rest <strong>of</strong> the population<br />

(intraspecies extrapolation).<br />

Essential for all procedures used in health risk assessment is the<br />

determination <strong>of</strong> the so-called critical effect. The critical effect may be<br />

defined as the adverse effect judged to be most appropriate as the basis for<br />

the risk assessment. Hence, the first step is the review <strong>of</strong> all available data<br />

on a chemical and the assessment <strong>of</strong> the adequacy <strong>of</strong> the database for the<br />

determination <strong>of</strong> the critical effect. On the basis <strong>of</strong> the critical effect,<br />

toxicants may be divided into two classes characterized by:<br />

– a threshold <strong>of</strong> response, i.e. the adverse effect on health is not expressed<br />

until the chemical, or the ultimately toxic metabolite, reaches a<br />

threshold dose or dose rate in the target tissue, or<br />

– no threshold <strong>of</strong> response, i.e. there is no threshold exposure level below<br />

which effects will not be expressed. This implies that there is some risk at<br />

any level <strong>of</strong> exposure. Examples are genotoxic carcinogens or germ cell<br />

mutagens.<br />

Based on these classes two general approaches to health risk assessment<br />

have been used.<br />

The first approach involves the use <strong>of</strong> ‘safety factors’ applied to the<br />

NOAEL or the lowest-observed-adverse-effect level (LOAEL) <strong>of</strong> a<br />

threshold effect determined in experimental animals (safety factors are<br />

recently referred to as ‘uncertainty’ or ‘assessment’ factors). The magnitude<br />

<strong>of</strong> the uncertainty factors varies between the regulatory bodies that are<br />

concerned with risk assessment, but usually they take into account the<br />

interspecies extrapolation (default factor 10) and intraspecies extrapolation<br />

(default factor 10). The magnitude <strong>of</strong> the default factors appears to be<br />

based more on the conventional use <strong>of</strong> the decimal system than on<br />

scientific reasons and have been proposed first by Lehman and Fitzhugh<br />

(1954) for the derivation <strong>of</strong> acceptable daily intakes (ADIs) for food<br />

additives. Additional uncertainty factors may be used for extrapolation to<br />

chronic exposure from subacute or subchronic exposure, adequacy <strong>of</strong> the<br />

database, extrapolation <strong>of</strong> a LOAEL to a NOAEL and severity <strong>of</strong> effects.<br />

The resulting overall uncertainty factor <strong>of</strong>ten reaches values <strong>of</strong> 1000 or<br />

higher, which is an indication <strong>of</strong> the imprecision <strong>of</strong> the derived tolerable<br />

intake. Refined extrapolation procedures using subdivisions <strong>of</strong> the default


170 EXTRAPOLATION OF TOXICITY DATA AND ASSESSMENT OF RISK<br />

factors or different default factors have recently been published (Lewis et<br />

al., 1990; Renwick, 1991, 1993).<br />

The second approach (for non-threshold effects) also relies mainly on<br />

default assumptions for dose-response extrapolation and cross-species<br />

extrapolation. Especially cancer risk assessment has been the subject <strong>of</strong><br />

much debate and there are a number <strong>of</strong> extrapolation methods reviewed<br />

recently by Park and Hawkins (1993) and Hallenbeck (1993). The default<br />

methodology in the . has been summarized by Frederick (1993). In<br />

principle, the risk assessment is based on a chronic rodent bioassay<br />

conducted at or near the maximum tolerated dose (MTD). The lifetime<br />

constant dose rates and the tumour incidence data for the individual dose<br />

groups are used to determine the dose response by fitting the data with a<br />

computer program. The linearized multistage cancer model (LMS) is <strong>of</strong>ten<br />

used to perform this step. The LMS model extrapolates the rodent tumor<br />

data observed at the MTD to a dose with a predefined risk and the 95 per<br />

cent upper bound on the dose-response curve is calculated. The interspecies<br />

extrapolation to humans is performed by a correction factor based on body<br />

weight or body surface. Subsequently, the dose is determined that<br />

corresponds to a maximum allowable calculated upper bound on risk. The<br />

resulting number does not describe the actual human risk under low-level<br />

environmental exposure, but provides an upper bound to human risk that<br />

is assumed not to be exceeded. The actual risk may be in the range between<br />

0 and the upper bound. In the process described, the dose is defined as<br />

administered dose or inhaled concentration. As a result, the lowdose<br />

extrapolation does not take into account non-linearities in tissue dosimetry<br />

and response. In addition, the interspecies extrapolation is performed using<br />

a default approach that does not account for mechanistic species<br />

differences.<br />

Use <strong>of</strong> PBPK models in risk assessment<br />

General description<br />

Physiologically based pharmacokinetic (PBPK) models have been used<br />

increasingly over the past decade to improve several aspects <strong>of</strong> the<br />

assessment <strong>of</strong> risk associated with human exposure to chemicals. Examples<br />

are PBPK models for styrene (Ramsey and Andersen, 1984; Csanády et al.,<br />

1994), dichloromethane (Andersen et al., 1987), 1,4-dioxane (Reitz et al.,<br />

1990a), chlor<strong>of</strong>orm (Reitz et al., 1990b), ethyl acrylate (Frederick et al.,<br />

1992), methanol (Horton et al., 1992) and 1,3-butadiene (Johanson and<br />

Filser, 1993). Recent reviews <strong>of</strong> the use <strong>of</strong> PBPK models in risk assessment<br />

have been published by several authors (Frederick, 1993; Travis, 1993;<br />

Wilson and Cox, 1993; Andersen and Krishnan, 1994).


N.FEDTKE 171<br />

PBPK models are based on the blood and tissue solubility <strong>of</strong> chemicals,<br />

their metabolism in various tissues and the physiology <strong>of</strong> the organism,<br />

thus incorporating the specific physiological description <strong>of</strong> animal species as<br />

well as specific physico-chemical descriptions <strong>of</strong> agents. Uptake,<br />

distribution, metabolism and excretion are described in physiologically<br />

realistic compartments (tissue groups) using computer simulation. The<br />

compartments are linked in parallel, represent the actual mammalian<br />

architecture, and include tissues such as lung and arterial blood, fatty<br />

tissue, poorly perfused tissues (muscles, skin), richly perfused tissues<br />

(brain, kidneys, heart, endocrine gland, gastro-intestinal tract), liver as the<br />

main metabolizing tissue, and mixed venous blood. The compartments are<br />

connected by arterial and venous blood flow and are characterized by a set<br />

<strong>of</strong> mass balance differential equations. The rate constants that describe the<br />

flow <strong>of</strong> material between the tissue groups and the rate <strong>of</strong> change in the<br />

chemical concentration <strong>of</strong> each compartment are proportional to blood<br />

flow, tissue solubility and compartment volumes. The basic mathematical<br />

description <strong>of</strong> a PBPK model for a volatile compound has been provided by<br />

Ramsey and Andersen (1984) and additional details may be found in<br />

appendices <strong>of</strong> manuscripts dealing with the development <strong>of</strong> PBPK models.<br />

Estimation <strong>of</strong> the constants used in PBPK models may be based on the<br />

literature in the case <strong>of</strong> physiological parameters such as ventilation rates,<br />

cardiac output, blood flow to tissues and tissue volumes. The EPA has<br />

compiled reference values for these parameters and their scaling (Arms and<br />

Travis, 1988). Chemical specific parameters such as blood and tissue<br />

solubilities may be determined from in vitro preparation (Sato and<br />

Nakajima, 1979; Gargas et al., 1989). The biochemical constants for<br />

metabolism may be derived from in vitro studies (Reitz et al. 1988;<br />

Carfagna and Kedderis, 1992; Johanson and Filser, 1993), in vivo<br />

toxicokinetic studies (Potter and Tran, 1993; Frederick et al., 1992) or in<br />

the case <strong>of</strong> volatile substances from gas uptake studies (Gargas et al., 1986,<br />

1990; Filser, 1992).<br />

Since the tissue groups have a defined biological meaning, scaling <strong>of</strong> the<br />

associated parameters between species is possible since many <strong>of</strong> the<br />

parameters used are correlated to body weight. Cardiac output, alveolar<br />

ventilation rate and V max are scaled by the 3/4 power <strong>of</strong> body weight<br />

whereas K m is assumed to be constant across species. However, the<br />

substitution <strong>of</strong> the physiological parameters with the appropriate values<br />

characteristic for the species <strong>of</strong> interest is preferred.<br />

The development <strong>of</strong> PBPK models is an iterative process involving<br />

comparison <strong>of</strong> the model simulations with experimental data and<br />

refinement <strong>of</strong> the estimates when the model fails to accurately predict the<br />

kinetic behaviour. Different exposure scenarios can be used to predict the<br />

concentrations <strong>of</strong> the parent chemical or its metabolites in the blood or the<br />

tissues, which are the target <strong>of</strong> toxic effects. The level <strong>of</strong> glutathione


172 EXTRAPOLATION OF TOXICITY DATA AND ASSESSMENT OF RISK<br />

depletion in hepatic and extrahepatic tissues (D’Souza et al., 1988;<br />

Frederick et al., 1992; Krishnan et al., 1992), kinetic interactions <strong>of</strong> parent<br />

compounds in mixed exposures (Tardif et al., 1993) or the amount <strong>of</strong><br />

adducts formed by macromolecular binding (Krishnan et al., 1992) are<br />

predictions that may also be generated by PBPK modeling. As a result <strong>of</strong><br />

the simulations, quantitative information on the internal dose <strong>of</strong> a<br />

chemical or its metabolites in the target tissue is obtained and can replace<br />

the administered dose conventionally used in risk assessment. After<br />

validation <strong>of</strong> the PBPK models in experimental animals, human PBPK<br />

models can be developed either by allometric scaling <strong>of</strong> the physiological<br />

and biochemical parameters or preferably using the actual human<br />

parameters. Following the prediction <strong>of</strong> the target tissue dosimetry in<br />

humans, the appropriate dose surrogates are related to the effect <strong>of</strong> interest<br />

and quantitative species differences are determined. This information<br />

provides the possibility to base the species extrapolation on scientific data<br />

instead <strong>of</strong> on arbitrarily assigned default factors and as a consequence the<br />

uncertainty <strong>of</strong> the extrapolation procedures applied in conventional risk<br />

assessment may be reduced.<br />

Description and use <strong>of</strong> the PBPK model for 2butoxyethanol<br />

2-Butoxyethanol (BE) is a widely produced glycol ether used as a key<br />

ingredient in water- or solvent-based coatings, industrial and consumer<br />

cleaning products, and as solvent in a variety <strong>of</strong> products. Haemolysis was<br />

identified as most sensitive indicator <strong>of</strong> BE-induced toxicity in several<br />

species <strong>of</strong> laboratory animals and has received the most attention as a<br />

critical effect for human risk assessment (ECETOC, 1985, 1994). The<br />

experimentally determined subchronic NOAEL for the rat is 25 ppm. The<br />

major metabolite <strong>of</strong> BE is 2-butoxyacetic acid (BAA) which has been<br />

identified as the metabolite responsible for the haemolysis <strong>of</strong> red blood<br />

cells in in vitro and in vivo studies (Bartnik et al., 1987; Ghanayem et al.,<br />

1987; Ghanayem, 1989). Changes in the deformability <strong>of</strong> rat erythrocytes<br />

appear to precede haemolysis upon treatment with BAA. Treatment <strong>of</strong><br />

human erythrocytes with BAA did not induce changes in deformability<br />

(Udden and Patton, 1994; Udden, 1994). The observed species differences<br />

may be due to differences in the lipid composition <strong>of</strong> erythrocyte<br />

membranes, differences in membrane proteins associated with anion<br />

transport processes, or differences in the erythrocyte cytoskeleton (Udden,<br />

1994; Udden and Patton, 1994). Humans are most likely to be exposed to<br />

BE by the dermal or inhalation routes due to the widespread use <strong>of</strong> BE in<br />

cleaning products. Assessment <strong>of</strong> the risk resulting from BE use has to<br />

account for these routes <strong>of</strong> exposure and the formation <strong>of</strong> BAA as the<br />

active metabolite. In order to assist in the risk assessment, PBPK models


N.FEDTKE 173<br />

were developed that describe the uptake, metabolism and disposition <strong>of</strong> BE<br />

and BAA (Johanson, 1986; Corley et al., 1993, 1994; Shyr et al., 1993).<br />

The model <strong>of</strong> Corley et al. (1993, 1994) is a refinement <strong>of</strong> Johanson’s<br />

model (1986) and consists <strong>of</strong> two submodels. The first submodel describes<br />

the uptake and disposition <strong>of</strong> BE and consists <strong>of</strong> the tissue compartments<br />

rapidly perfused organs, slowly perfused organs, fat, skin, muscle,<br />

gastrointestinal tract, and liver as the metabolizing tissue. The BE<br />

submodel allows uptake via the inhalation and dermal routes and in<br />

addition provides the possibility <strong>of</strong> uptake via IV infusion and the<br />

gastrointestinal tract in order to validate the model with laboratory data.<br />

The second submodel tracks the disposition <strong>of</strong> BAA in the same tissue<br />

compartments, but the kidney was removed from the rapidly perfused<br />

organs as separate tissue to allow for the excretion <strong>of</strong> BAA metabolites.<br />

The two submodels are linked together by the metabolism <strong>of</strong> BE to BAA<br />

via a saturable enzymatic pathway catalyzed by alcohol and aldehyde<br />

dehydrogenases in the liver. Competing pathways (BE conjugation and<br />

BE O-dealkylation) are lumped together and described by an additional<br />

enzymatic pathway with Michaelis-Menten kinetics. The model assumes<br />

that BAA is bound to proteins in blood and is eliminated by a saturable<br />

process in the kidneys. The rate <strong>of</strong> BAA elimination by the kidneys is<br />

described as the sum <strong>of</strong> glomerular filtration rate <strong>of</strong> BAA and the acid<br />

transport <strong>of</strong> BAA assuming that no reabsorption occurs. The biochemical<br />

constants determined experimentally in the rat were scaled to humans by<br />

(body weight) 0.7 . In the validation process, the model successfully described<br />

a wide variety <strong>of</strong> rat and human data from different laboratories using<br />

several routes <strong>of</strong> administration.<br />

BAA was predicted to be formed more rapidly in rats compared with<br />

humans, but to be eliminated slower in humans than in rats. In summary,<br />

higher maximum concentrations <strong>of</strong> BAA in blood (C max) and also higher<br />

areas under the BAA concentration-time curves (AUC) were predicted for<br />

rats than for humans, especially as the vapour concentration was<br />

increased. For the purpose <strong>of</strong> dose-response and interspecies extrapolation,<br />

BAA-C max and BAA-AUC were used as estimates <strong>of</strong> the internal dose<br />

surrogate; C max can be related directly to the in vitro haemolysis studies<br />

with BAA and is responsive to the dose-rate. The in vitro studies performed<br />

(Bartnik et al., 1987; Ghanayem et al., 1987; Ghanayem, 1989; Udden,<br />

1994; Udden and Patton, 1994) suggest that approximately 0.2 mM BAA<br />

is required to produce slight haemolysis <strong>of</strong> rat red blood cells. At about 2<br />

mM BAA nearly complete haemolysis was observed. The model predicts<br />

for nose-only exposure that these concentrations are reached in the rat at<br />

BE exposure concentrations <strong>of</strong> about 100 ppm and 800 ppm for 6 h,<br />

respectively, which is consistent with observations in vivo (Tyler, 1984;<br />

Sabourin et al., 1992).


174 EXTRAPOLATION OF TOXICITY DATA AND ASSESSMENT OF RISK<br />

For human red blood cells, the minimum BAA-concentration necessary<br />

to induce slight haemolysis is about 40 times higher compared with rats,<br />

i.e. 8 mM. The model predicts for human nose-only exposure that the C max<br />

<strong>of</strong> BAA in blood is slightly lower than the value observed in rats at a BE<br />

exposure concentration <strong>of</strong> about 100 ppm for 6 h and is only about 50 per<br />

cent <strong>of</strong> the BAA rat blood concentration at 800 ppm. In any case, the<br />

minimum toxic concentration <strong>of</strong> approximately 8 mM BAA in human<br />

blood is not achieved.<br />

The AUC has a time component which is important since haemolysis is<br />

not an instantaneous response (Udden, 1994; Udden and Patton, 1994).<br />

With respect to the AUCs for BAA, the model predicts that the values for<br />

rat and human blood are similar up to a BE exposure concentration <strong>of</strong><br />

about 500 ppm. Higher BE concentrations cause higher AUCs for BAA in<br />

rat blood than in human blood. Thus, the model predicted a BAA-AUC in<br />

man at 22 ppm BE vapour exposure that was similar to the BAA-AUC in<br />

rats achieved at 25 ppm BE vapour exposure, the established subchronic<br />

NOAEL.<br />

The simulation <strong>of</strong> the dermal BE uptake assumed that 10 per cent <strong>of</strong> the<br />

body surface <strong>of</strong> rats and humans were exposed for 6 h to BE solutions in<br />

water (5–100 per cent) and that no losses <strong>of</strong> BE occurred from the dosing<br />

solution. The simulation predicted C max blood concentrations for BAA in<br />

rats that were highest (about 3 mM) for a 40 per cent BE solution. For<br />

humans, BAA-C max was predicted to reach about 1.3 mM for the same BEconcentration.<br />

Predicted BAA-AUCs were about tw<strong>of</strong>old higher in rats<br />

compared with humans. Under these worst-case assumptions, no BE<br />

concentration is expected to achieve BAA concentrations in human blood<br />

that would cause haemolysis.<br />

ECETOC (1994) used the described PBPK model for BE and BAA<br />

disposition in combination with mechanistic data obtained by in vitro<br />

experiments to recommend an occupational exposure limit for BE:<br />

– BAA-AUC was used as the internal dose surrogate and 22 ppm BE<br />

vapour was predicted to cause a BAA-AUC in human blood similar to<br />

the BAA-AUC in rats exposed to a BE-concentration <strong>of</strong> 25 ppm<br />

(identified as the subchronic rat NOAEL). At 22 ppm BE vapour, the<br />

BAA-C max (33 µM) is predicted to be several hundredfold below the<br />

BAA concentration that causes pre-haemolytic effects in human red<br />

blood cells (8 mM).<br />

– ECETOC did not use an uncertainty factor for intraspecies<br />

extrapolation, since the in vitro studies indicated no increased sensitivity<br />

<strong>of</strong> red blood cells from individuals regarded as susceptible to haemolytic<br />

effects such as older persons, persons with hereditary spherocytosis or<br />

sickle cell disease (Udden, 1994; Udden and Patton, 1994).


– An uncertainty factor for time extrapolation (subchronic to chronic<br />

exposure) was also not applied, since the red blood cell haemolysis was<br />

regarded as a transient phenomenon observed predominantly on the<br />

first few days <strong>of</strong> exposure thus indicating that longer exposure would<br />

not have resulted in a lower rat NOAEL.<br />

– Although there is some uncertainty about the actual magnitude <strong>of</strong> the<br />

contribution <strong>of</strong> dermal uptake to the total uptake during BE vapour<br />

exposure, ECETOC concluded that even under worst-case conditions<br />

the BAA concentrations achieved are not sufficient to cause haemolysis<br />

in man and there is no need for the adjustment <strong>of</strong> the predicted human<br />

NOAEL for route.<br />

In conclusion, an occupational exposure limit <strong>of</strong> 20 ppm (8 h TWA) was<br />

recommended, also taking into account all other effects that may be<br />

associated with BE-exposure. This value is similar to the rat NOAEL <strong>of</strong> 25<br />

ppm for the most sensitive parameter, i.e. haemolysis, and was derived<br />

using scientific data instead <strong>of</strong> applying default factors to the rat NOAEL,<br />

a procedure which would have overpredicted the human risk associated<br />

with BE-exposure.<br />

Conclusion<br />

N.FEDTKE 175<br />

The use <strong>of</strong> PBPK models and mechanistic data in risk assessment tends to<br />

reduce the uncertainties in comparison with default methodologies by<br />

replacing the administered dose with the delivered dose and also tends to<br />

reveal uncertainties concealed in default methodologies (Wilson and Cox,<br />

1993). However, there are also limitations in the development <strong>of</strong> PBPK<br />

models. One limitation is that the mechanism <strong>of</strong> the toxic effect has to be<br />

known, otherwise the replacement <strong>of</strong> the external dose by internal dose<br />

surrogates is not possible. In addition, extensive validation <strong>of</strong> the model is<br />

necessary in order to replace default approaches in risk assessment. For the<br />

time being, the development <strong>of</strong> PBPK models appears to be restricted to<br />

high production chemicals where the existing data base allows<br />

identification <strong>of</strong> an accepted mechanism <strong>of</strong> toxic action and validation <strong>of</strong><br />

the model. Concern has been expressed that the use <strong>of</strong> point estimates in<br />

PBPK modelling instead <strong>of</strong> ranges <strong>of</strong> biologically plausible values leads to<br />

an increase in the uncertainty (Portier and Kaplan, 1989). However, a<br />

recent study from the Delivered Dose Work Group <strong>of</strong> the American<br />

<strong>Industrial</strong> Health Council came to the conclusion that incorporation <strong>of</strong><br />

‘pharmacokinetic information in a risk assessment,…, leads to both a more<br />

accurate estimate <strong>of</strong> risk and a better specification <strong>of</strong> the true uncertainty’<br />

(Wilson and Cox, 1993). A detailed discussion <strong>of</strong> the sources <strong>of</strong><br />

uncertainties is also provided in this reference.


176 EXTRAPOLATION OF TOXICITY DATA AND ASSESSMENT OF RISK<br />

If the data base is sufficient, PBPK models provide scientific credibility to<br />

interspecies extrapolation, extrapolation across routes <strong>of</strong> administration,<br />

extrapolation from high-dose to low-dose and intraspecies extrapolation.<br />

Recent concepts link the original tissue dose concept <strong>of</strong> PBPK models to<br />

biologically based tissue response models, thus relating the delivered dose<br />

via the mechanism <strong>of</strong> action to the toxic response and developing<br />

integrated biological models (Conolly et al., 1988; Moolgavker et al.,<br />

1988; Cohen and Ellwein, 1990; Conolly and Andersen, 1991). Such<br />

approaches enable scientists to ask the right questions and to design new<br />

mechanistic studies that will lead toward the goal <strong>of</strong> a scientifically-based<br />

risk assessment.<br />

Acknowledgement<br />

The author thanks Richard A.Corley and the Glycol Ether Panel <strong>of</strong> the<br />

Chemical Manufactures Association for providing data on 2butoxyethanol.<br />

References<br />

ANDERSEN, M.E. and KRISHNAN, K., 1994, Physiologically based<br />

pharmacokinetics and cancer risk assessment, Environ. Health Perspect.<br />

Suppl., 102 (Suppl. 1), 103–8.<br />

ANDERSEN, M.E., CLEWELL, H.J., GARGAS, M.L., SMITH, F.A. and REITZ,<br />

R.H., 1987, Physiologically based pharmacokinetics and the risk assessment<br />

process, for methylene chloride, Toxicol. Appl. Pharmacol., 87, 185–205.<br />

ARMS, A.D. and TRAVIS, C.C., 1988, Reference physiological parameters in<br />

pharmacokinetic modeling, United States Evironmental Protection Agency,<br />

EPA/600/6– 88/004, reproduced by US Department <strong>of</strong> Commerce, National<br />

Technical Information Service, Springfield, VA.<br />

BARTNIK, F.G., REDDY, A.K., KLECAK, G., ZIMMERMANN, V., HOSTYNEK,<br />

J.J. and KUENSTLER, K., 1987, Percutaneous absorption, metabolism, and<br />

hemolytic activity <strong>of</strong> n-butoxyethanol, Fundam. Appl. Toxicol., 8, 59–70.<br />

CARFAGNA, M.A. and KEDDERIS, G.L., 1992, Isolated hepatocytes as in vitro<br />

models for the biotransformation and toxicity <strong>of</strong> chemicals in vivo, CIIT<br />

Activities, 12, 1–6.<br />

COHEN, S.M. and ELLWEIN, L.B., 1990, Proliferative and genotoxic cellular<br />

effects in 2-acetylamin<strong>of</strong>luorene bladder and liver carcinogenesis: biological<br />

modeling <strong>of</strong> the ED01 study, Toxicol. Appl. Pharmacol, 104, 79–93.<br />

CONOLLY, R.B. and ANDERSEN, M.E., 1991, Biologically based<br />

pharmacodynamic models: tools for toxicological research and risk<br />

assessment, Ann. Rev. Pharmacol. Toxicol., 31, 503–23.<br />

CONOLLY, R.B., REITZ, R.H., CLEWELL, H.J., III and ANDERSEN, M.E.,<br />

1988, Pharmacokinetics, biochemical mechanism and mutational


N.FEDTKE 177<br />

accumulation: a comprehensive model for chemical carcinogenesis, Toxicol.<br />

Lett., 43, 189–200.<br />

CORLEY, R.A., BORMETT, G.A., MARKLEY, B.J. and STACK, C.R., 1993, A<br />

physiologically-based pharmacokinetic model for 2-butoxyethanol and its<br />

metabolite, 2-butoxyacetic acid, The Toxicologist, 13, 355, 1387A.<br />

CORLEY, R.A., BORMETT, G.A. and GHANAYEM, B.I., 1994, Kinetics <strong>of</strong> 2butoxyethanol<br />

and its major metabolite 2-butoxyacetic acid in rats and humans,<br />

Toxicol. Appl. Pharmacol., submitted.<br />

CSANÁDY, G.A., MENDRALA, A.L., NOLAN, R.J. and FILSER, J.G., 1994, A<br />

physiologic pharmacokinetic model for styrene and styrene-7,8-oxide in mouse,<br />

rat and man, Arch. Toxicol. (in press).<br />

D’SOUZA, R.W., FRANCIS, W.R. and ANDERSEN, M.E., 1988, Physiological<br />

model for tissue glutathione depletion and increased resynthesis after ethylene<br />

dichloride exposure, J. Pharmacol. Exp. Therap., 245, 563–8.<br />

ECETOC, 1985, Technical Report No. 17. The <strong>Toxicology</strong> <strong>of</strong> Glycol Ethers and<br />

its Relevance to Man: an Updating <strong>of</strong> ECETOC Technical Report No. 14,<br />

Brussels: European Centre for Ecotoxicology and <strong>Toxicology</strong> <strong>of</strong> Chemicals.<br />

ECETOC, 1994, Special Report No. 7. Butoxyethanol Criteria Document<br />

(including a supplement for 2-butoxyethyl acetate), Brussels: European Centre<br />

for Eco-toxicology and <strong>Toxicology</strong> <strong>of</strong> Chemicals.<br />

EEC, 1992, Council Directive 92/32/EEC <strong>of</strong> 30 April 1992 amending for the 7th<br />

time Council Directive 67/548/EEC on the approximation <strong>of</strong> laws, regulations<br />

and administrative provisions relating to the classification, packaging and<br />

labelling <strong>of</strong> substances, Offic. J. Europ. Communities, L 154/1.<br />

EEC, 1993a, Council Regulation EEC 793/93 <strong>of</strong> 23 March 1993 on the evaluation<br />

and control <strong>of</strong> the risks <strong>of</strong> existing substances, Offic. J. Europ. Communities, L<br />

84.<br />

EEC, 1993b, Commission Directive 93/67/EEC <strong>of</strong> 20 July 1993 laying down the<br />

principles for assessment <strong>of</strong> risks to man and the environment <strong>of</strong> substances<br />

notified in accordance with Council Directive 67/548/EEC, Offic. J. Europ.<br />

Communities, L 227/9.<br />

FILSER, J.G., 1992, The closed chamber technique—uptake, endogenous<br />

production, excretion, steady state kinetics and rates <strong>of</strong> metabolism <strong>of</strong> gases<br />

and vapours, Arch. Toxicol, 66, 1–10.<br />

FREDERICK, C.B., 1993, Limiting the uncertainty in risk assessment by the<br />

development <strong>of</strong> physiologically based pharmacokinetic and pharmacodynamic<br />

models, Toxicol. Lett., 68, 159–75.<br />

FREDERICK, C.B., POTTER, D.W., CHANG-MATEU, M.I. and ANDERSEN,<br />

M. E., 1992, A physiologically based pharmacokinetic and pharmacodynamic<br />

model to describe the oral dosing <strong>of</strong> rats with ethyl acrylate and its implications<br />

for risk assessment, Toxicol. Appl. Pharmacol., 114, 246–60.<br />

GARGAS, M.L., ANDERSEN, M.E. and CLEWELL, H.J., III, 1986, A physiologically<br />

based simulation approach for determining metabolic constants from gas<br />

uptake data, Toxicol. Appl. Pharmacol., 86, 341–52.<br />

GARGAS, M.L., BURGESS, R.J., VOISARD, D.E., CASON, G.H. and<br />

ANDERSEN, M.E., 1989, Partition coefficients <strong>of</strong> low-weight volatile<br />

chemicals in various liquids and tissues, Toxicol. Appl. Pharmacol., 98, 87–99.


178 EXTRAPOLATION OF TOXICITY DATA AND ASSESSMENT OF RISK<br />

GARGAS, M.L., CLEWELL, H.J. and ANDERSEN, M.E., 1990, Gas uptake<br />

inhalation techniques and the rates <strong>of</strong> metabolism <strong>of</strong> chloromethanes,<br />

chloroethanes and chloroethylenes in the rat, Inhal. Toxicol., 2, 285–309.<br />

GHANAYEM, B.I., 1989, Metabolic and cellular basis <strong>of</strong> 2-butoxyethanol-induced<br />

hemolytic anemia in rats and assessment <strong>of</strong> human risk in vitro, Biochem.<br />

Pharmacol., 8, 1679–84.<br />

GHANAYEM, B.I., BURKA, L.T. and METTHEWS, H.B., 1987, Metabolic basis<br />

<strong>of</strong> ethylene glycol monobutyl ether (2-butoxyethanol) toxicity: role <strong>of</strong> alcohol<br />

and aldehyde dehydrogenases, J. Pharmacol. Exp. Therap., 242, 222–31.<br />

HALLENBECK, W.H., 1993, Quantitative Risk Assessment <strong>of</strong> Environmental and<br />

Occupational Health, Chelsea, MI: Lewis Publisher, Inc.<br />

HORTON, V.L., HIGUCHI, M.A. and RICKERT, D.E., 1992, Physiologically<br />

based pharmacokinetic model for methanol in rats, monkeys and humans,<br />

Toxicol. Appl. Pharmacol., 117, 26–36.<br />

JOHANSON, G., 1986, Physiologically-based pharmocokinetic modeling <strong>of</strong><br />

inhaled 2-butoxyethanol in man, Toxicol. Lett., 34, 23–31.<br />

JOHANSON, G. and FILSER, J.G., 1993, A physiologically based pharmacokinetic<br />

model for butadiene and its metabolite butadiene monoxide in rat and mouse<br />

and its significance for risk extrapolation, Arch. Toxicol., 67, 151–63.<br />

KRISHNAN, K., GARGAS, M.L., FENNELL, T.R. and ANDERSEN, M.E., 1992,<br />

A physiologically based description <strong>of</strong> ethylene oxide dosimetry in the rat,<br />

Toxicol. Ind. Health, 8, 121–40.<br />

LEHMAN, A.J. and FITZHUGH, O.G., 1954, 100-fold margin <strong>of</strong> safety, Assoc.<br />

Food Drug Off. US. Bull., 18, 33–5.<br />

LEWIS, S.C., LYNCH, J.R. and NIKIFOROV, A.I., 1990, A new approach to<br />

deriving exposure guidelines from ‘no-observed-adverse-effect levels’, Regul.<br />

Toxicol. Pharmacol., 11, 314–30.<br />

MOOLGAVKER, S.H., DEWANJI, A. and VENZON, D.J., 1988, A stochastic<br />

two-stage model for cancer risk assessment. I. The hazard function and the<br />

probability <strong>of</strong> tumor, Risk Anal., 8, 383–92.<br />

PARK, C.N. and HAWKINS, N.C., 1993, An overview <strong>of</strong> cancer risk assessment,<br />

<strong>Toxicology</strong> Methods, 3, 63–86.<br />

PORTIER, C.J. and KAPLAN, N.L., 1989, Variability <strong>of</strong> safe dose estimates when<br />

using complicated models <strong>of</strong> the carcinogenic process. A case study: methylene<br />

chloride, Toxicol. Appl. Pharmacol, 13, 533–44.<br />

POTTER, D.W. and TRAN, T.-B., 1993, Apparent rates <strong>of</strong> glutathione turnover in<br />

rat tissues, Toxicol. Appl. Pharmacol., 120, 186–92.<br />

RAMSEY, J.C. and ANDERSEN, M.E., 1984, A physiologically based description<br />

<strong>of</strong> the inhalation pharmacokinetics <strong>of</strong> styrene in rats and humans, Toxicol.<br />

Appl. Pharmacol., 73, 159–75.<br />

REITZ, R.H., MENDRALA, A.L., PARK, C.N., ANDERSEN, M.E. and<br />

GUENGERICH, F.P., 1988, Incorporation <strong>of</strong> in vitro enzyme data into the<br />

physiologically based pharmacokinetic model for methylene chloride:<br />

implications for risk assessment, Toxicol. Lett., 43, 97–116.<br />

REITZ, R.H., MCCROSKY, P.S., PARK, C.N., ANDERSEN, M.E. and GARGAS,<br />

M.L., 1990a, Development <strong>of</strong> a physiologically based pharmacokinetic model<br />

for risk assessment with 1,4-dioxane, Toxicol. Appl. Pharmacol, 105, 37–54.


N.FEDTKE 179<br />

REITZ, R.H., MANDRELA, A.L., CORLEY, R.A., QUAST, J.F., GARGAS, M.<br />

L., ANDERSEN, M.E., STAATS, D.A. and CONOLLY, R.B., 1990b,<br />

Estimating the risk <strong>of</strong> liver cancer associated with human exposures to<br />

chlor<strong>of</strong>orm using physiologically based pharmacokinetic modeling, Toxicol.<br />

Appl. Pharmacol., 105, 443–59.<br />

RENWICK, A.G., 1991, Safety factors and establishment <strong>of</strong> acceptable daily<br />

intakes, Food Add. Contam., 8, 135–50.<br />

RENWICK, A.G., 1993, Data derived safety for the evaluation <strong>of</strong> food additives<br />

and environmental contaminants, Food Add. Contam., 10, 337–50.<br />

SABOURIN, P.J., MEDINSKY, M.A., BIRNBAUM, L.S., GRIFFITH, W.C. and<br />

HENDERSON, R.F., 1992, Effect <strong>of</strong> exposure concentration on the<br />

disposition <strong>of</strong> inhaled butoxyethanol by F-344 rats, Toxicol. Appl. Pharmacol,<br />

114, 232–8.<br />

SATO, A. and NAKAJIMA, T, 1979, Partition coefficients <strong>of</strong> some aromatic<br />

hydrocarbons and ketones in water, blood and oil, Br. J. Ind. Med., 36, 231–<br />

4.<br />

SHYR, L.J., SABOURIN, P.J., MEDINSKY, M.A., BIRNBAUM, L.S. and<br />

HENDERSON, R.F., 1993, Physiologically based modeling <strong>of</strong> 2-butoxyethanol<br />

disposition in rats following different routes <strong>of</strong> exposure, Environ. Res., 63,<br />

202–18.<br />

SILBERGELD, E.G., 1993, Risk assessment: the perspective and experience <strong>of</strong> US<br />

Environmentalists, Environ, Health Persp., 101, 100–4.<br />

TARDIF, R., LAPARÉ, S., KRISHNAN, K. and BRODEUR, J., 1993,<br />

Physiologically based modeling <strong>of</strong> the toxicokinetic interaction between<br />

toluene and m-xylene in the rat, Toxicol. Appl. Pharmacol, 120, 266–73.<br />

TRAVIS, C.C., 1993, Interspecies extrapolation <strong>of</strong> toxicological data, in Maibach,<br />

H. I. (Ed.), CRC Series in Dermatology: Clinical and Basic Science, pp. 387–<br />

410, London: CRC Press.<br />

TYLER, T.R., 1984, Acute and subchronic toxicity <strong>of</strong> ethylene glycol monobutyl<br />

ether, Environ. Health Persp., 57, 185–91.<br />

UDDEN, M.L., 1994, Hemolysis and decreased deformability <strong>of</strong> erythrocytes<br />

exposed to butoxyacetic acid, a metabolite <strong>of</strong> 2-butoxyethanol: II. Resistance<br />

in red blood cells from humans with potential susceptibility, J. Appl. Toxicol,<br />

14, 97–102.<br />

UDDEN, M.L. and PATTON, C.S., 1994, Hemolysis and decreased deformability<br />

<strong>of</strong> erythrocytes exposed to butoxyacetic acid, a metabolite <strong>of</strong> 2-butoxyethanol:<br />

I. Sensitivity in rats and resistance in normal humans, J. Appl. Toxicol, 14, 91–<br />

6.<br />

WILSON, A. and Cox, L.A., 1993, Managing Statistical Uncertainties in PBPK<br />

Modeling, Denver, CO: Cox Associates.


14<br />

Molecular Approaches to Assess Cancer Risks<br />

ALAN S.WRIGHT, J.PAUL ASTON, NICO J.VAN SITTERT<br />

and WILLIAM P.WATSON<br />

Sittingbourne Research Centre, Sittingbourne<br />

Introduction<br />

Carcinogenesis is a complex process which is not yet fully understood.<br />

Nevertheless, it is generally accepted that carcinogenesis involves the<br />

accumulation <strong>of</strong> mutations in critical genes: proto-oncogenes and/or<br />

tumour suppressor genes. These mutations transform normal cells into<br />

‘initiated’ cells possessing the full complement <strong>of</strong> genetic changes necessary<br />

for malignancy (Figure 14.1). The critical mutations may result from<br />

exposures to radiation, to genotoxic chemicals or they may arise<br />

‘spontaneously’ as a consequence <strong>of</strong> miscoding errors during the normal<br />

replication <strong>of</strong> DNA. Concomitantly, mutations will also accumulate in<br />

other genes which, although not critical for cancer per se may,<br />

nevertheless, influence cellular character thereby contributing to the<br />

multifaceted nature <strong>of</strong> cancer.<br />

The precise nature and number <strong>of</strong> critical genetic changes required for<br />

initiation have not yet been established but will probably vary from case to<br />

case. Many researchers envisage a strict temporal sequence <strong>of</strong> genetic<br />

changes in carcinogenesis. However, it is probable that the critical<br />

mutations can occur in any sequence and at any time. Indeed, it is clear<br />

that one or more <strong>of</strong> the critical mutations can occur in parental cells.<br />

Transmission (inheritance) <strong>of</strong> these mutations either through the germ line<br />

or via somatic cell division increases the susceptibility <strong>of</strong> the progeny to<br />

carcinogens. Furthermore, it is important to note that each <strong>of</strong> the critical<br />

mutations necessary for malignancy may have a different cause. This<br />

potential for multiple causation has important implications in risk<br />

assessment (vide infra).<br />

Fully initiated cells may not automatically proliferate to form tumours.<br />

One possible explanation is that the surrounding normal cells restrain<br />

the initiated or latent cancer cell by providing essential growth regulators<br />

which are no longer produced by the initiated cell. Nevertheless, partially<br />

and fully initiated cells have a replicative and/or survival advantage over<br />

normal cells. Tissue injury caused by physical trauma, chemical agents or


Figure 14.1 Schematic representation <strong>of</strong> the carcinogenic process.<br />

A.S.WRIGHT ET AL. 181<br />

viruses may have a derestraining effect thereby triggering or facilitating the<br />

replication <strong>of</strong> partially or fully initiated cells to form benign tumours or<br />

malignant tumours. Increased functional demands may also serve to<br />

promote tumour development in affected tissues.<br />

It is clear that chemicals which promote tumour development are very<br />

important determinants <strong>of</strong> carcinogenesis. Indeed, promoting agents<br />

display a marked tendency for organotropism. Promoter action is,<br />

therefore, probably the most important determinant <strong>of</strong> the site <strong>of</strong> tumour<br />

development. Yet genotoxic chemicals which initiate the carcinogenic<br />

process are perhaps viewed with even greater concern. The reasons for this<br />

high level <strong>of</strong> concern hinge mainly on evidence that the mutagenic or<br />

initiating actions <strong>of</strong> genotoxic chemicals are additive, cumulative and<br />

essentially irreversible. Furthermore, in contrast to most other classes <strong>of</strong><br />

toxic chemicals, including promoters operating via cytotoxic mechanisms,<br />

there is no theoretical reason or experimental evidence to support the view<br />

that mutagenic actions <strong>of</strong> genotoxic chemicals are thresholded. For these<br />

reasons even very low exposures <strong>of</strong> genotoxic chemicals are viewed with<br />

concern. These concerns have focused scientific and regulatory attention on<br />

a need to develop sound approaches to manage cancer risks—particularly<br />

low level risks associated with low exposures to genotoxic chemicals<br />

encountered in the occupational or environmental settings. Indeed, apart<br />

from clinical applications, high exposures to genotoxic chemicals cannot be<br />

countenanced.


182 MOLECULAR APPROACHES TO ASSESS CANCER RISKS<br />

Management <strong>of</strong> cancer risks (key requirements)<br />

The management <strong>of</strong> toxicological risks implies a capacity to control<br />

exposures within acceptable safety limits. Effective control is, therefore,<br />

dependent not only on the qualitative detection and identification <strong>of</strong><br />

hazardous chemicals but also on a capacity to determine human exposure<br />

and to evaluate the health risks. This last requirement necessitates a<br />

knowledge <strong>of</strong> potency, i.e. quantitative human dose-response relationships.<br />

In the case <strong>of</strong> genotoxic chemicals, the relevant data reside in the very low<br />

region <strong>of</strong> the dose-response curve.<br />

The concept <strong>of</strong> acceptable risk is readily accepted when applied to the<br />

many classes <strong>of</strong> toxic chemicals operating by a thresholded mechanism<br />

indicative <strong>of</strong> the virtual absence <strong>of</strong> risk at sub-threshold doses. Absolute<br />

safety margins for genotoxic chemicals cannot be guaranteed (vide supra)<br />

leading to the adoption <strong>of</strong> conservative safety measures. Indeed, cursory<br />

analysis might suggest that quantitative risk data are not required for the<br />

effective management <strong>of</strong> cancer risks associated with genotoxic chemicals.<br />

Thus, it is generally accepted that human contact with carcinogens should<br />

be minimised. Purely qualitative identification <strong>of</strong> the hazard would permit<br />

the design <strong>of</strong> measures to limit human exposure and minimise carcinogenic<br />

impact. Indeed, a purely qualitative indication <strong>of</strong> genotoxicity can be an<br />

absolute deterrent to the development <strong>of</strong> new products. Nevertheless,<br />

certain exposures, e.g. to indigenous genotoxic chemicals and natural food<br />

components, are unavoidable. Furthermore, measures to reduce exposures<br />

to ‘avoidable’ genotoxic hazards, e.g. certain combustion products, key<br />

industrial base chemicals and intermediates, are <strong>of</strong>ten difficult and costly.<br />

Quantitative risk assessment is needed to prioritise these hazards and, most<br />

importantly, to determine safety margins. Certainly a failure to determine<br />

carcinogenic potency would lead to uncertainty about the adequacy <strong>of</strong><br />

safety margins and, probably, to unnecessary measures to further reduce<br />

exposures. Thus, despite their additive and cumulative actions, even<br />

genotoxic chemicals can pose a negligible health risk. Of course, the<br />

definition <strong>of</strong> a negligible, i.e. acceptable, risk is a socio-political judgement<br />

which nevertheless has to be realistic in the case <strong>of</strong> unavoidable hazards<br />

and achievable in the case <strong>of</strong> avoidable hazards.<br />

Detection <strong>of</strong> genotoxic carcinogens<br />

Concerns about genotoxic hazards have provided an incentive for the<br />

development <strong>of</strong> a broad range <strong>of</strong> rapid tests to detect intrinsic genotoxic<br />

activity or potential. The principal aim <strong>of</strong> these approaches is to predict<br />

carcinogenic activity or, more accurately, cancer initiating activity. The<br />

most widely used tests are the coupled microsomal-microbial mutation<br />

assays developed by Ames et al. (1973). However, such approaches are


A.S.WRIGHT ET AL. 183<br />

viewed as too remote to be <strong>of</strong> value in estimating cancer risks. The trend is<br />

towards increasingly sensitive and precise technology—particularly generic<br />

methods with potential for direct application in humans.<br />

Advances in molecular biology have permitted the development <strong>of</strong> a new<br />

generation <strong>of</strong> point mutation assays based on DNA base mismatch<br />

technology (Thilly, 1991; Lu and Hsu, 1992). This technology has a<br />

precision far exceeding that <strong>of</strong> conventional biological methods and a<br />

sensitivity permitting direct applications in humans. The full potential <strong>of</strong> this<br />

technology has not yet been realised. However, it seems probable that<br />

detection levels will ultimately obviate a need for prior phenotypic<br />

selection: paving the way to universal application. Avoidance <strong>of</strong> phenotypic<br />

selection would represent a powerful advantage over existing<br />

methodologies by providing a much more direct and reliable route to<br />

determining overall background mutation rates and increments due to<br />

specific exposures <strong>of</strong> key relevance to cancer risk assessment (vide infra).<br />

The most prospective <strong>of</strong> the current assays are those designed to detect<br />

primary DNA damage. Among these procedures, 32 P-post-radiolabelling<br />

technology developed by Randerath et al. (1981) to detect DNA adducts is<br />

by far the most sensitive. The justification for application <strong>of</strong> such a<br />

prospective approach to detect exposure to genotoxic carcinogens hinges<br />

on the causal relationship established between genotoxic activity and<br />

cancer. In general, genotoxic character is conferred by possession <strong>of</strong> a<br />

centre(s) <strong>of</strong> electrophilic reactivity. This reactivity permits the chemical to<br />

undergo chemical reactions with nucleophilic centres in the target molecule<br />

(DNA). In many instances the electrophilic centre(s) is introduced into an<br />

inactive precursor chemical by metabolic activation. Primary products, e.g.<br />

DNA adducts, formed when genotoxic chemicals react with DNA are<br />

generally promutagenic (or lethal) and their occurrence leads to an<br />

increased risk <strong>of</strong> mutation and cancer. There is no known category <strong>of</strong><br />

chemical which forms DNA adducts that can be excluded from this<br />

generalisation. Not all DNA adducts are strongly promutagenic. However,<br />

because electrophiles do not display absolute specificity in their reactions<br />

with nucleophiles, the detection <strong>of</strong> even a weakly promutagenic adduct,<br />

e.g. N 7 -alkyldeoxyguanosine, signals the formation <strong>of</strong> a more strongly<br />

promutagenic adduct, e.g. O 6 -alkyldeoxyguanosine. If follows that the<br />

detection <strong>of</strong> DNA adducts provides qualitative evidence <strong>of</strong> (human)<br />

exposure to a genotoxic carcinogen.


184 MOLECULAR APPROACHES TO ASSESS CANCER RISKS<br />

Identification <strong>of</strong> human carcinogens<br />

Classical epidemiological approaches<br />

Until very recently, epidemiological approaches to detect and identify<br />

environmental carcinogens were based exclusively on the analysis <strong>of</strong><br />

tumour incidence and chromosome aberrations in human populations.<br />

However, the endpoints <strong>of</strong> these biological methods lack the intrinsic<br />

resolving power needed to dis criminate between different contributory<br />

factors. Indeed, it is only in instances <strong>of</strong> specific, high and, <strong>of</strong>ten, localised<br />

exposures that these methods have been effective in identifying specific<br />

causative agents. Nevertheless, the results <strong>of</strong> epidemiological studies<br />

indicate that chemicals, which may include both natural and xenobiotic<br />

compounds in food, drink or in the local or general environment, play a<br />

major and broad role in the aetiology <strong>of</strong> human cancer. The identification<br />

<strong>of</strong> these chemical factors is a major goal in cancer prevention.<br />

In vitro genotoxicity assays<br />

In addition to applications in screening prospective chemical products, in<br />

vitro genotoxicity assays, particularly the Ames test, provided the first<br />

practicable, systematic approach to identify environmental carcinogens.<br />

However, this approach places very heavy demands on the time and effort<br />

required to fractionate environmental samples and test individual<br />

compounds. More importantly, however, like the animal cancer studies<br />

these assays complement or have largely supplanted, the approach is not<br />

specifically targeted towards identifying and prioritising human hazards.<br />

For example, these short-term in vitro test do not provide direct evidence<br />

<strong>of</strong> human exposure or effects.<br />

Molecular epidemiology<br />

32 P-Post-radiolabelling technology for the analysis <strong>of</strong> DNA adducts<br />

provides the basis <strong>of</strong> a very sensitive and generic approach to detect<br />

exposures to genotoxic carcinogens. This technology has universal<br />

application and can be applied to detect DNA adducts formed in<br />

laboratory species or humans during exposures to both known and, as yet,<br />

unidentified genotoxic chemicals at the low concentrations encountered in<br />

the environment and the workplace. Elucidation <strong>of</strong> the chemical structures<br />

<strong>of</strong> adducts in human DNA would provide a basis for identifying the<br />

causative agents and their sources or origins. This possibility <strong>of</strong> identifying<br />

the chemical initiators <strong>of</strong> human cancer is an exciting prospect.<br />

Unfortunately, however, these adducts are present at very low abundances<br />

and this is a major obstacle to identification. Thus, the methods for


detecting DNA adducts are much more sensitive than the physicochemical<br />

methods needed for structural characterisation. A number <strong>of</strong> strategies<br />

have been adopted in attempts to solve this problem.<br />

Protein adducts<br />

Genotoxic chemicals that react with DNA also react with nucleophilic<br />

centres in proteins and may also undergo ‘spontaneous’ and enzymecatalysed<br />

reactions with glutathione leading to the excretion <strong>of</strong> the<br />

corresponding mercapturic acids. Ins<strong>of</strong>ar as the formation <strong>of</strong> protein<br />

adducts and mercapturic acids reflect the formation <strong>of</strong> the corresponding<br />

DNA adducts, their detection may also furnish evidence <strong>of</strong> exposure to a<br />

genotoxic carcinogen.<br />

The potential for reaction <strong>of</strong> genotoxic chemicals with proteins (and<br />

glutathione) is much greater than with DNA. Furthermore, human<br />

proteins, e.g. haemoglobin, are available in much larger quantities and are<br />

more accessible than human tissue DNA. These advantages have been<br />

exploited, particularly in the pioneering work <strong>of</strong> Ehrenberg’s group, to<br />

develop a range <strong>of</strong> procedures for the qualitative and quantitative analysis<br />

<strong>of</strong> protein adducts (Osterman-Golkar et al., 1976; Calleman et al., 1978;<br />

Ehrenberg and Osterman-Golkar, 1980). (For a review <strong>of</strong> the available<br />

methods see Skipper and Naylor, 1991.) The most powerful and generic<br />

approach is undoubtedly that developed by Törnqvist et al. (1986a). An<br />

initial purification or enrichment step is key to any successful method for<br />

the analysis <strong>of</strong> low levels <strong>of</strong> organic residues. The amino-groups <strong>of</strong> the Nterminal<br />

valine residues <strong>of</strong> the α-and<br />

β-chains <strong>of</strong> human haemoglobin are<br />

major targets for reaction with a broad range <strong>of</strong> genotoxic chemicals.<br />

Törnqvist achieved selective enrichment <strong>of</strong> adducted N-terminal valine<br />

residues <strong>of</strong> haemoglobin by devising a modified Edman degradation which<br />

resulted in the scission <strong>of</strong> adducted residues whilst leaving the nonadducted<br />

N-terminal valines intact. This procedure provides the basis for<br />

identifying the adducting moieties and their quantitation by GC/MS.<br />

Applications <strong>of</strong> this technology have furnished evidence <strong>of</strong> background<br />

exposures to a range <strong>of</strong> alkylating species. Protein adduct technology has<br />

the potential for considerable further refinement. The possibility <strong>of</strong> using<br />

immunoaffinity technology to enrich both known and unidentified protein<br />

adducts is currently being explored.<br />

DNA adducts and immunoenrichment<br />

A.S.WRIGHT ET AL. 185<br />

The need for effective enrichment technology for DNA adducts is even<br />

more pressing than for protein adducts. Ideally, the enrichment procedure<br />

should be applied at the earliest possible stage <strong>of</strong> analysis. The procedure


186 MOLECULAR APPROACHES TO ASSESS CANCER RISKS<br />

should be rapid and mild in order to minimise the formation <strong>of</strong> artefacts.<br />

Currently, immunoaffinity technology holds the greatest promise.<br />

Immunoenrichment <strong>of</strong> DNA adducts necessitates antibodies possessing<br />

the appropriate specificities and affinities to permit selective binding <strong>of</strong><br />

adducts at the very low abundances encountered in hydrolysates or enzyme<br />

digests <strong>of</strong> human DNA. The immune system does not normally respond to<br />

small molecules per se. However, the system can be induced to produce<br />

effective antibodies by immunising with the small molecule (hapten)<br />

coupled to a protein. Such treatment induces a spectrum <strong>of</strong> antibodyproducing<br />

cells, each producing a specific antibody. Most <strong>of</strong> these<br />

antibodies recognise various regions (epitopes) <strong>of</strong> the carrier protein while<br />

a few may specifically recognise and bind the small molecule <strong>of</strong> interest.<br />

Suitable antibody-producing cells can be selected and cloned to provide a<br />

permanent source <strong>of</strong> homogenous antibody (monoclonal antibody, Mab).<br />

Mabs can be raised against virtually any organic chemical although some<br />

lower molecular weight compounds (


adducts are not viewed as particularly promutagenic. Nevertheless the N 7 -<br />

atom <strong>of</strong> dG residues in DNA is a major target for adduction and the<br />

detection <strong>of</strong> N 7 -dG adducts signals the production <strong>of</strong> adducts at other<br />

(more critical) sites in DNA (vide supra).<br />

In certain instances, a class <strong>of</strong> adducting moieties may possess a common<br />

structural feature that can be exploited for immunoenrichment. For<br />

example, a Mab has been raised against the major DNA adduct <strong>of</strong> benzo(a)<br />

pyrene (r-7,t-8,t-9-trihydroxy-c-10-(N 2 -deoxyguanosylphosphate)-7,8,9,<br />

10-tetrahydrobenzo(a)pyrene) in a collaborative study with Dr Baan’s<br />

group. This Mab recognises DNA adducts formed by a broad range <strong>of</strong><br />

polycyclic aromatic hydrocarbons (PCAs) including benzo(a)pyrene (BP),<br />

chrysene, benz(a)anthracene, 5-methylchrysene, picene and dibenz(a,h)<br />

anthracene. It seems probable that this Mab recognises the common<br />

trihydric alcohol structure produced when the reactive diol epoxides <strong>of</strong><br />

each <strong>of</strong> these polycyclic compounds reacts with nucleophilic centres in<br />

DNA or other macromolecules. The fact that the Mab does not bind the<br />

corresponding fluoranthene adduct is consistent with the spatial<br />

environment <strong>of</strong> the hydroxyl groups in fluoranthene-DNA adducts which<br />

is completely different from those generated from the other PCAs employed<br />

in this study.<br />

The performance <strong>of</strong> the Mab raised against the major BP-DNA adduct in<br />

the enrichment <strong>of</strong> PCA-DNA adducts is being evaluated using the<br />

immobilised Mab coupled to cyanogen bromide-activated Sepharose 4B.<br />

Results obtained to date demonstrate that the immobilised Mab selectively<br />

adsorbs the major BP-DNA adduct from DNA hydrolysates at abundances<br />

below 1 adduct per 10 9 nucleotide units. Results with the other PCA-DNA<br />

adducts are not yet available. However, the results obtained with the major<br />

BP-DNA adduct underlines the potential <strong>of</strong> immunoenrichment technology<br />

in the qualitative and quantitative analysis <strong>of</strong> adducts. Furthermore, such<br />

results provide an incentive to pursue the development <strong>of</strong> class-specific<br />

antibodies in order to permit or facilitate the identification <strong>of</strong> the chemical<br />

initiators <strong>of</strong> human cancer.<br />

Mercapturic acids<br />

A.S.WRIGHT ET AL. 187<br />

Qualitative analysis <strong>of</strong> mercapturic acids also provides a basis for<br />

identifying human exposures to genotoxic chemicals (vide supra). However,<br />

the available analytical procedures are complex and tend to lack specificity<br />

and sensitivity. During the last dozen years we have undertaken a number<br />

<strong>of</strong> studies aimed at developing compound- and class-specific antibodies to<br />

facilitate the analysis <strong>of</strong> mercapturic acids.<br />

Conventional approaches to generate antibodies to low molecular weight<br />

(MW) organic chemicals involves the covalent attachment <strong>of</strong> the small<br />

molecule (hapten) to a strongly antigenic protein, e.g. keyhole limpet


188 MOLECULAR APPROACHES TO ASSESS CANCER RISKS<br />

haemocyanin or bovine serum albumin, for the immunisation <strong>of</strong> mice. This<br />

strategy is usually effective in the case <strong>of</strong> strongly antigenic haptens, e.g.<br />

aromatic nitro compounds <strong>of</strong> PCA-DNA adducts. However, all <strong>of</strong> our<br />

attempts to use this approach to generate antibodies against relatively low<br />

MW and weakly antigenic mercapturic acids, e.g. S-(2-hydroxyethyl)-Nacetylcysteine,<br />

failed. Antibodies were generated but these were directed<br />

against the strongly antigenic carrier protein(s).<br />

Covalent binding to macromolecules is believed to provide the basis <strong>of</strong><br />

allergic responses, e.g. skin sensitisation reactions, to small molecules and,<br />

possibly, a basis for the induction <strong>of</strong> auto-immune responses. Thus, the<br />

binding <strong>of</strong> the small molecule transforms normal proteins into ‘foreign’<br />

proteins which trigger an immune response. Recently we have employed<br />

this principle in an attempt to direct the immune response specifically<br />

against mercapturic acid haptens by immunising mice with the haptens<br />

bound to a non-antigenic carrier protein, i.e. mouse serum albumin.<br />

Preliminary results indicate that this tactic has been successful. Overall the<br />

treatment induced fewer antibody-producing cells. However, the antibodies<br />

that were generated show high affinities and specificity toward model<br />

mercapturic acids including S-(2-hydroxyethyl) and S-phenylmercapturic<br />

acid. Studies are in progress to investigate the performance <strong>of</strong> these<br />

antibodies in an immunoenrichment mode.<br />

The preliminary results <strong>of</strong> our studies using non-antigenic protein<br />

carriers are very encouraging and have provided fresh insights which may<br />

assist in directing immune responses against the specific structural features<br />

<strong>of</strong> interest. Improvements in our ability to tailor the antibody will prove<br />

extremely valuable in optimising the properties <strong>of</strong> antibodies to meet<br />

specific needs, e.g. to enrich DNA, protein or mercapturic acid adducts for<br />

application in identifying the chemical initiators <strong>of</strong> human cancer and<br />

quantifying exposures to these agents.<br />

Cancer risk assessment<br />

Human exposure monitoring (determination <strong>of</strong> dose)<br />

The assessment <strong>of</strong> cancer risks posed by exposure to genotoxic chemicals<br />

has two components: determination <strong>of</strong> the dose and determination <strong>of</strong> the<br />

effect (increment in cancer incidence) caused by that dose. The introduction<br />

<strong>of</strong> the target dose concept by Ehrenberg in the early 1970s has provided the<br />

key to modern strategies to assess genotoxic risks (Ehrenberg, 1974, 1979;<br />

Ehrenberg et al., 1974). This new dose concept was developed to provide a<br />

measure <strong>of</strong> the critical dose, i.e. the dose <strong>of</strong> the ultimate genotoxic agent(s)<br />

penetrating to DNA. Target dose is much more relevant to risk assessment<br />

than is exposure dose. The determination <strong>of</strong> target dose automatically


compensates for individual or species differences in the operation <strong>of</strong><br />

metabolic and biokinetic factors that control the quantitative (and<br />

qualitative) relationships between the exposure and the dose <strong>of</strong> the ultimate<br />

toxicant delivered to the target. Measurements <strong>of</strong> target dose may be<br />

applied to improve the extrapolation <strong>of</strong> risk data from experimental<br />

models to humans and may also provide improved definitions <strong>of</strong> risks to<br />

individuals. The determination <strong>of</strong> target dose in humans may be viewed,<br />

therefore, as an approach towards direct risk monitoring as well as a more<br />

relevant approach to monitor human exposures to genotoxic chemicals.<br />

Determination <strong>of</strong> target dose<br />

The determination <strong>of</strong> target dose raises numerous technical and theoretical<br />

problems. Target dose can be determined by measuring primary products,<br />

e.g. DNA adducts, formed when genotoxic agents react with DNA. The<br />

kinetics <strong>of</strong> formation and decay <strong>of</strong> these adducts must also be determined<br />

(vide infra) in order to transform measurements <strong>of</strong> amounts <strong>of</strong> adducts into<br />

estimates <strong>of</strong> target dose. Human tissue DNA is not readily accessible for<br />

monitoring purposes: surrogate dose monitors are required. There are<br />

numerous possibilities including the determination <strong>of</strong> adducts in white<br />

blood cell DNA or <strong>of</strong> the corresponding adducts in the haemoglobin <strong>of</strong><br />

circulating erythrocytes.<br />

Such indirect approaches require validation. Haemoglobin is the most<br />

extensively studied surrogate, not only because <strong>of</strong> its accessibility and<br />

relative abundance but also because <strong>of</strong> the relative stability <strong>of</strong> haemoglobin<br />

adducts and the longevity <strong>of</strong> erythrocytes which permit retrospective<br />

estimates <strong>of</strong> dose received by the erythrocytes over a period <strong>of</strong> about 4<br />

months. Current evidence indicates that all electrophiles that undergo<br />

covalent reactions with DNA also react with haemoglobin. Furthermore<br />

the amounts <strong>of</strong> haemoglobin adducts are quantitatively related to the rates<br />

<strong>of</strong> formation <strong>of</strong> DNA adducts in the tissues. However the proportional<br />

relationships between the doses delivered to tissue DNA and to<br />

haemoglobin or to any other surrogate will vary from chemical to chemical<br />

and will have to be established using experimental models.<br />

Measurement <strong>of</strong> haemoglobin adducts<br />

A.S.WRIGHT ET AL. 189<br />

Genotoxic chemicals undergo covalent reactions with a variety <strong>of</strong><br />

nucleophilic centres in haemoglobin including the sulphydryl group <strong>of</strong><br />

cysteine, the N 1 and N 3 atoms <strong>of</strong> histidine and the amino groups <strong>of</strong> Nterminal<br />

valine residues. Ehrenberg’s group (Osterman-Golkar et al., 1976;<br />

Calleman et al, 1978; Törnqvist et al., 1986a) has pioneered the<br />

development <strong>of</strong> methods to detect, identify and quantify adducts formed at<br />

each <strong>of</strong> these centres. A review <strong>of</strong> these and methods for the analysis <strong>of</strong>


190 MOLECULAR APPROACHES TO ASSESS CANCER RISKS<br />

‘labile’ adducts formed, for example, during exposure to aromatic amines<br />

(Green et al., 1984; Albrecht and Neumann, 1985) is beyond the scope <strong>of</strong><br />

this paper (for reviews see Farmer, 1991; Skipper and Naylor, 1991).<br />

However, probably the most powerful and valuable approach was<br />

developed by Törnqvist et al. (1986a, b) who showed that adducts with the<br />

N-terminal valine residues <strong>of</strong> haemoglobin could be specifically enriched by<br />

scission in a modified Edman reaction followed by extraction. This<br />

enrichment procedure greatly facilitates sample analysis by GC/MS.<br />

Immunoassays are also being introduced as alternatives to physicochemical<br />

methods for the determination <strong>of</strong> protein adducts (Wraith et al., 1988).<br />

However, as in the case <strong>of</strong> DNA adducts, the biggest impact <strong>of</strong><br />

immunotechnology on the analysis <strong>of</strong> protein adducts will probably be in<br />

the immunoenrichment <strong>of</strong> low levels <strong>of</strong> adducts for analysis by physicochemical<br />

methods.<br />

Determination <strong>of</strong> biological effects<br />

Tumour incidence<br />

The determination <strong>of</strong> target dose is essential for assessing cancer risks<br />

posed by low-level exposures to genotoxic chemicals. The other requisite is<br />

know ledge <strong>of</strong> the human dose-carcinogenic response relationships in the<br />

low-dose range. The lack <strong>of</strong> intrinsic resolving power <strong>of</strong> classical<br />

epidemiological methods (vide supra) prevents effective applications to<br />

detect small carcinogenic effects associated with low exposures to any<br />

particular genotoxic chemical. Furthermore, the detection limits <strong>of</strong> animal<br />

cancer studies fall short <strong>of</strong> ‘acceptable’ risk limits by three to four orders <strong>of</strong><br />

magnitude (Wright, 1991). This poor sensitivity compels the use <strong>of</strong> high<br />

test doses in order to ensure that significant carcinogens do not go<br />

undetected. However, it is generally accepted that high doses <strong>of</strong> chemicals<br />

may induce tumours by non-specific mechanisms, e.g. via tissue injury and<br />

compensatory cell proliferation, that do not operate at low doses (Ames,<br />

1989; Wright, 1991). Many, if not all genotoxic chemicals induce cell<br />

injury at high (thresholded) doses. Clearly, extrapolation <strong>of</strong> such high dose<br />

risk data to the relevant low-dose range may, at the very least, lead to a<br />

gross overestimation <strong>of</strong> risk.<br />

Determination <strong>of</strong> mutagenic potency<br />

In considering the impact that a low-level exposure to a genotoxic<br />

chemical may have on cancer incidence, it is reasonable to suggest that the<br />

mutagenic propensity <strong>of</strong> the chemical, although <strong>of</strong> a low order, would<br />

nevertheless be the overriding risk factor. Thus, it is probable that any


A.S.WRIGHT ET AL. 191<br />

intrinsic promoter activity <strong>of</strong> the chemical arising, for example as a<br />

consequence <strong>of</strong> cell injury, would be negligible at low exposures—<br />

particularly when viewed in the context <strong>of</strong> the overall promoter pressure<br />

exerted on the populations at risk. Thus, it is unlikely that the added<br />

cancer risk would be greater than and may approximate to the increment in<br />

critical mutations caused by the specific exposure (Figure 14.1).<br />

Experimental studies indicate that all known categories <strong>of</strong> genotoxic<br />

agents ranging from methylating agents to polycyclic aromatic compounds<br />

can induce the critical mutations leading to malignancy (Figure 14.1).<br />

Furthermore, at low doses, the possibility that exposure to a particular<br />

genotoxic chemical would induce more than one <strong>of</strong> the critical mutations in<br />

any particular cell is extremely remote. Indeed it is probable that each<br />

critical mutation is induced by a different agent or mechanism, i.e.<br />

chemical, radiation or ‘spontaneous’. In this sense, each critical mutation<br />

would have equal status, i.e. no single event would be any more or any less<br />

critical than any other to the final outcome. Accordingly, the increment in<br />

cancer risk would equate with the increment in any decisive, e.g. oncogeneactivating,<br />

mutation in any critical gene. The induction <strong>of</strong> such mutations<br />

is almost certainly a direct function <strong>of</strong> overall mutagenic activity <strong>of</strong> the<br />

chemical, i.e. linked to the number <strong>of</strong> mutational events rather than the type<br />

<strong>of</strong> mutations induced by a given dose <strong>of</strong> the chemical. At low exposures,<br />

therefore, the increment in cancer incidence due to a specific genotoxic<br />

agent would approximate to the small increase in the total mutational load<br />

caused by the exposure, i.e. relative to the overall background level <strong>of</strong><br />

mutations due to all causes, multiplied by the overall cancer incidence in<br />

the population at risk. (The latter function introduces a measure <strong>of</strong> the net<br />

impact <strong>of</strong> promoter and anti-promoter pressure acting on initiated cells in<br />

the population at risk.) Of course risks may also be calculated on the basis<br />

<strong>of</strong> specific tumours and specific tissues.<br />

The determination <strong>of</strong> small increments in mutation associated with lowlevel<br />

exposures to genotoxic chemicals in human populations presents<br />

enormous technical problems not least due to the much larger and variable<br />

background <strong>of</strong> mutations due to all causes. Increasing the sensitivity <strong>of</strong><br />

mutation assays per se (vide supra) is unlikely to improve the situation.<br />

High resolving power is also needed to discriminate between effects due to<br />

different contributory factors. Nevertheless, while direct approaches to<br />

assess absolute cancer risks posed by such low-level exposures may elude<br />

us we can nevertheless begin to determine relative cancer risks and<br />

prioritise genotoxic chemicals on the basis <strong>of</strong> experimental determinations<br />

<strong>of</strong> mutagenic potency and estimates <strong>of</strong> target doses resulting from<br />

environmental or occupational exposures. Thus, according to the foregoing<br />

the relative cancer risk posed by a low exposure to a genotoxic agent<br />

would approximate to the number <strong>of</strong> mutations induced per unit target<br />

dose×estimated human target dose. Once relative cancer risks have been


192 MOLECULAR APPROACHES TO ASSESS CANCER RISKS<br />

established, the determination <strong>of</strong> the ‘absolute’ risks for one genotoxic<br />

chemical would permit calculation <strong>of</strong> the ‘absolute’ risks for the others.<br />

Such ‘absolute’ risks will nevertheless vary from population to population<br />

dependent upon variations in promoter pressure.<br />

Determination <strong>of</strong> ‘absolute’ cancer risks<br />

Increments in human mutation caused by low-level exposures to genotoxic<br />

chemicals are essential for risk estimation but cannot be determined<br />

directly (vide supra). Such increments may be estimated using experimental<br />

models. However, unless the variations in background are <strong>of</strong> a very low<br />

order, it is unlikely that even the most sensitive <strong>of</strong> the emerging mutation<br />

assays will permit the measurement <strong>of</strong> small increments <strong>of</strong> mutation at<br />

low, e.g. environmental, exposures. Extrapolation to low doses will be<br />

required and must necessarily be conservative, i.e. linear extrapolation to<br />

the origin.<br />

In addition to ‘high’ dose-low dose extrapolation, it will be necessary to<br />

apply corrections for differences between the model and humans in the<br />

operation <strong>of</strong> systemic factors that govern the relationships between<br />

exposure and mutagenic effect. Estimates <strong>of</strong> target dose in the human<br />

population at risk and in the experimental model compensate for<br />

differences in metabolic and biokinetic factors that determine the<br />

relationships between exposure and the critical dose. In effect, the<br />

determination <strong>of</strong> target dose provides a measure <strong>of</strong> the rates <strong>of</strong> formation <strong>of</strong><br />

the key (primary and critical) chemical lesions leading to mutation. The<br />

final stage in translating the experimentally-determined risk data to<br />

humans is to apply corrections for systemic factors that determine the<br />

progression <strong>of</strong> the key lesions into mutations.<br />

The equivalent radiation dose concept<br />

The principal systemic factors determining the progression <strong>of</strong> key chemical<br />

lesions in DNA into mutations are the rates and fidelities <strong>of</strong> DNA repair<br />

and replication (Wright et al., 1988). Ehrenberg and co-workers have<br />

suggested that the repair <strong>of</strong> primary DNA damage induced by low doses <strong>of</strong><br />

radiation may be proportionate to that induced by low doses <strong>of</strong> genotoxic<br />

chemicals. They have further suggested that the determination <strong>of</strong> the<br />

relative mutagenic effectiveness or potencies <strong>of</strong> radiation and any<br />

particular genotoxic chemical may be <strong>of</strong> value in correcting for species<br />

differences in factors determining the progression <strong>of</strong> primary DNA damage<br />

into mutations. The model proposed by Ehrenberg (1980) is based on the<br />

determination <strong>of</strong> the dose-response curves for the induction <strong>of</strong> the same<br />

mutation in the same experimental system by low target doses <strong>of</strong> the test<br />

chemical and acute -radiation. A consistent ratio between the two curves,


A.S.WRIGHT ET AL. 193<br />

which need not be linear, would indicate proportionality over the low dose<br />

range <strong>of</strong> interest and would permit the mutagenic potency <strong>of</strong> the chemical<br />

to be expressed in terms <strong>of</strong> radiation equivalents, i.e. the number <strong>of</strong> rads<br />

giving the same response or risk as a unit <strong>of</strong> chemical dose (expressed in<br />

terms <strong>of</strong> target dose, e.g. millimolar hour, mM h). The significance <strong>of</strong> such<br />

radiation dose-equivalents hinges on their possible extrapolative value.<br />

Thus, in order to be useful in assisting the translation <strong>of</strong> experimentallydetermined<br />

mutagenicity data, the rad-equivalence value for the test<br />

chemical must have a similar numerical value in both the test system used<br />

to determine mutagenicity and in humans. However, it is improbable that<br />

rad-equivalence values can be directly determined in humans.<br />

Rad-equivalence values for the induction <strong>of</strong> mutations have been<br />

determined for a number <strong>of</strong> intrinsically reactive mon<strong>of</strong>unctional alkylating<br />

agents using a wide range <strong>of</strong> genetic endpoints in a variety <strong>of</strong> biological<br />

systems including bacteria, plants and mammalian species—the latter,<br />

mainly in vitro (Ehrenberg et al., 1974; Ehrenberg, 1976, 1979; Calleman,<br />

1984; Kolman et al., 1989). The rad-equivalence value for a given<br />

alkylating agent was approximately the same (within a factor <strong>of</strong> two) in<br />

each <strong>of</strong> the test systems. On the basis <strong>of</strong> such evidence Calleman et al.<br />

(1978) concluded that there was no reason to presume that a value for radequivalence<br />

established in these disparate systems would differ in humans.<br />

The best studied example is ethylene oxide. Currently a conjoint programme<br />

is underway at the Universities <strong>of</strong> Stockholm and Leiden to determine radequivalence<br />

values in rodents in vivo using a variety <strong>of</strong> endpoints including<br />

the clonal HGPRT mutation assay and induction <strong>of</strong> pre-neoplastic nodules<br />

in rat liver. Preliminary findings are encouraging (Ehrenberg, personal<br />

communication).<br />

Demonstration <strong>of</strong> their extrapolative value would justify application <strong>of</strong><br />

rad-equivalence values to compute small increments in mutation induced in<br />

humans by low exposures (determined as target dose) to genotoxic<br />

chemicals. The determination <strong>of</strong> these increments is the basis <strong>of</strong> the risk<br />

model (vide supra) in which the increment in cancer risk due to a particular<br />

chemical in a population is viewed as approximating to the increment in<br />

mutation induced by the chemical. However, the fact that risk coefficients<br />

have not yet been established for radiation-induced mutations in humans<br />

precludes applications <strong>of</strong> rad-equivalence values to estimate mutational<br />

risks, e.g. small increments in mutation, in humans. Of course, Ehrenberg<br />

realised that a genotoxic chemical(s) may prove to be superior to radiation<br />

as a reference standard for estimating mutational risks. Indeed, the use <strong>of</strong><br />

chemicals that are representative <strong>of</strong> classes <strong>of</strong> genotoxic chemicals, repair<br />

pathways, etc. is envisaged in this developing ‘equivalence’ strategy<br />

(Törnqvist and Osterman-Golkar, 1991). However, at the time the original<br />

strategy was formulated there was and still is no reliable data relating low<br />

level exposure to a genotoxic chemical and the attendant risk <strong>of</strong> mutation


194 MOLECULAR APPROACHES TO ASSESS CANCER RISKS<br />

or cancer in human populations. In contrast, risk coefficients have been<br />

established for the induction <strong>of</strong> cancer by low levels <strong>of</strong> -radiation in<br />

human populations within, approximately, a factor <strong>of</strong> 2.<br />

According to the basic presumptions (vide supra), the increased cancer<br />

risks in a human population caused by low-level exposures to radiation or<br />

to a genotoxic chemical are predominantly due to the mutagenic<br />

propensities <strong>of</strong> these genotoxic factors. It would, therefore, seem<br />

reasonable to suggest that cells that had been initiated by exposure to<br />

radiation or to genotoxic chemicals or combinations <strong>of</strong> factors (vide supra)<br />

would, nevertheless, be subject to the same general promoter and<br />

modulating influences acting on the population. On this basis,<br />

experimentally-determined rad-equivalence values for genotoxic chemicals<br />

may be used to convert target doses <strong>of</strong> the chemicals (determined in human<br />

populations receiving low-level exposures) into the equivalent doses <strong>of</strong><br />

radiation (for the induction <strong>of</strong> mutation). The respective cancer risks<br />

associated with any particular monitored target dose <strong>of</strong> a genotoxic<br />

chemical may then be obtained by direct reference to the human cancer risk<br />

coefficients established for radiation.<br />

References<br />

ALBRECHT, W. and NEUMANN, H.-G., 1985, Biomonitoring <strong>of</strong> aniline and<br />

nitrobenzene. Haemoglobin binding in rats and analysis <strong>of</strong> adducts, Arch.<br />

Toxicol, 57, 1–5.<br />

AMES, B.N., 1989, Environmental pollution and the causes <strong>of</strong> human cancer: six<br />

errors, in DeVita, V.T., Jr, Hellman, S. and Rosenberg, S.A. (Eds) Important<br />

Advances in Oncology, pp. 237–47, Philadelphia, PA: Lippincott.<br />

AMES, B.N., DURSTON, W.E., YAMASAKI, E. and LEE, F.E., 1973, Carcinogens<br />

are mutagens: a simple test system combining liver homogenates for activation<br />

and bacteria for detection, Proc. Nat. Acad. Sci. USA, 70, 2281–5.<br />

CALLEMAN, C.J., 1984, Haemoglobin as a dose monitor and its application to<br />

the risk estimation <strong>of</strong> ethylene oxide, PhD Thesis, p. 25, Stockholm: University<br />

<strong>of</strong> Stockholm.<br />

CALLEMAN, C.J., EHRENBERG, L., JANSSON, B., OSTERMAN-GOLKAR, S.,<br />

SEGERBÄCK, D., SVENSSON, K. and WACHTMEISTER, C.A., 1978,<br />

Monitor ing and risk assessment by means <strong>of</strong> alkyl groups in haemoglobin in<br />

persons occupationally exposed to ethylene oxide, J. Environm. Pathol. Tox.,<br />

2, 427–42.<br />

COOPER, D.P., GRIFFIN, K.A. and POVEY, A.C., 1992, Immunoaffinity<br />

purification combined with 32 P-postlabelling for the detection <strong>of</strong> O 6 -<br />

methylguanine in DNA from human tissues, Carcinogenesis, 13, 469–75.<br />

EHRENBERG, L., 1974, Genotoxicity <strong>of</strong> environmental chemicals, Acta. Biol.<br />

Yugosl., Ser. F Genetika, 6, 367–98.<br />

EHRENBERG, L., 1976, Methods <strong>of</strong> Comparing Effects <strong>of</strong> Radiation and<br />

Chemicals, Brighton IAEA Consultant Meeting.


A.S.WRIGHT ET AL. 195<br />

EHRENBERG, L., 1979, Risk assessment <strong>of</strong> ethylene oxide and other compounds,<br />

in McElheny, V.K. and Abrahamson, S. (Eds) Assessing Chemical Mutagens:<br />

The Risk to Humans (Banbury Report 1), pp. 157–90, Cold Spring Harbour,<br />

New York, CSH Press.<br />

EHRENBERG, L., 1980, Purposes and methods <strong>of</strong> comparing risks <strong>of</strong> radiation<br />

and chemicals, in Radiobiological Equivalents <strong>of</strong> Chemical Pollutants, pp. 11–<br />

36, Vienna: International Atomic Energy Agency.<br />

EHRENBERG, L., HIESCHE, K.D., OSTERMAN-GOLKAR, S. and WENNBERG,<br />

I., 1974, Evaluation <strong>of</strong> genetic risks <strong>of</strong> alkylating agents: tissue doses in the<br />

mouse from air contaminated with ethylene oxide, Mutat. Res., 24, 83–103.<br />

EHRENBERG, L. and OSTERMAN-GOLKAR, S., 1980, Alkylation <strong>of</strong><br />

macromolecules for detecting mutagenic agents, Teratogen., Carcinogen.<br />

Mutagen., 1, 105–27.<br />

FARMER, P.B., 1991, Analytical approaches for the determination <strong>of</strong><br />

proteincarcinogen adducts using mass spectrometry, in Groopman, J.D. and<br />

Skipper, P. L. (Eds) Molecular Dosimetry and Human Cancer: Analytical,<br />

Epidemiological and Social Considerations, pp. 189–210, Boca Raton: CRC<br />

Press.<br />

GREEN, L.C., SKIPPER, P.L., TURESKY, R.J., BRYANT, M.S. and<br />

TANNENBAUM, S.R., 1984, In vivo dosimetry <strong>of</strong> 4-aminobiphenyl in rats via<br />

a cysteine adduct in haemoglobin, Cancer Res., 44, 4254–9.<br />

KOLMAN, A., NASLUND, M., OSTERMAN-GOLKAR, S., SCALIA-TOMBA,<br />

G.P. and MEYER, A.L., 1989, Comparative studies <strong>of</strong> in vitro transformation<br />

by ethylene oxide and gamma-radiation <strong>of</strong> cells, Mutagenesis, 4, 58–61.<br />

Lu, A-L. and Hsu, I-C., 1992, Detection <strong>of</strong> single DNA base mutations with<br />

mismatch repair enzymes, Genomics, 14, 249–55.<br />

OSTERMAN-GOLKAR, S., EHRENBERG, L., SEGERBÄCK, D. and<br />

HALLSTROM, I., 1976, Evaluation <strong>of</strong> genetic risks <strong>of</strong> alkylating agents. II.<br />

Haemoglobin as a dose monitor, Mutat. Res., 34, 1–10.<br />

RANDERATH, K., REDDY, M.V. and GUPTA, R.C., 1981, 32 P-labeling test for<br />

DNA damage, Proc. Natl Acad. Sci. USA, 78, 6126–9.<br />

SKIPPER, P. L and NAYLOR, S., 1991, Mass spectrometric analysis <strong>of</strong><br />

proteincarcinogen adducts, in Garner, R.C, Farmer, P.B., Steel, G.T. and<br />

Wright, A.S. (Eds) Human Carcinogen Exposure: Biomonitoring and Risk<br />

Assessment, pp. 61–8, Oxford: Oxford University Press.<br />

THILLY, W.G., 1991, Mutational spectrometry: opportunities and limitations in<br />

human risk assessment, in Garner, R.C, Farmer, P.B., Steel, G.T. and Wright,<br />

A. S. (Eds) Human Carcinogen Exposure: Biomonitoring and Risk<br />

Assessment, pp. 127–33, Oxford: Oxford University Press.<br />

TÖRNQVIST, M. and OSTERMAN-GOLKAR, S., 1991, Monitoring <strong>of</strong> in vivo<br />

dose by macromolecular adducts: usefulness in risk estimation, in Groopman,<br />

J.D. and Skipper, P.L. (Eds) Molecular Dosimetry and Human Cancer:<br />

Analytical, Epidemi ological and Social Considerations, pp. 89–102, Boca<br />

Raton: CRC Press.<br />

TÖRNQVIST, M., MOWRER, J., JENSEN, S. and EHRENBERG, L., 1986a,<br />

Monitoring <strong>of</strong> environmental cancer initiators through haemoglobin adducts<br />

by a modified Edman degradation method, Analyt. Biochem., 154, 255–66.


196 MOLECULAR APPROACHES TO ASSESS CANCER RISKS<br />

TÖRNQVIST, M., OSTERMAN-GOLKAR, S., KAUTIAINEN, A., JENSEN, S.,<br />

FARMER P.B. and EHRENBERG, L., 1986b, Tissue doses <strong>of</strong> ethylene oxide<br />

in cigarette smokers determined from adduct levels in haemoglobin,<br />

Carcinogenesis, 7, 1519–21.<br />

WRAITH, M.J., WATSON, W.P., EADSFORTH, C.V., VAN SITTERT, N.J.,<br />

TÖRNQVIST, M. and WRIGHT, A.S., 1988, An immunoassay for monitoring<br />

human exposure to ethylene oxide in Bartsch, H., Hemminki, K., and O’Neill,<br />

I.K. (Eds) Methods for Detecting DNA Damaging Agents in Humans:<br />

Applications in Cancer Epidemiology and Prevention, IARC Scientific<br />

Publications No. 89, pp. 271–4, Lyon, France: International Agency for<br />

Research on Cancer.<br />

WRIGHT, A.S., 1991, Emerging strategies for the determination <strong>of</strong> human<br />

carcinogens, detection, identification, exposure monitoring and risk<br />

evaluation, in Garner, R.C, Farmer, P.B., Steel, G.T. and Wright, A.S. (Eds)<br />

Human Carcinogen Exposure: Biomonitoring and Risk Assessment, pp. 3–23,<br />

Oxford: Oxford University Press.<br />

WRIGHT, A.S., BRADSHAW, T.K. and WATSON, W.P., 1988, Prospective<br />

detection and assessment <strong>of</strong> genotoxic hazards: a critical appreciation <strong>of</strong> the<br />

contribution <strong>of</strong> L.G.Ehrenberg, in Bartsch, H., Hemminki, K. and O’Neill, I.K.<br />

(Eds) Methods for Detecting DNA Damaging Agents in Humans: Applications<br />

in Cancer Epidemiology and Prevention, IARC Scientific Publications No. 89,<br />

pp. 237–47, Lyon, France: International Agency for Research on Cancer.


15<br />

Evaluation <strong>of</strong> Toxicity to the Immune System<br />

HANS-WERNER VOHR<br />

Bayer AG, Wuppertal<br />

Introduction<br />

A number <strong>of</strong> years ago the new field <strong>of</strong> immunotoxicology was established.<br />

Very early on, calls were heard from various sides demanding that the<br />

development <strong>of</strong> new chemicals should also take into account the influence<br />

<strong>of</strong> these substances on the immune system (Dean, 1979; Luster et al., 1988;<br />

Trizio, et al., 1988). These demands led on the one hand to the initiation <strong>of</strong><br />

a number <strong>of</strong> investigations and collaborative studies (ICICIS; BGA; US-<br />

NTP), on the other hand to thoughts by the authorities and industry on the<br />

introduction <strong>of</strong> guidelines (US-EPA, 1982, 1990; Sjoblad, 1988; ECETOC,<br />

1990; UK-DOH, 1991; Hinton, 1992; OECD, 1992ab.<br />

If we define immunotoxicology as the science <strong>of</strong> adverse effects <strong>of</strong><br />

substances on the immune system we can say further that these side-effects<br />

can lead to either immunopotentiation or immunosuppression. The former<br />

can lead to induction <strong>of</strong> autoimmune reactions and to Type I-IV<br />

hypersensitivity reactions, the latter to reduced resistance to infection,<br />

development <strong>of</strong> cancer and also to autoimmune phenomena.<br />

On the basis <strong>of</strong> this definition, immunotoxicological investigations have<br />

already been carried out for years during the development <strong>of</strong> substances;<br />

namely with respect to DTH reactions (Type IV) in the guinea pig (Bühler,<br />

1965; Magnusson and Kligman, 1969). As an alternative to these tests in<br />

the guinea pig, the so-called local lymph node assay (LLNA) in the mouse<br />

according to Kimber et al. (1989) was developed and validated and has<br />

meanwhile been adopted as alternative test in the OECD guidelines (OECD,<br />

1992a, b; Botham et al., 1991).<br />

The development or selection <strong>of</strong> suitable tests for immunotoxicological<br />

screening and thus for incorporation in guidelines presents considerable<br />

problems. Most <strong>of</strong> the tests which have been proposed for<br />

immunotoxicological investigations and most knowledge and experience in<br />

immunology are based on mouse models. The standard animal in the early<br />

phase <strong>of</strong> toxicological testing, however, is the rat. Transference <strong>of</strong> the tests<br />

is not always easy, partly because <strong>of</strong> lack <strong>of</strong> suitable reagents.


198 EVALUATION OF TOXICITY TO THE IMMUNE SYSTEM<br />

The next problem is to find which tests can suitably be used—simply and<br />

without having to treat additional animals—for reliable identification <strong>of</strong><br />

interactions with the immune system. Another question which has not yet<br />

been solved is relating to the dosages and the changes in immunological<br />

parameters which are still tolerable and at which times these should be<br />

determined.<br />

National and international collaborative studies<br />

Most <strong>of</strong> the guideline drafts favour a two-tiered to three-tiered approach<br />

for the screening <strong>of</strong> immunotoxic side effects, with the greatest disparities<br />

in respect <strong>of</strong> the proposals for tier I tests, ranging from just organ weights<br />

and a little more emphasis on the histology <strong>of</strong> the lymphatic organs, to a<br />

series <strong>of</strong> elaborate, supplementary function tests including, in some cases,<br />

satellite groups.<br />

Any discussion about basic tests is hampered by a lack <strong>of</strong> data from<br />

routine toxicological and/or epidemiological studies although a few years<br />

ago a number <strong>of</strong> collaborative studies were initiated, namely: ICICIS<br />

(international), US-NTP (international, USA (two)), BGA (Federal German<br />

Health Office, Germany), GEVI (France). The aim <strong>of</strong> all these studies is to<br />

check on different histological parameters and functional tests for their<br />

possibility to flag an immunotoxic potential <strong>of</strong> a compound in a routine 14<br />

or 28 day study (rats) in an interlaboratory trial. Two immunosuppressive<br />

standards (azathioprine and/or cyclosporin A) have been used so far.<br />

Although the experimental phase is finished the evaluation <strong>of</strong> the data is<br />

still underway.<br />

Table 15.1 summarises the immunotoxicological experiments and<br />

collaborative studies currently in progress. With a few exceptions all<br />

investigations are based on a 28-day gavage study in rats. These basic tests<br />

were supplemented by extended histopathology and functional tests.<br />

ICICIS<br />

The first substance to be investigated in the international collaborative<br />

study was azathioprine. However this first attempt suffered from lack <strong>of</strong><br />

harmonisation between the models used by the 28 participants world-wide.<br />

This made the comparability and the evaluation <strong>of</strong> the results particularly<br />

problematic.<br />

The second substance which was tested by ICICIS in a more restrictive<br />

design with slightly fewer participants was cyclosporin A. The<br />

experimental section as well as the first evaluation <strong>of</strong> the results has now<br />

been completed.<br />

As the final evaluation—including statistics—<strong>of</strong> ICICIS is likely to take<br />

some time (years?) it will not be discussed further at this point.


Table 15.1 Immunotoxicology collaborative studies<br />

Fischer 344 study (Kimber-White)<br />

H.-W.VOHR 199<br />

An other interesting approach was done by Kimber-White and colleagues.<br />

In this study the duration <strong>of</strong> treatment was 14 days, Fischer 344 rats were<br />

used as experimental animals. Apart from the usual parameters the plaque<br />

assay (PFCA), mitogenic stimulation (ConA, LPS) and NK activity were<br />

measured.


200 EVALUATION OF TOXICITY TO THE IMMUNE SYSTEM<br />

No effects were found with respect to organ weights or cell counts<br />

analysis but there were marked effects in the PFCA. Here there was a good<br />

correlation between the laboratories and clearly dose-dependent effects<br />

were seen. Mitogenic stimulation was not and NK activity only slightly<br />

affected.<br />

Results <strong>of</strong> the BGA collaborative study<br />

In order to put the discussion on a somewhat sounder footing it is<br />

imperative to test the various models for detection <strong>of</strong> immunotoxicological<br />

potential in practice. For this reason Bayer AG is taking part in a<br />

collaborative trial initiated by the German Federal Health Office in Berlin<br />

(BGA study). In this collaborative study standard immunotoxic substances<br />

are investigated in parallel under restrictive conditions in several<br />

laboratories. On the other hand Bayer AG has introduced a set <strong>of</strong><br />

functional immunological tests into the routine toxicological testing <strong>of</strong><br />

agrochemicals in rats in order to test the informative value <strong>of</strong> these<br />

parameters in practice (Vohr, 1995).<br />

In the course <strong>of</strong> this collaborative study cyclosporin A was investigated<br />

as the first substance in a very well harmonised design. One reason for<br />

choosing cyclosporin A was to permit comparison with the ICICIS results.<br />

The study was based on OECD guideline 407 which was supplemented by<br />

a number <strong>of</strong> histopathological, haematological, clinical-chemical and<br />

functional parameters. The final evaluation, including pathology and<br />

statistics will take a few more months. Nevertheless a first look at the data<br />

showed that there were no marked effects with respect to either organ<br />

weights (lymphatic organs) or blood cells. On the other hand dose-related<br />

effects were found for many parameters in the functional tests.<br />

Examples are shown <strong>of</strong> these parameters and their changes at the various<br />

dosages. Both sexes show dose-related changes from the lowest dosage (1<br />

mg kg −1 ) upwards with respect to the surface markers <strong>of</strong> the<br />

immunocompetent cells. The PFCA and the measurements <strong>of</strong> IgA<br />

antibodies in serum also proved to be sensitive parameters.<br />

In immunotoxic investigations based on data obtained in rats treated<br />

with test substances (28-day test) we found that secondary influences <strong>of</strong>ten<br />

occur and that occasionally one single parameter is changed at the highest<br />

dosage. Apart from these non-specific effects, dose-related reactions were<br />

observed from the lowest or middle dose upwards (for example in the case<br />

<strong>of</strong> cytostatic substances). Such genuinely immunotoxic compounds were<br />

reliably identified by surface markers (like CD4/CD45R or PanB) and<br />

changes in the serum Ig-titres. Changes in other parameters such as cell<br />

counts and macrophage activity verified these findings.


Findings <strong>of</strong> the US-NTP study (Luster)<br />

The US-NTP study investigated 51 substances, 35 <strong>of</strong> which were declared<br />

immunotoxic, in a comprehensive test battery in mice for changes in<br />

functional parameters after 28-day administration <strong>of</strong> the substances. The<br />

correlations <strong>of</strong> each <strong>of</strong> these parameters with the given classification and<br />

with the results <strong>of</strong> host-resistance studies were calculated. The correlations<br />

after combinations <strong>of</strong> individual tests were also calculated (Luster et al.,<br />

1992, 1993).<br />

The conclusion drawn from these investigations was that the<br />

immunotoxic potential <strong>of</strong> a substance can be determined relatively reliably<br />

by combination <strong>of</strong> 2–3 specific tests. The most powerful tests proposed by<br />

Luster et al. for such a combination include surface markers, NK test and<br />

PFCA. Serum titres <strong>of</strong> Ig —particularly IgA—were unfortunately not<br />

determined.<br />

With regard to the correlation with host resistance (HR) results it was<br />

found that if effects were shown in the HR model there were always effects<br />

on functional parameters, too. There were, however, also cases in which<br />

there were effects in the functional tests although the HR studies were<br />

negative. Although these investigations were carried out on mice and the<br />

choice and classification <strong>of</strong> the substances are not entirely undisputed,<br />

these findings are nevertheless confirmed by our own experience.<br />

Discussion and prospect<br />

H.-W.VOHR 201<br />

It is undoubtedly too early to make any judgement. However, it appears<br />

that apart from the histology—particularly the immuno-histology—a few<br />

additional parameters such as analysis <strong>of</strong> surface markers <strong>of</strong> subpopulations<br />

and serum titre assays <strong>of</strong> IgG, IgM and IgA are sufficient as screening<br />

indicators to show the possible immunotoxic potential <strong>of</strong> a substance. One<br />

<strong>of</strong> these is the PFCA, which presupposes, however, that satellite groups are<br />

used or that the authorities accept injection <strong>of</strong> SRBC as GLP treatment.<br />

Positive findings in a combination <strong>of</strong> these tests should then occasion<br />

further elucidation <strong>of</strong> the immunotoxic potential.<br />

In summary it can be concluded from the currently available results <strong>of</strong> the<br />

collaborative studies that the following criteria must be fulfilled if a<br />

substance is to be characterised as possibly immunotoxic. The substance<br />

must:<br />

1. Induce significant dose-related changes in one <strong>of</strong> the effective tests<br />

listed above, or<br />

2. induce significant changes in the highest dose group in a combination<br />

<strong>of</strong> 2–3 <strong>of</strong> these tests.


202 EVALUATION OF TOXICITY TO THE IMMUNE SYSTEM<br />

But also findings in lymphoid organs which are remarkable with respect to<br />

quality, severity or quantity <strong>of</strong> changes should be <strong>of</strong> sufficient relevance to<br />

warrant further assessment.<br />

Finally I would like to point out the problems involved in the evaluation<br />

<strong>of</strong> immunological changes.<br />

• In contrast to other data obtained in routine toxicology, historical data<br />

and routine experience with respect to the functional tests are so far<br />

almost entirely lacking. On account <strong>of</strong> this paucity <strong>of</strong> data a decision as<br />

to the right effect level for the functional tests can only be made on the<br />

basis <strong>of</strong> subjective judgement at present. The discussion <strong>of</strong> uniform<br />

criteria is only just beginning.<br />

• If effects only occur secondarily, e.g. on account <strong>of</strong> inflammatory<br />

processes, they are not specifically immunotoxic. It must be discussed<br />

whether such effects on the immune system, which is only doing its<br />

normal job in these cases, can be classed as immunotoxic at all.<br />

• The immune system can show an ‘oscillating’ response to a substance. A<br />

substance may have a stimulating effect in a low dose range and an<br />

inhibitory effect in a high range or vice versa. Such reactions are also<br />

dose-related effects.<br />

• Most immunotoxic investigations have so far used known<br />

immunosuppressive drugs. There are, however, little data—particularly<br />

from rat studies— on immunostimulant substances. Since the other<br />

unwanted side-effect apart from immunosuppression is immune<br />

potentiation, future collaborative investigations must urgently<br />

concentrate on such substances. Before this, no final evaluation and thus<br />

no recommendations on relevant tests can be made.<br />

For pharmaceuticals (Hinton, 1992), pesticides (US-EPA, 1990, 1993) and<br />

veterinary medicinal chemicals (EEC, 1991) final drafts or notes for<br />

guidance for the screening <strong>of</strong> the immunotoxic potential <strong>of</strong> a compound<br />

already exist. These draft proposals for immunotoxicity parameters for<br />

incorporation into new guidelines are shown in Tables 15.2 (FDA) and<br />

15.3 (US-EPA).<br />

For industrial chemicals advanced screening <strong>of</strong> lymphoid organs also<br />

with respect to functional parameters had been expected to be incorporated<br />

into the adopted OECD guideline No. 407 (1992). But recommendations<br />

made by van Loveren and Vos (1992) have not yet been taken into<br />

consideration for this update <strong>of</strong> OECD guideline 407. The proposal <strong>of</strong><br />

these authors recommended more histopathology (gut associated lymphoid<br />

tissue), measurement <strong>of</strong> serum immunoglobulins, bone marrow cellularity,<br />

cyt<strong>of</strong>luorimetry <strong>of</strong> spleen cells and measurement <strong>of</strong> NK cell activity.<br />

A task force ‘Immunotoxicology’ initiated by ECETOC has put forward<br />

proposals on hazard identification and risk assessment <strong>of</strong> immunotoxic


Table 15.2 Summary <strong>of</strong> immunotoxicity testing recommendations for direct food additives<br />

H.-W.VOHR 203<br />

Abbreviations: CBC=complete blood cell count; WBC=white blood cell count; Ig=Immunoglobin; NK=natural killer;<br />

IL-2=interleukin 2; SRBC=sheep red blood cells; TNP-LPS=trinitrophenol lipopolysaccharide.<br />

* Recommended for inclusion in basic testing.<br />

potential <strong>of</strong> a compound on the basis <strong>of</strong> the routine 28 day treatment <strong>of</strong><br />

rats (Basketter et al., 1994; ECETOC, 1994). A central point in these


204 EVALUATION OF TOXICITY TO THE IMMUNE SYSTEM<br />

Table 15.3 Proposed amendment to the subdivision F guideline requirements to<br />

provide for an evaluation <strong>of</strong> the immunotoxicity <strong>of</strong> chemical pesticides<br />

proposals is a flow diagram for the evaluation <strong>of</strong> results from this basic<br />

study for hazard identification and conclusions drawn from it. This flow<br />

diagram, shown in Figure 15.1, could be helpful for the evaluation <strong>of</strong><br />

results obtained from incorporation <strong>of</strong> a basic immunotoxicity test battery<br />

into studies with routinely treated animals.<br />

References<br />

BASKETTER, D. et al., 1994, The identification with sensitising or<br />

immunosuppressive properties in routine toxicology, Food Chem. Toxicol. (in<br />

press).<br />

BOTHAM, P.A. et al., 1991, Skin sensitization—a critical review <strong>of</strong> predictive test<br />

methods in animals and men Food Chem. Toxicol, 29, 275–86.<br />

BÜHLER, E.V., 1965, Delayed contact hypersensitivity in the guinea pig, Arch.<br />

Dermatol. 91, 171.<br />

DEAN, J.H., PADARATHSINGH, M.L. and JERRELS, T.R., 1979, Assessment <strong>of</strong><br />

immunobiological effects induced by chemicals, drugs or food additives. I. Tier<br />

testing and screening approach, Drug Chem. Toxicol. 2, 5–17.<br />

ECETOC, 1990, Skin Sensitization Testing, Monograph No. 14, Brussels.


Figure 15.1 Flow diagram (taken from the ECETOC monograph:<br />

Immunotoxicity).<br />

H.-W.VOHR 205<br />

ECETOC, 1994, Immunotoxicity: hazard identification and risk assessment,<br />

Monograph No. 21, Brussels.<br />

EEC, 1991, Annex to directive 81/852/EEC—Note <strong>of</strong> guidance, 22.


206 EVALUATION OF TOXICITY TO THE IMMUNE SYSTEM<br />

HINTON, D.M., 1992, Testing guidelines for evaluation <strong>of</strong> the immunotoxic<br />

potential <strong>of</strong> direct food additives, Crit. Rev. Food Sci. Nutrit, 32, 173–90.<br />

KIMBER, I., HILTON, J. and WEISENBERGER, C. 1989, The murine local lymph<br />

node assay for identification <strong>of</strong> contact allergens: a preliminary evaluation <strong>of</strong><br />

in situ measurement <strong>of</strong> lymphocyte proliferation, Contact Dermatit. 21, 215–<br />

20.<br />

LUSTER, M.I. et al., 1988, Development <strong>of</strong> a battery to assess chemical-induced<br />

immunotoxicity: National <strong>Toxicology</strong> Program’s guidelines for<br />

immunotoxicity evaluation in mice, Fundam. Appl. Toxicol. 10, 2–19.<br />

LUSTER, M.I. et al., 1992, Risk assessment in immuno-toxicology. I. Sensitivity<br />

and predictability <strong>of</strong> immune tests, Fundam. Appl. Toxicol., 18, 200–10.<br />

LUSTER, M.I. et al., 1993, Risk assessment in immuno-toxicology. II. Relationship<br />

between immune and host resistance tests, Fundam. Appl. Toxicol., 21, 71–82.<br />

MAGNUSSON, B. and KLIGMANN, A.M., 1969, The identification <strong>of</strong> contact<br />

allergens by animal assay. The guinea pig maximisation test, J. Invest.<br />

Dermatol., 52, 268.<br />

OECD, 1992a, Organisation for Economic Cooperation and Development,<br />

Guidelines for testing <strong>of</strong> chemicals No. 407, adopted 12 July, 1992.<br />

OECD, 1992b, Organisation for Economic Cooperation and Development,<br />

Guidelines for testing <strong>of</strong> chemicals—skin sensitisation, No. 406, adopted 17<br />

July, 1992.<br />

SJOBLAD, R., 1988, Potential future requirements for immunotoxicology testing<br />

<strong>of</strong> pesticides, Toxicol. Indust. Hlth, 4, 391.<br />

TRIZIO, D. et al., 1988, Identification <strong>of</strong> immunotoxic effects <strong>of</strong> chemicals and<br />

assessment <strong>of</strong> their relevance to man. Food Chem. Toxic., 26, 527–39.<br />

UK Department <strong>of</strong> Health, 1991, Proposed to update OECD Guideline 407.<br />

US-EPA, 1982, Code <strong>of</strong> Federal Regulations, Washington DC, 152–18, 152–24 and<br />

158–165.<br />

US-EPA, 1990, Draft immunotoxicity study screen for testing chemical pesticides.<br />

VAN LOVEREN, H. and Vos, J.G., 1992, Evaluation <strong>of</strong> OECD Guideline 407 for<br />

assessment <strong>of</strong> toxicity <strong>of</strong> chemicals with respect to potential adverse effects to<br />

the immune system. RIVM Report No. 158801001, Bilthoven: National<br />

Institute <strong>of</strong> Public Health and Environmental Protection.<br />

VOHR, H.-W., 1995, Experiences with an advanced screening procedure for the<br />

identification <strong>of</strong> chemicals with an immunotoxic potential in routine<br />

toxicology (a position paper). <strong>Toxicology</strong>, (in press),


16<br />

New Strategies: the Use <strong>of</strong> Long-term Cultures <strong>of</strong><br />

Hepatocytes in Toxicity Testing and Metabolism<br />

Studies <strong>of</strong> Chemical Products Other than<br />

Pharmaceuticals<br />

VERA ROGIERS, 1 MAY AKRAWI, 2 SANDRA COECKE, 1<br />

YVES VANDENBERGHE, 1 ELIZABETH SHEPHARD, 2 IAN<br />

PHILLIPS 3 and ANTOINE VERCRUYSSE 1<br />

1 Vrije Universiteit Brussel, Brussels; 2 University College<br />

London, London; 3 University <strong>of</strong> London, London<br />

Introduction: metabolism and toxicity <strong>of</strong> chemical<br />

products are closely linked<br />

Lipophilic compounds are metabolized in the liver by phase 1 and phase 2<br />

reactions into more polar, more hydrophilic metabolites, which are usually<br />

less biologically active. Bioactivation, however, may occur, forming toxic<br />

species by phase 1, cytochrome P450 (CYP) dependent oxidation (e.g.<br />

epoxidation <strong>of</strong> C=C to reactive epoxide intermediates (Guengerich et al.<br />

1991), CYP dependent reduction (e.g. dehalogenation <strong>of</strong> CCl 4 toa free<br />

radical intermediate) (Timbrell, 1993) or even by phase 2 reactions (e.g.<br />

reactive episulphonium ion formation by glutathione conjugation <strong>of</strong><br />

dibromoethane) (Van Bladeren, 1988; Coles and Ketterer, 1990; Timbrell,<br />

1993).<br />

The major process involved in the bioactivation <strong>of</strong> chemical carcinogens<br />

is their oxidation catalyzed by CYP enzymes. Thirty or more different CYP<br />

forms exist within each animal species (Nebert et al., 1989), each with at<br />

least some distinct elements <strong>of</strong> catalytic specificity and regulation. The role<br />

<strong>of</strong> some <strong>of</strong> these CYPs in the activation and detoxication <strong>of</strong> chemical<br />

carcinogens has already been determined. For example:<br />

– CYP2E1 is a major catalyst involved in the oxidation <strong>of</strong> benzene,<br />

styrene, CCl 4, CHCl 3, ethylene dichloride, vinylchloride, acrylonitrile,<br />

vinyl carbamate (Guengerich et al., 1991), ethanol (Perrot et al., 1989),<br />

dialkylnitrosamines (Yoo et al., 1988), isobutene (Cornet et al., 1991)<br />

and some other small molecules.<br />

– CYP1A1 is involved in the oxidation <strong>of</strong> polycyclic aromatic<br />

hydrocarbons (Shimada et al., 1989b).<br />

– CYP1A2 activates arylamines (Shimada et al., 1989a).


208 USE OF LONG-TERM CULTURES OF HEPATOCYTES IN TOXICITY TESTING<br />

– CYP3A4 is a major catalyst in the activation <strong>of</strong> aflatoxins, pyrrolizidine<br />

alkaloids and polycyclic hydrocarbon dihydrodiols (Shimada et al.,<br />

1989a; Shimada and Guengerich, 1989).<br />

The balance between the rates <strong>of</strong> formation <strong>of</strong> reactive metabolites and<br />

detoxication will greatly determine the potential toxic response <strong>of</strong> a<br />

chemical. Variables, known to affect normal biotransformation, such as<br />

enzyme induction and inhibition can also change the bioactivation rate <strong>of</strong><br />

chemicals. A clear example is the metabolism <strong>of</strong> halogenated biphenyls<br />

after treatment with arochlor 1254 (Borlakoglu and Wilkins, 1993).<br />

Consequently, the toxicity <strong>of</strong> chemical products <strong>of</strong>ten depends upon their<br />

specific biotransformation, the presence, absence, induction and inhibition<br />

<strong>of</strong> specific phase 1 and phase 2 enzymes involved in their metabolism.<br />

Towards an in vitro approach <strong>of</strong> risk assessment<br />

Nearly all toxicological studies on chemical products, including industrial<br />

chemicals, agrochemicals, pharmaceuticals, additives, materials in contact<br />

with food and cosmetics, have been carried out in vivo using experimental<br />

animals, in particular small vertebrates (News and Views, 1993).<br />

Important scientific, technological, ethical and economic considerations,<br />

however, justify the actual search for in vitro alternatives, replacing or<br />

improving existing in vivo methods and reducing the number <strong>of</strong> animals<br />

involved (Frazier and Goldberg, 1990; Roberfroid, 1991).<br />

Since hepatocytes can be isolated from different species (Guguen-<br />

Guillouzo et al., 1982; Green et al., 1986; van’t Klooster et al., 1992)<br />

including man (Guguen-Guillouzo et al., 1982; Rogiers, 1993), they can<br />

represent a powerful tool for short-term risk assessment studies when used<br />

as suspensions <strong>of</strong> freshly isolated cells (up to 3–4 h) or as short-term<br />

cultures (up to 2 days) (Klaassen and Stacey, 1982; Guillouzo, 1986). They<br />

can be useful in biotransformation, cytotoxicity, hepatotoxicity and<br />

genotoxicity studies, in species selection and in mechanistic studies<br />

(Blaauboer, 1994). Long-term cultures <strong>of</strong> hepatocytes (up to several weeks)<br />

represent a somewhat different approach in risk assessment. Such systems<br />

are <strong>of</strong> interest for the assessment <strong>of</strong> long-term toxicity <strong>of</strong> xenobiotics<br />

including the occurrence <strong>of</strong> enzyme induction, the effects on xenobiotic<br />

biotransformation, lipid peroxidation, accumulation <strong>of</strong> triglycerides,<br />

changes in glutathione content, interaction between compounds and the<br />

hepatoprotection afforded by certain molecules. Consequently, long-term<br />

cultures have been particularly applied in the development <strong>of</strong><br />

pharmaceuticals, in pharmaco-toxicological studies (Guillouzo, 1986,<br />

1992; Rogiers and Vercruysse, 1993; Skett, 1995). As far as chemical<br />

products other than pharmaceuticals and in particular industrial chemicals<br />

and agrochemicals are concerned, the practical needs during development


are different. The compounds brought onto the European market are<br />

labelled and need to fulfill only the legal demands <strong>of</strong> the specific category<br />

to which they belong.<br />

Testing is only obligatory when they reach the market and the type <strong>of</strong><br />

tests needed per category <strong>of</strong> chemicals is clearly outlined. In Europe legal<br />

categories consist <strong>of</strong> dangerous compounds (88/379/EEC, 93/18/EEC), 1a,b<br />

phytopharmaceuticals (91/414/EEC, 93/71/EEC), 2 biocides (93/C239/03), 3<br />

cosmetics (76/ 768/EEC, 93/35/5/EEC) 4a,b and food additives. Of the latter<br />

category the most comprehensive surveys are carried out by the Joint<br />

Expert Committee on Food Additives (JECFA) <strong>of</strong> the World Health<br />

Organization and the Food and Agriculture Organization <strong>of</strong> the United<br />

Nations (Conning, 1993).<br />

Less sophisticated in vitro studies have been performed on industrial<br />

chemicals and agrochemicals than is the case for pharmaceuticals. The only<br />

field in which isolated hepatocytes have already been incorporated into<br />

routine screening <strong>of</strong> industrial chemicals for regulatory purposes is in<br />

genotoxicity testing (Swierenga et al., 1991). The potentialities, however,<br />

<strong>of</strong> in vitro testing for these compounds, in particular <strong>of</strong> the use <strong>of</strong> long-term<br />

cultures, has not yet been explored in depth, although induction, inhibition,<br />

biotransformation, chronic toxicity, interaction between chemicals and<br />

mechanistic studies are <strong>of</strong> great interest for these compounds too.<br />

Human exposure to chemical products such as pesticides, eventually<br />

reaching the food chain as residues or <strong>of</strong> potential risk for workers and<br />

operators spraying the fields, is such an interesting research area. For<br />

example, Alachlor ® , a herbicide, <strong>of</strong> which millions <strong>of</strong> tons are used per<br />

year, has been classified as a potential human carcinogen (Leslie et al.,<br />

1989) because <strong>of</strong> tumour formation in rats and DNA damage observed in<br />

isolated rat hepatocytes (Bonfanti et al., 1992).<br />

In vivo studies concerning its biotransformation in rat, mouse and<br />

monkey, however, pointed to the observation that Alachlor ® is metabolized<br />

via different pathways in rodents and monkeys, suggesting a lower risk for<br />

man than assumed (internal report Monsanto, 1988). In the future, this<br />

type <strong>of</strong> biotransformation study could easily be performed with short- and<br />

long-term cultures <strong>of</strong> hepatocytes derived from different species, including<br />

man, providing relevant human information without interspecies<br />

extrapolation.<br />

Long-term hepatocyte cultures<br />

V.ROGIERS ET AL. 209<br />

During culture, hepatocytes undergo phenotypic changes as a function <strong>of</strong><br />

culture time affecting selectively components <strong>of</strong> phase 1 and/or phase 2<br />

biotransformation (Nakamura et al., 1983; Guillouzo, 1986; Mooney et<br />

al., 1992; Kocarek et al., 1993; Rogiers and Vercruysse, 1993). These<br />

changes are interpreted as dedifferentiation. In the literature, data have


210 USE OF LONG-TERM CULTURES OF HEPATOCYTES IN TOXICITY TESTING<br />

been presented that part <strong>of</strong> this loss <strong>of</strong> functionality can be prevented by<br />

several factors including soluble medium factors, extracellular matrix<br />

components and cell-cell interactions (reviews Guillouzo et al., 1990;<br />

Rogiers, 1993). At present no ‘ideal’ long-term hepatocyte culture model<br />

exists but valuable alternatives are being developed which take into<br />

account some <strong>of</strong> the factors mentioned. A recent development consists <strong>of</strong><br />

hepatocytes cultured in a collagen gel sandwich configuration (Dunn et al.,<br />

1991; Lee et al., 1992). This promising system is claimed to maintain longterm<br />

differentiation probably due to the reinstatement <strong>of</strong> the cellular<br />

polarity <strong>of</strong> the hepatocytes as a function <strong>of</strong> the extracellular matrix (Dunn<br />

et al., 1991; Lee et al., 1992). To date, only few results concerning xenobiotic<br />

metabolism, are available (Koebe et al., 1994). Another recently introduced<br />

model substantially improved the maintenance <strong>of</strong> xenobiotic metabolism<br />

by culturing hepatocytes on a mixture <strong>of</strong> crude membrane fractions with<br />

collagen type 1, combined with the use <strong>of</strong> culture medium supplemented<br />

with aprotinin and selenium (Saad et al., 1993).<br />

Also co-cultures <strong>of</strong> hepatocytes with rat epithelial cells <strong>of</strong> primitive<br />

biliary origin represent a rather new and valuable tool in xenobiotic<br />

biotransformation research and testing (Bégué et al., 1984a). The model<br />

has been developed in order to mimic better the microenvironment <strong>of</strong> the<br />

liver cells in vivo. It is until now, the only long-term hepatocyte culture<br />

system <strong>of</strong> which enough biotransformation data exist. Co-cultures <strong>of</strong><br />

hepatocytes retain to a great extent the morphological and biochemical<br />

characteristics <strong>of</strong> adult hepatocytes in vivo, including phase 1 and phase 2<br />

xenobiotic metabolism pathways (Guillouzo, 1986; Rogiers et al., 1992;<br />

Akrawi et al., 1993a). The following text reviews the actual knowledge<br />

concerning xenobiotic biotransformation in cocultured hepatocytes with<br />

emphasis on the authors’ own research.<br />

Phase 1<br />

reactions in co-cultured hepatocytes<br />

It has been claimed that as much as 100 per cent <strong>of</strong> the CYP content and Naminopyrine<br />

demethylation activity can be maintained in co-cultured rat<br />

hepatocytes with primitive biliary duct cells (Bégué et al., 1984b). Some<br />

other phase 1 enzymatic activities also appear to be maintained since drugs<br />

such as ketotifen (Le Bigot et al., 1987) and testosterone (Utesch, 1992)<br />

were metabolized by several pathways. Maier (1988) however, has<br />

reported that aldrin epoxidase activity underwent a significant decrease as<br />

a function <strong>of</strong> culture time. Niemann et al. (1991) was able to show that, as<br />

was also the case for mono-cultured rat hepatocytes, enzymatic activities<br />

belonging to the 3-methyl-cholanthrene-inducible family were better<br />

maintained than those belonging to the phenobarbital inducible family. In<br />

our own experiments with adult rat hepatocytes co-cultured with rat liver


V.ROGIERS ET AL. 211<br />

epithelial cells (Rogiers et al., 1990b), it was found that a steady-state<br />

situation for at least 10 days was obtained in which the total CYP content<br />

and 7-ethoxycoumarin O-deethylase and aldrin epoxidase activities were<br />

maintained at 25,100 and 15 per cent, respectively, <strong>of</strong> their corresponding<br />

values in freshly isolated hepatocytes. Both the total CYP content and 7ethoxycoumarin<br />

O-deethylase activity, but not the aldrin epoxidase<br />

activity, were induced by exposure to phenobarbital (Rogiers et al., 1990b)<br />

or sodium valproate (Rogiers et al., 1988b). Furthermore, the combination<br />

<strong>of</strong> inducing agents with co-cultivation <strong>of</strong> rat hepatocytes with rat epithelial<br />

cell clones has recently been proposed as the best way for stabilization <strong>of</strong><br />

the CYP system. This method is better than the use <strong>of</strong> a perfusion system<br />

or changing the extracellular matrix from collagen to matrigel (Wegner et<br />

al., 1991). The results mentioned above indicate a degree <strong>of</strong> selection in the<br />

ability <strong>of</strong> co-cultured hepatocytes to maintain the expression and<br />

inducibility <strong>of</strong> individual members <strong>of</strong> the CYP superfamily. From the work<br />

<strong>of</strong> Akrawi (Akrawi et al., 1993a), using mRNA analysis <strong>of</strong> co-cultured rat<br />

hepatocytes, it appeared that the abundance <strong>of</strong> CYP2B mRNAs declined to<br />

about 30 per cent <strong>of</strong> the initial value by 4 days but that thereafter it remained<br />

constant. The inducibility by phenobarbital (Akrawi et al., 1993a) and<br />

sodium valproate (Rogiers et al., 1992) was also maintained. These results<br />

were confirmed by Western blotting (Akrawi et al. 1993b). RNase<br />

protection assays using probes capable <strong>of</strong> distinguishing between CYP2B1<br />

and CYP2B2 mRNAs demonstrated that the relative abundance and<br />

inducibility <strong>of</strong> each <strong>of</strong> the mRNAs were the same in co-cultures as in vivo.<br />

Co-cultured hepatocytes also maintained the expression <strong>of</strong> the CYP1A2<br />

gene and <strong>of</strong> genes coding for two other components <strong>of</strong> the CYP-mediated<br />

monooxygenase, namely NADPH cytochrome P450 reductase and<br />

cytochrome b 5 (Akrawi et al., 1993a). Both components were inducible by<br />

valproate and phenobarbital (Akrawi et al., 1993b; Shephard et al., 1994;<br />

Rogiers et al., 1994).<br />

In addition, we have shown using Western blotting (Akrawi et al.,<br />

1993b) and mRNA analysis (Akrawi et al., 1994; Shephard et al., 1994)<br />

that the expression <strong>of</strong> CYP4A and its specific inducibility (induced by<br />

valproate and not by phenobarbital) were maintained in co-cultured rat<br />

hepatocytes. By using antisense RNA probes that could discriminate<br />

between RNAs encoding different members <strong>of</strong> the CYP4A subfamily it was<br />

further demonstrated that CYP4A1, CYP4A2 and CYP4A3 were all<br />

induced by valproate, although to differing extents. None <strong>of</strong> these mRNAs<br />

was increased by phenobarbital (Akrawi et al., 1994; Shephard et al.<br />

1994). The results were very similar to those observed in vivo (Shephard et<br />

al., 1994).<br />

Flavin-containing monooxygenase (FMO), a less well-known phase 1<br />

biotransformation enzyme than the CYP system, is responsible for the<br />

oxygenation <strong>of</strong> drugs, pesticides and dietary components (Ziegler, 1980). It


212 USE OF LONG-TERM CULTURES OF HEPATOCYTES IN TOXICITY TESTING<br />

may activate as well as deactivate a number <strong>of</strong> important molecules such as<br />

thioether-containing pesticides (Hajjar and Hodgson, 1980), 3,3′-dichlorobenzidine<br />

(Iba and Thomas, 1988), N-methyl-4-aminobenzene (Kadlubar<br />

et al., 1976) and others. The expression <strong>of</strong> FMOs is also maintained better<br />

and for a longer time in co-cultures <strong>of</strong> rat hepatocytes than it is in monocultures<br />

<strong>of</strong> these cells (Coecke et al., 1993). In co-cultures a steady state<br />

situation is obtained at a level <strong>of</strong> approximately 40 per cent <strong>of</strong> its initial<br />

value in freshly isolated hepatocytes (Coecke et al., 1993).<br />

Hormonal regulation <strong>of</strong> FMO is retained (Coecke et al., 1995a,b). It was<br />

shown that 17β-oestradiol significantly decreased FMO activity in cocultures<br />

<strong>of</strong> male rat hepatocytes which was not the case for testosterone<br />

and 5α-dihydrotestosterone. These data are in accordance with in vivo<br />

results (Coecke et al., 1995b). In addition, the thyroid hormone thyroxine<br />

and its metabolite L-triiodothyronine were found to cause a significant<br />

decrease in FMO activity suggesting a suppressive role in the regulation <strong>of</strong><br />

FMO in rat liver. In vivo data on this subject are not available in the<br />

literature.<br />

Phase 2<br />

reactions in co-cultured hepatocytes<br />

The activity, expression and regulation <strong>of</strong> the different glutathione Stransferase<br />

(GST) isoenzymes in co-cultured rat hepatocytes, has been<br />

extensively studied by our group (Vandenberghe et al., 1988a,b, 1989,<br />

1990a,b, 1991). As far as the enzymatic activity is concerned, it was<br />

observed that the composition <strong>of</strong> the culture medium was <strong>of</strong> much less<br />

influence in co-cultures than was the case in mono-cultures (Vandenberghe<br />

et al., 1988b). In cocultures GST activity was maintained for a longer<br />

period and at a more stable level, comparable to the in vivo situation. This<br />

observation was confirmed later by Niemann et al. (1991) and Utesch and<br />

Oesch (1992), although the latter investigators reported a high variability<br />

in their results depending on the batch <strong>of</strong> epithelial cells. GST activity in cocultured<br />

rat hepatocytes was found to be increased or decreased by<br />

phenobarbital and valproate, respectively (Rogiers et al., 1988a; Rogiers et<br />

al., 1992).<br />

These results are in good accordance with previously obtained in vivo<br />

data (Rogiers et al., 1988a; Rogiers et al., 1992). They were also confirmed<br />

at the protein level using the Western blot technique (Rogiers et al., 1995).<br />

Furthermore, GST activity is increased significantly, in a dose dependent<br />

way by ethanol. Both GST protein and mRNA amounts (in particular, GST<br />

subunits 3 and 4) were increased by this compound (Coecke et al., 1995c).<br />

Results obtained using different substrates suggested that the GST isoenzyme<br />

pr<strong>of</strong>ile changes as soon as hepatocytes are seeded in culture<br />

(Vandenberghe et al., 1988b). By a combination <strong>of</strong> GSH agarose affinity


V.ROGIERS ET AL. 213<br />

chromatography and reversed phase HPLC, the GST subunits <strong>of</strong> cocultured<br />

rat hepatocytes were purified and separated (Vandenberghe et al,<br />

1988a). Alterations comparable to those observed for mono-cultures<br />

suggested changes towards a more ‘foetal-like’ state. Less variations,<br />

however, were noticed in the GST subunit pattern <strong>of</strong> co-cultured<br />

hepatocytes when various media conditions were compared. Incorporation<br />

<strong>of</strong> 35 S-methionine in the medium showed the ability <strong>of</strong> co-cultured rat<br />

hepatocytes to synthesize the different GST subunits and suggested that<br />

changes in GST subunit expression under various culture conditions were<br />

the result <strong>of</strong> in vitro ‘de novo’ synthesis (Vandenberghe et al., 1990a).<br />

Northern blot analysis, using specific cDNA probes showed that the<br />

mRNA levels encoding GST subunits 1/2, 3/4 and 7 were very dependent<br />

on the culture medium.<br />

Again in co-cultures, the changes observed were much less marked than<br />

was the case for mono-cultures (Morel et al., 1989; Vandenberghe et al.,<br />

1990b). As already mentioned for conventionally cultured rat hepatocytes,<br />

phenobarbital had inducing effects on all the GST subunits, but to a<br />

different extent for each subunit (Vandenberghe, 1989). The increased<br />

steady-state mRNA levels observed in co-cultures after phenobarbital<br />

exposure were the result <strong>of</strong> an increased transcriptional activity <strong>of</strong> the GST<br />

genes together with a stabilizing effect <strong>of</strong> the compound (Vandenberghe et<br />

al., 1991).<br />

Also <strong>of</strong> interest is that the hormonal regulation <strong>of</strong> GST is maintained in<br />

co-cultures <strong>of</strong> male rat hepatocytes. 17β-Oestradiol, triiodothyronine and<br />

thyroxine cause a significant decrease in GST activity. Both the overall GST<br />

activity and in particular that <strong>of</strong> GST 3–3 and 3–4 are decreased (Coecke<br />

et al., 1995c). In contrast, male sex hormones and human growth hormone<br />

had little effect on the overall activity. The effects <strong>of</strong> triiodothyronine and<br />

thyroxine were particularly oriented towards GST subunits 3 and 4 and<br />

towards an as yet unidentified GST subunit, which was significantly<br />

increased (Coecke et al., 1995c). 17β-Oestradiol shifted the GST subunit<br />

pattern towards the one observed in freshly isolated cells whereas growth<br />

hormone had no specific effect on the individual protein classes (Coecke et<br />

al., 1995c).<br />

These results clearly show a hormonal regulation <strong>of</strong> GST in co-cultured<br />

rat hepatocytes, although previous work with mono-cultures failed to<br />

prove any direct effect (Gebhardt et al., 1990). The effects are most<br />

pronounced for the Mu-class GSTs. In man, Mu-class GST genes are<br />

structurally very similar to the rat genes and are <strong>of</strong> particular interest<br />

because 45 per cent <strong>of</strong> the European population fails to express a<br />

transferase at the GST M 1 locus (Zhong et al., 1993). It is this class <strong>of</strong> GSTs<br />

that is very effective in deactivating mutagenic and carcinogenic epoxides.<br />

UDP glucuronyltransferases (UDP-GT) have been much less studied in<br />

cocultures than GST. From the work <strong>of</strong> Niemann et al. (1991) it appears


214 USE OF LONG-TERM CULTURES OF HEPATOCYTES IN TOXICITY TESTING<br />

that 1-naphthol UDP-GT activity is maintained well in co-cultures whereas<br />

this is not true for morphine UDP-GT, although for both a steady state<br />

situation was reached. These data point to a shift towards a more<br />

differentiated pattern since 1-naphthol is considered to be more specific for<br />

the late foetal form <strong>of</strong> UDP-GT and morphine for the neonatal form. The<br />

maintenance <strong>of</strong> 1-naphthol UDP-GT has been confirmed by Utesch and<br />

Oesch (1992). Other studies on preservation <strong>of</strong> phase 2 enzymatic activity<br />

in co-cultures have dealt with the identification <strong>of</strong> the metabolites formed<br />

when drugs are added to the culture medium. In human co-culture it could<br />

be shown that the glucuronide metabolite <strong>of</strong> ketotifen was still present<br />

after 3 weeks whereas it became undetectable after 6 days in mono-culture<br />

(Bégué et al., 1984b). Similar observations have been made for other drugs<br />

such as caffeine and theophylline (Ratanasavanh et al., 1990).<br />

Conclusions<br />

At present no ideal culture system for hepatocytes can be proposed. In all<br />

models reported in the literature, phenotypic changes occur, affecting the<br />

various components <strong>of</strong> phase 1 and phase 2 xenobiotic metabolism to a<br />

different extent. An interesting conclusion, however, remains from the<br />

observation that, when co-cultures and mono-cultures <strong>of</strong> hepatocytes are<br />

compared, cocultures exhibit higher biotransformation capacities which are<br />

better and preserved for longer than is the case for mono-cultures. The<br />

inducibility by common inducers is fairly well maintained and seems, to a<br />

certain extent, comparable with the in vivo situation. In addition, hormonal<br />

regulation <strong>of</strong> phase 1 and phase 2 key enzymes seems to be well maintained<br />

and comparable with the in vivo situation. Co-cultures <strong>of</strong> hepatocytes with<br />

rat liver epithelial cells are therefore already <strong>of</strong> importance as an alternative<br />

model for risk assessment. In particular, when long-term effects <strong>of</strong> a<br />

chemical are to be expected.<br />

Some experience already exists concerning the application <strong>of</strong> co-cultured<br />

hepatocytes for the study <strong>of</strong> pharmaceuticals. As far as chemical products<br />

other than pharmaceuticals are concerned, experience is lacking although<br />

interesting results are to be expected particularly in those cases where<br />

chemicals can interfere with the human organism via the food chain or by<br />

occupational exposure. In vitro exploration <strong>of</strong> this new field in toxicology<br />

is a challenge for the coming years.<br />

Notes<br />

1a. 88/379/EEC<br />

Directive du Conseil du 7 juin 1988 concernant le rapprochement des<br />

dispositions législatives, réglementaires et administratives des Etats membres


elatives à la classification, à l’emballage et à 1’étiquetage des préparations<br />

dangereuses. Journal Officiel des Communautés européennes no L187, 16<br />

juillet 1988, p. 14.<br />

1b. 93/18/EEC<br />

Directive 93/18/CEE de la Commission du 5 avril 1993 portant troisième<br />

adaptation au progrès technique de la directive 88/379/CEE du Conseil<br />

concernant le rapprochement des dispositions législatives, réglementaires et<br />

administratives des Etats membres relatives à la classification, à l’emballage<br />

et à 1'étiquetage des preparations dangereuses. Journal Officiel des<br />

Communautés européennes no L104, 29 Avril 1993, p. 46.<br />

2a. 91/414/EEC<br />

Richtlijn van de Raad van 15 juli 1991 betreffende het op de markt<br />

brengen van gewasbeschermingsmiddelen. Publikatieblad van de Europese<br />

Gemeenschappen no L230, 19 Augustus 1991, p. 1.<br />

2b. 93/71/EEC<br />

Directive 93/71/CEE de la Commission du 27 juillet 1993 modifiant la<br />

directive 91/414/CEE du Conseil concernant la mise sur le marché<br />

des produits phytopharmaceutiques. Journal Officiel des Communautés<br />

européennes no L221, 31 Août 1993, p. 27.<br />

3. 93/C239/03<br />

Voorstel voor een richtlijn van de Raad betreffende het op de markt<br />

brengen van biociden. Publicatieblad van de Europese Gemeenschappen no<br />

C239, 3 September 1993, p. 3.<br />

4a. 76/768/EEC<br />

Richtlijn van de Raad van 27 juli 1976 betreffende de onderlinge<br />

aanpassing van de wetgevingen der Lid-Staten inzake kosmetische produkten.<br />

Publikatieblad van de Europese Gemeenschappen no L262, 27 juli 1976, p.<br />

169.<br />

4b. 93/35/EEC<br />

Directive 93/35/CEE du Conseil du 14 juin 1993 modifiant pour la sixième<br />

fois la directive 76/768/CEE concernant le rapprochement des lé-gislations des<br />

Etats membres relatives aux produits cosmétiques. Journal Officiel des<br />

Communautés européennes no L151, 23 juin 1993 p. 32.<br />

References<br />

V.ROGIERS ET AL. 215<br />

AKRAWI, M., ROGIERS, V., VANDENBERGHE, Y., PALMER, C.,<br />

VERCRUYSSE, A., SHEPHARD, E. and PHILLIPS I., 1993a, Maintenance<br />

and induction in cocultured rat hepatocytes <strong>of</strong> components <strong>of</strong> the cytochrome<br />

P450 mediated monooxygenase, Biochemical Pharmacology, 45, 1583–91.<br />

AKRAWI, M., SHEPHARD, E.A., PHILLIPS, I.R., VERCRUYSSE, A. and<br />

ROGIERS, V., 1993b, Effects <strong>of</strong> phenobarbital and valproate on the<br />

expression <strong>of</strong> cytochrome P-450 mediated mono-oxygenases in co-cultured rat<br />

hepatocytes, <strong>Toxicology</strong> In Vitro, 7, 477–80.<br />

AKRAWI, M., ROGIERS, V., VERCRUYSSE, A., PHILLIPS, I.R. and SHEPHARD,<br />

E.A., 1994, Long-term preservation and induction <strong>of</strong> drug metabolizing


216 USE OF LONG-TERM CULTURES OF HEPATOCYTES IN TOXICITY TESTING<br />

enzymes in co-cultured rat hepatocytes, Biochemical Society Transaction, 22,<br />

124S.<br />

BÉGUÉ, J.M., GUGUEN-GUILLOUZO, C., PASDELOUP, N. and GUILLOUZO,<br />

A., 1984a, Prolonged maintenance <strong>of</strong> active cytochrome P450 in adult rat<br />

hepatocytes co-cultured with another liver cell type, Hepatology, 4, 839–42.<br />

BÉGUÉ, J.M., LE BIGOT, J.F., GUGUEN-GUILLOUZO, C., KIECHEL, J.R. and<br />

GUILLOUZO, A., 1984b, Cultured human adult hepatocytes: a new model for<br />

drug metabolism studies, Biochemical Pharmacology, 32, 1643–46.<br />

BÉGUÉ, J.M., BAFFET, G., CAMPION, J.P. and GUILLOUZO, A., 1988,<br />

Differential response <strong>of</strong> primary cultures <strong>of</strong> human and rat hepatocytes to<br />

aflatoxin B1-induced cytotoxicity and protection by the hepatoprotective agent<br />

(+)-cyamidanol-3, Biology <strong>of</strong> the Cell, 63, 327–33.<br />

BLAAUBOER, B.J., BOOBIS, A.R., CASTELL, J.V., COECKE, S., GROOTHUIS,<br />

G.M.M., GUILLOUZO, A., HALL, T.J., HAWKSWORTH, G.M.,<br />

LORENZON, G., MILTENBURGER, H.G., ROGIERS, V., SKETT, P.,<br />

VILLA, P. and WIEBEL, F., 1994, The practical applicability <strong>of</strong> hepatocyte<br />

cultures in routine testing, Alternatives to Laboratory Animals, 22, 231–41.<br />

BONFANTI, M., TAVERNA, P., CHIAPPETTA, L., VILLA, P., D’INCALCI, A.,<br />

BAGNATTI, R. and FANELLI, R., 1992, DNA damage induced by alachlor<br />

after in vitro activation by rat hepatocytes, <strong>Toxicology</strong>, 72, 207–19.<br />

BORLAKOGLU, J.T. and WILKINS, J.P., 1993, Microsomal oxidation <strong>of</strong> bromo-,<br />

chloro-, and fluorobiphenyls, Comparative Biochemistry and Physiology, 105,<br />

119– 25.<br />

COECKE, S., SEGAERT, A., VERCRUYSSE, A. and ROGIERS, V., 1993,<br />

Expression <strong>of</strong> flavin-containing monooxygenase activity in adult rat<br />

hepatocytes under various culture conditions, <strong>Toxicology</strong> in Vitro, 7, 487–91.<br />

COECKE, S., CALLAERTS, A., VERLEYE, G., PHILLIPS, I.R., VERCRUYSSE,<br />

A., SHEPHARD, E.A. and ROGIERS, V., 1995a, Effect <strong>of</strong> thyroid hormones<br />

on flavin-containing monooxygenase activity in co-cultured adult rat<br />

hepatocytes, Biochemical and Biophysical Research Communications,<br />

submitted.<br />

COECKE, S., CORNET, M., VERLEYE, G., PHILLIPS, I.R., VERCRUYSSE, A.,<br />

SHEPHARD, E.A. and ROGIERS, V., 1995b, Hormonal regulation <strong>of</strong><br />

microsomal flavin-containing monooxygenase activity by sex steroids and<br />

growth hormone in co-cultured male rat hepatocytes, Endocrinology,<br />

submitted.<br />

COECKE, S., DEBAST, G., CALLAERTS, A., FORIERS, A., AKRAWI, M.,<br />

SONCK, W., VERLEYE, G., PHILLIPS, I.R., VERCRUYSSE, A., SHEPHARD,<br />

E.A. and ROGIERS, V., 1995c, Hormonal regulation <strong>of</strong> glutathione Stransferase<br />

expression in co-cultured adult rat hepatocytes, Biochemical<br />

Journal, submitted.<br />

COLES, B. and KETTERER, B., 1990, The role <strong>of</strong> glutathione transferases in<br />

chemical carcinogenesis, Critical Reviews in Biochemistry and Molecular<br />

Biology, 25, 47–70.<br />

CONNING, D.M., 1993, <strong>Toxicology</strong> <strong>of</strong> food and food additives, in Ballantyne, B.,<br />

Marrs, T. and Turner, P. (Eds) General and Applied <strong>Toxicology</strong>, Vol. 2, pp.<br />

1343– 71, New York: Stockton Press, The Macmillan Press.


V.ROGIERS ET AL. 217<br />

CORNET, M., SONCK, W., CALLAERTS, A., CSANADY, G., VERCRUYSSE,<br />

A., LAIB, R. and ROGIERS, V., 1991, In vitro biotransformation <strong>of</strong> 2methylpropene<br />

(isobutene): epoxide formation in mice liver, Archives <strong>of</strong><br />

<strong>Toxicology</strong>, 65, 263–7.<br />

DUNN, J.C.Y., TOMPKINS, R.G. and YARMUSH, M.L., 1991, Long-term in<br />

vitro function <strong>of</strong> adult hepatocytes in a collagen sandwich configuration,<br />

Biotechnology Progress, 7, 237–45.<br />

FRAZIER, J.M. and GOLDBERG, A.M., 1990, Alternatives to and reduction <strong>of</strong><br />

animal use in biomedical research, education and testing, Alternatives to<br />

Laboratory Animals, 18, 65–74.<br />

GEBHARDT, R., FITZKE, H., FAUSEL, M., EISENMANN-TAPPE, I. and<br />

MECKE, D., 1990, Influence <strong>of</strong> hormones and drugs on glutathione Stransferase<br />

levels in primary cultures <strong>of</strong> adult rat hepatocytes, Cell Biology and<br />

<strong>Toxicology</strong>, 6, 365–78.<br />

GREEN, C.E., LE VALLEY, S.E. and TYSON, C.A., 1986, Comparison <strong>of</strong><br />

amphetamine metabolism using isolated hepatocytes from five species<br />

including human, Journal <strong>of</strong> Pharmacology and Experimental Therapeutics,<br />

237, 931–6.<br />

GUENGERICH, F.P., KIM, D.H. and IWASAKI, M., 1991, Role <strong>of</strong> human<br />

cytochrome P-450 IIE1 in the oxidation <strong>of</strong> many low molecular weight cancer<br />

suspects, Chemical Research in <strong>Toxicology</strong>, 4, 168–79.<br />

GUGUEN-GUILLOUZO, C., CAMPION, J.P., BRISSOT, P., GLAISE, D.,<br />

LAUNOIS, B., BOUREL, M. and GUILLOUZO, A., 1982, High yield<br />

preparation <strong>of</strong> isolated human adult hepatocytes by enzymatic perfusion <strong>of</strong> the<br />

liver, Cell Biology International Reports, 6, 625–8.<br />

GUGUEN-GUILLOUZO, C. and GUILLOUZO, A., 1986, Methods for<br />

preparation <strong>of</strong> adult and fetal hepatocytes, in Guillouzo, A. and Guguen-<br />

Guillouzo, C. (Eds) Isolated and Cultured Hepatocytes, pp. 1–12, Paris: Les<br />

éditions INSERM and London: John Libbey Eurotext.<br />

GUILLOUZO, A., 1986, Utilisation des hepatocytes isolés et en culture pour des<br />

études de métabolisme et de cytotoxicité des xénobiotiques, in Guillouzo, A.<br />

and GuguenGuillouzo, C. (Eds) Hépatocytes isolés et en culture, pp. 327–46,<br />

Paris: Les Editions INSERM and London: John Libbey Eurotext.<br />

GUILLOUZO, A., 1992, Hepatotoxicity, in Frazier, J.M. (Ed). In vitro Toxicity<br />

Testing: Applications to Safety Evaluation, pp. 45–83, New York: Marcel<br />

Dekker Inc.<br />

GUILLOUZO, A., MOREL F., RATANASAVANH, D., CHESNE, C. and<br />

GUGUEN-GUILLOUZO, C., 1990, Long-term culture <strong>of</strong> functional<br />

hepatocytes, <strong>Toxicology</strong> in Vitro, 4, 415–27.<br />

HAJJAR, N.P. and HODGSON, E., 1980, Flavin adenine dinucleotide-dependent<br />

monooxygenase: its role in the sulfoxidation <strong>of</strong> pesticides in mammals,<br />

Science, 209, 1134–6.<br />

IBA, M.M. and THOMAS, P.E., 1988, Activation <strong>of</strong> 3,3'-dichlorobenzidine in rat<br />

liver microsomes to mutagens: involvement <strong>of</strong> cytochrome P450d,<br />

Carcinogenesis, 9, 717–23.<br />

JOVER, R., PONSODA, X., GOMEZ-LECHON, M.J. and CASTELL, J.V., 1992,<br />

Potentiation <strong>of</strong> heroin and methadone hepatotoxicity by ethanol: an in vitro<br />

study using cultured human hepatocytes, Xenobiotica, 22, 471–8.


218 USE OF LONG-TERM CULTURES OF HEPATOCYTES IN TOXICITY TESTING<br />

KADLUBAR, F.F., MILLER, J.A. and MILLER, E.C., 1976, Microsomal Noxidation<br />

<strong>of</strong> the hepatocarcinogen N-methyl-4-aminobenzene and the<br />

reactivity <strong>of</strong> N-hydroxy arylamines to reactive sulfuric acid esters, Cancer<br />

Research, 36, 1196– 206.<br />

KLAASSEN, C.D. and STACEY, N.H., 1982, Use <strong>of</strong> isolated hepatocytes in<br />

toxicity assessment, in Plaa, G. and Hewitt, W.R. (Eds) <strong>Toxicology</strong> <strong>of</strong> the<br />

Liver, pp. 147–79, New York: Raven Press.<br />

KOCAREK, T.A., SCHUETZ, E.G. and GUZELIAN, P.S., 1993, Differentiated<br />

induction <strong>of</strong> cytochrome P450b/e and P450p mRNAs by dose <strong>of</strong><br />

phenobarbital in primary cultures <strong>of</strong> adult rat hepatocytes, Molecular<br />

Pharmacology, 38, 440–4.<br />

KOEBE, H.G., PAHERNIK, S., EYER, P. and SCHILDBERG, F.W., 1994,<br />

Collagen gel immobilization, a useful cell culture technique for long-term<br />

metabolic studies on human hepatocytes, Xenobiotica, 24, 95–107.<br />

LE BIGOT, J.F., BÉGUÉ, J.M., KIECHEL, J.R. and GUILLOUZO, A., 1987,<br />

Species differences in metabolism <strong>of</strong> ketotifen in rat, rabbit and man:<br />

demonstration <strong>of</strong> similar pathways in vivo and in cultured hepatocytes, Life<br />

Sciences, 40, 883–90.<br />

LEE, J., MORGAN, J.R., TOMPKINS, R.G. and YARMUSH, M.L., 1992, The<br />

importance <strong>of</strong> proline on long-term hepatocyte function in a collagen gel<br />

sandwich configuration: regulation <strong>of</strong> protein secretion, Biotechnology and<br />

Bioengineering, 40, 298–305.<br />

LESLIE, C., REIDY, G.F. and STACEY, N.H., 1989, Effect <strong>of</strong> <strong>of</strong>urace, oxadixyl,<br />

and alachlor on xenobiotic biotransformation in the rat liver, Archives <strong>of</strong><br />

Environmental Contamination and <strong>Toxicology</strong>, 18, 876–80.<br />

MAIER, P., 1988, Development <strong>of</strong> in vitro toxicity tests with cultures <strong>of</strong> freshly<br />

isolated rat hepatocytes, Experientia, 44, 807–17.<br />

MOONEY, D., HANSEN, L., VACANTI, J., LANGER, R., FARMER, S. and<br />

INGBERG, D., 1992, Switching from differentiation to growth in hepatocytes:<br />

control by extracellular matrix, Journal <strong>of</strong> Cellular Physiology, 151, 497–505.<br />

MOREL, F., VANDENBERGHE, Y., PEMBLE, S., TAYLOR, J.B.,<br />

RATANASAVANH, D., ROGIERS, V., KETTERER, B. and GUILLOUZO,<br />

A., 1989, Regulation <strong>of</strong> glutathione S-transferase subunits 3 and 4 in cultured<br />

rat hepatocytes, FEBS Letters, 258, 99–102.<br />

NAKAMURA, T., YOSHIMOTO, K., NAKAYAMA, Y., TOMITA, Y. and<br />

ICHIHARA, A., 1983, Reciprocal modulation <strong>of</strong> growth and differentiated<br />

functions <strong>of</strong> mature rat hepatocytes in primary culture by cell-cell contact and<br />

cell membrane, Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences <strong>of</strong> the United<br />

States <strong>of</strong> America, 80, 7229–33.<br />

NEBERT, D.W., NELSON, D.R., ADESNIK, M., COON, M.J., ESTABROOK, R.<br />

W., GONZALEZ, F.J., GUENGERICH, F.P., GUNSALUS, I.C., JOHNSON,<br />

E.F., KEMPER, B., LEVIN, W., PHILLIPS, I., SATO, R. and WATERMAN, M.,<br />

1989, The P450 superfamily: updated listing <strong>of</strong> all genes and recommended<br />

nomenclature for the chromosomal loci, DNA, 8, 1–13.<br />

NEWS and VIEWS, 1993, Scientific procedures on living animals in Great Britain<br />

in 1992, Alternatives to Laboratory Animals, 21, 406–8.


V.ROGIERS ET AL. 219<br />

NIEMANN, C., GAUTHIER, J.C., RICHERT, L., IVANOV, M.A., MELCION, C.<br />

and CORDIER, A., 1991, Rat adult hepatocytes in primary pure and mixed<br />

monolayer culture, Biochemical Pharmacology, 42, 373–9.<br />

PERROT, N., NALPAS, B., YANG, C.S. and BEAUNE, P.H., 1989, Modulation <strong>of</strong><br />

cytochrome-P450 isozymes in human liver, by ethanol and drug intake,<br />

European Journal <strong>of</strong> Clinical Investigation, 19, 549–55.<br />

RATANASAVANH, D., BAFFET, G., LATINIER, M.F., RISSEL, M. and<br />

GUILLOUZO, A., 1988, Use <strong>of</strong> hepatocyte co-cultures in the assessment <strong>of</strong><br />

drug toxicity from chronic exposure, Xenobiotica, 18, 765–71.<br />

RATANASAVANH, D., BERTHOU, F., DREANO, Y., MONDINE, P.,<br />

GUILLOUZO, A. and RICHE, C., 1990, Methylcholanthrene but not<br />

phenobarbital enhances caffeine and theophylline metabolism in cultured<br />

human hepatocytes, Biochemical Pharmacology, 266, 683–8.<br />

ROBERFROID, M.B., 1991, Long term policy in toxicology, in Hendriksen, C.F.M.<br />

and Koëter, H.B.W.M. (Eds) Animals in Biochemical Research, pp. 35–48,<br />

Amsterdam: Elsevier Science Publishers B.V.<br />

ROGIERS, V., 1993, Cultures <strong>of</strong> human hepatocytes in in vitro<br />

pharmacotoxicology, in Rogiers, V., Sonck, W., Shephard, E. and Vercruysse,<br />

A. (Eds) Human Cells in in vitro Pharmaco-toxicology—Present Status within<br />

Europe, pp. 77–115, Brussels: VUB Press.<br />

ROGIERS, V., VANDENBERGHE, Y., CALLAERTS, A., SONCK, W., MAES, V.<br />

and VERCRUYSSE, A., 1988a, The inducing and inhibiting effects <strong>of</strong> sodium<br />

valproate in vivo on the biotransformation systems <strong>of</strong> xenobiotics in isolated<br />

rat hepatocytes, Xenobiotica, 18, 665–73.<br />

ROGIERS, V., VANDENBERGHE, Y., CORNET, M., CALLAERTS, A., SONCK,<br />

W., GUILLOUZO, A. and VERCRUYSSE, A., 1988b, The use <strong>of</strong> cultures and<br />

cocultures <strong>of</strong> rat hepatocytes for the study <strong>of</strong> the effects <strong>of</strong> valproate on phase<br />

I and II biotransformation systems <strong>of</strong> xenobiotics, in Woodhouse, K.W.,<br />

Yelland, C. and James, O.F.W. (Eds) The Liver, Metabolism and Ageing, pp.<br />

75–82, Amsterdam: Elsevier Biomedical Press.<br />

ROGIERS, V., VANDENBERGHE, Y., CALLAERTS, A., SONCK, W. and<br />

VERCRUYSSE, A., 1990a, Effects <strong>of</strong> dimethylsulphoxide on phase I and phase<br />

II biotransformation in cultured rat hepatocytes, <strong>Toxicology</strong> In Vitro, 4, 239–<br />

42.<br />

ROGIERS, V., VANDENBERGHE, Y., CALLAERTS, A., VERLEYE, G.,<br />

CORNET, M., MERTENS, K., SONCK, W. and VERCRUYSSE, A., 1990b,<br />

Phase I and phase II xenobiotic biotransformation in cultures and co-cultures<br />

<strong>of</strong> adult rat hepatocytes, Biochemical Pharmacology, 40, 1701–6.<br />

ROGIERS, V., CALLAERTS, A., VERCRUYSSE, A., AKRAWI, M., SHEPHARD,<br />

E. and PHILLIPS, I., 1992, Effects <strong>of</strong> valproate on xenobiotic<br />

biotransformation systems in rat liver: in vivo and in vitro experiments,<br />

Pharmaceutisch Weekblad, Scientific Edition, 14, 117–31.<br />

ROGIERS, V. and VERCRUYSSE, A., 1993, Rat hepatocyte cultures and cocultures<br />

in biotransformation studies <strong>of</strong> xenobiotics, <strong>Toxicology</strong>, 82, 193–208.<br />

ROGIERS, V., AKRAWI, M., VERCRUYSSE, A., SHEPHARD, E. and PHILLIPS,<br />

I. R., 1995, Effects <strong>of</strong> valproate on the expression <strong>of</strong> the cytochrome P450<br />

system in rat liver and co-cultured rat hepatocytes, Biochemical Journal, in<br />

press.


220 USE OF LONG-TERM CULTURES OF HEPATOCYTES IN TOXICITY TESTING<br />

SAAD, B., SCHOLL, F.A., THOMAS, H., SCHAWALDER, H., STREIT, V.,<br />

WAECHTER, F. and MAIER, P., 1993, Crude liver membrane fractions and<br />

extracellular matrix components as substrata regulate differentially the<br />

preservation and inducibility <strong>of</strong> cytochrome P-450 isoenzymes in cultured rat<br />

hepatocytes, European Journal <strong>of</strong> Biochemistry, 213, 805–14.<br />

SHEPHARD, E.A., AKRAWI, M., ROGIERS, V., BOOTMAN, M.,<br />

VERCRUYSSE, A. and PHILLIPS, I.R., 1995, Valproate increases the<br />

expression <strong>of</strong> the CYP4A subfamily in vivo and in vitro, Biochemical Journal,<br />

submitted.<br />

SHIMADA, T. and GUENGERICH, F.P., 1989, Evidence for cytochrome P-450<br />

NF, the nifedipine oxidase, being the principal enzyme involved in the<br />

bioactivation <strong>of</strong> aflatoxins in human liver, Proceedings <strong>of</strong> the National<br />

Academy <strong>of</strong> Sciences <strong>of</strong> the United States <strong>of</strong> America, 86, 462–5.<br />

SHIMADA, T., IWASAKI, M., MARTIN, M.V. and GUENGERICH, F.P., 1989a,<br />

Human liver microsomal cytochrome P-450 enzymes involved in the<br />

bioactivation <strong>of</strong> procarcinogens detected by umu gene response in Salmonella<br />

typhimurium TA1535/pSK 1002, Cancer Research, 49, 3218–28.<br />

SHIMADA, T., MARTIN, M.V., PRUESS-SCHWARTZ, D., MARNETT, L.J. and<br />

GUENGERICH, F.P., 1989b, Roles <strong>of</strong> individual human cytochrome P-450<br />

enzymes in the bioactivation <strong>of</strong> benzo(a)pyrene, 7,8-dihydroxy-7,8dihydrobenzo(a)<br />

pyrene, and other dihydrodiol derivatives <strong>of</strong> polycyclic<br />

aromatic hydrocarbons, Cancer Research, 49, 6304–12.<br />

SKETT, P., 1995, Problems in using isolated and cultured hepatocytes for<br />

xenobiotic metabolism/metabolism-based toxicity testing—Solutions?<br />

<strong>Toxicology</strong> in Vitro (in press).<br />

SWIERENGA, S.H.H., BRADLAW, J.A., BRILLINGER, R.L., GILMAN, J.P. W.,<br />

NESTMANN, E.R. and SAN, R.C., 1991, Recommended protocols based on a<br />

survey <strong>of</strong> current practice in genotoxicity testing laboratories: I. Unscheduled<br />

DNA synthesis assay in rat hepatocyte cultures, Mutation Research, 246, 235–<br />

53.<br />

TEE, L.B.G., DAVIES, D.S., SEDDON, C.E. and BOOBIS, A.R., 1987, Species<br />

differences in the hepatotoxicity <strong>of</strong> paracetamol are due to differences in the<br />

rate <strong>of</strong> conversion to its cytotoxic metabolite, Biochemical Pharmacology, 36,<br />

1041–52.<br />

TIMBRELL, J., 1993, Biotransformation <strong>of</strong> xenobiotics, in Ballantyne, B. (Marrs, T.<br />

and Turner, P. (Eds) General and Applied <strong>Toxicology</strong>, pp. 88–119, New York:<br />

Stockton Press.<br />

UTESCH, D. and OESCH, F., 1992, Dependency <strong>of</strong> the in vitro stabilization <strong>of</strong><br />

differentiated functions in liver parenchymal cells on the type <strong>of</strong> cell line used<br />

for coculture, In vitro Cellular Development Biology, 28, 193–8.<br />

VAN BLADEREN, P., 1988, Formation <strong>of</strong> toxic metabolites from drugs and other<br />

xenobiotics by glutathione conjugation, Trends in Pharmacological Sciences,<br />

9, 295–8.<br />

VANDENBERGHE, Y., GLAISE, D., MEYER, D., GUILLOUZO, A. and<br />

KETTERER, B., 1988a, Glutathione transferase isoenzymes in cultured rat<br />

hepatocytes, Biochemical Pharmacology, 37, 2482–95.<br />

VANDENBERGHE, Y., RATANASAVANH, D., GLAISE, D. and GUILLOUZO,<br />

A., 1988b, Influence <strong>of</strong> medium composition and culture conditions on


V.ROGIERS ET AL. 221<br />

glutathione S-transferase activity in adult rat hepatocytes during culture, In<br />

Vitro Cellular and Developmental Biology, 24, 281–8.<br />

VANDENBERGHE, Y., MOREL, F., FORIERS, A., KETTERER, B.,<br />

VERCRUYSSE, A., GUILLOUZO, A. and ROGIERS, V., 1989, Effect <strong>of</strong><br />

phenobarbital on the expression <strong>of</strong> glutathione S-transferase isoenzymes in<br />

cultured rat hepatocytes, FEBS Letters, 251, 59–64.<br />

VANDENBERGHE, Y., FORIERS, A., ROGIERS, V. and VERCRUYSSE, A.,<br />

1990a, Changes in expression and ‘de novo’ synthesis <strong>of</strong> glutathione Stransferase<br />

subunits in cultured adult rat hepatocytes, Biochemical<br />

Pharmacology, 39, 685–90.<br />

VANDENBERGHE, Y., MOREL, F., PEMBLE, S., TAYLOR, J.B., ROGIERS, V.,<br />

RATANASAVANH, D., VERCRUYSSE, A., KETTERER, B. and<br />

GUILLOUZO, A., 1990b, Changes in expression <strong>of</strong> mRNA coding for<br />

glutathione S-transferase subunits 1–2 and 7 in cultured rat hepatocytes,<br />

Molecular Pharmacology, 37, 372–6.<br />

VANDENBERGHE, Y., TEE, L., MOREL, F., ROGIERS, V., GUILLOUZO, A.<br />

and YEOH, G., 1991, Regulation <strong>of</strong> glutathione S-transferase gene expression<br />

by phenobarbital in cultured adult rat hepatocytes, FEBS Letters, 284, 103–8.<br />

VAN’T KLOOSTER, G.A.E., WOUTERSEN-VAN NIJANTEN, F.M.A., KLEIN,<br />

W.R., BLAAUBOER, B.J., NOORDHOEK, J. and VAN MIERT, A.S.J.P.A. M.,<br />

1992, Effects <strong>of</strong> various medium formulations and attachment substrata on the<br />

performance <strong>of</strong> cultured ruminant hepatocytes in biotransformation studies,<br />

Xenobiotica, 22, 523–34.<br />

WEGNER, H., MECKE, D. and GEBHARDT, R., 1991, Attempts to stabilize the<br />

cytochrome P450 dependent monooxygenase activity in primary cell-culture.<br />

Abstract in Third International ISSX meeting: Drug metabolism: molecules,<br />

models and man, p. 317, Amsterdam, June.<br />

Yoo, J.S.H., GUENGERICH, F.G. and YANG, C.S., 1988, Metabolism <strong>of</strong> Nnitrosodialkylamines<br />

by human liver microsomes, Cancer Research, 48, 1499–<br />

504.<br />

ZHONG, S., SPURR, N.K., HAYES, J.D. and WOLF, C.R., 1993, Deduced amino<br />

acid sequence gene structure and chromosome location <strong>of</strong> a novel human class<br />

Mu glutathione S-transferase, GSTM4, Biochemical Journal, 291, 41–50.<br />

ZIEGLER, D.M., 1980, Microsomal flavin-containing monooxygenase:<br />

oxygenation <strong>of</strong> nucleophilic nitrogen and sulfur compounds, in Jakoby, W.B.<br />

(Ed.) Enzymatic Basis <strong>of</strong> Detoxification, pp. 201–7, New York: Academic<br />

Press.


PART FIVE<br />

Mechanisms <strong>of</strong> toxicity <strong>of</strong> industrial<br />

chemicals


17<br />

Peroxisome Proliferation<br />

BRIAN G.LAKE and ROGER J.PRICE<br />

BIBRA International, Carshalton, Surrey<br />

Introduction<br />

Peroxisomes (sometimes referred to as ‘microbodies’) are single<br />

membranelimited cytoplasmic organelles which are characterised by their<br />

content <strong>of</strong> catalase and a number <strong>of</strong> hydrogen peroxide generating oxidase<br />

enzymes (Cohen and Grasso, 1981; Reddy and Lalwani, 1983). In rat liver,<br />

peroxisomes are normally spherical or oval in shape, approximately 0.5 µm<br />

in diameter and contain a finely granular matrix with a crystalline nucleoid<br />

core. A number <strong>of</strong> reviews have been published dealing with various<br />

aspects <strong>of</strong> hepatic peroxisome proliferation (Cohen and Grasso, 1981;<br />

Reddy and Lalwani, 1983; Hawkins et al., 1987; Stott, 1988; Lock et al.,<br />

1989; Moody et al., 1991; Bentley et al., 1993; Lake, 1993). This chapter<br />

will focus on mechanisms <strong>of</strong> hepatocarcinogenesis, species differences in<br />

response and risk assessment <strong>of</strong> rodent peroxisome proliferators.<br />

Peroxisome proliferation in rodent liver<br />

Since the initial observations on the hepatic effects <strong>of</strong> the hypolipidaemic<br />

agent cl<strong>of</strong>ibrate (Paget, 1963; Hess et al., 1965) many compounds have<br />

been shown to produce hepatic peroxisome proliferation in rats and mice.<br />

Liver enlargement is due to both hyperplasia and hypertrophy and<br />

organelle proliferation is associated with a differential induction <strong>of</strong><br />

peroxisomal enzyme activities. Peroxisomes, like mitochondria, contain a<br />

complete fatty acid β-oxidation cycle (Lazarow and DeDuve, 1976). While<br />

the enzymes <strong>of</strong> the β-oxidation cycle (normally assessed as cyanideinsensitive<br />

palmitoyl-CoA oxidation) are markedly induced, only small<br />

changes are observed in other peroxisomal enzyme activities such as<br />

catalase and D-amino acid oxidase. Apart from stimulating peroxisomal<br />

fatty acid metabolism, peroxisome proliferators also increase microsomal<br />

fatty acid ( -l)- and particularly -hydroxylase activities. This is due to<br />

induction <strong>of</strong> cytochrome P-450 isoenzymes in the CYP4A subfamily and is


224 PEROXISOME PROLIFERATION<br />

normally measured as lauric acid 12-hydroxylase (Sharma et al., 1988a, b;<br />

Gibson, 1989). Peroxisome proliferators also markedly increase carnitine<br />

acetyltransferase activity which is localised in peroxisomal, mitochondrial<br />

and microsomal fractions (Ishii et al., 1980; Bieber et al., 1981). In rat liver<br />

good correlations have been reported between the induction <strong>of</strong><br />

peroxisomal fatty acid β-oxidation and organelle proliferation and between<br />

the induction <strong>of</strong> peroxisomal and microsomal fatty acid oxidising enzyme<br />

activities (Lake et al., 1984a; Lin, 1987; Sharma et al., 1988a, b; Dirven et<br />

al., 1992).<br />

Several laboratories have demonstrated that the characteristics <strong>of</strong><br />

peroxisome proliferation in vivo may also be observed in vitro in primary<br />

rat and mouse hepatocyte cultures. Indeed, hepatocyte cultures have been<br />

employed for studying various aspects <strong>of</strong> peroxisome proliferation<br />

including structureactivity relationships and species differences in response<br />

(Gray et al., 1982; Elcombe, 1985; Bieri, 1993; Lake and Lewis, 1993;<br />

Foxworthy and Eacho, 1994).<br />

Rodent peroxisome proliferators<br />

Many different classes <strong>of</strong> chemicals have been found to produce<br />

peroxisome proliferation in the rat and mouse (Cohen and Grasso, 1981;<br />

Reddy and Lalwani, 1983; Stott, 1988; Moody et al., 1991; Bentley et al.,<br />

1993; Lake and Lewis, 1993). Classes <strong>of</strong> industrial chemicals include<br />

plasticisers, chlorinated solvents (e.g. trichloroethylene, perchloroethylene),<br />

chlorinated paraffins and other chemicals (e.g. perfluoro-n-octanoic acid).<br />

Types <strong>of</strong> plasticisers known to produce peroxisome proliferation include<br />

phthalate esters (e.g. di-(2-ethyl-hexyl)phthalate (DEHP), di-(isodecyl)<br />

phthalate), adipate esters (e.g. di-(2-ethyl-hexyl)adipate (DEHA)) and other<br />

compounds (e.g. tri-(2-ethylhexyl) trimellitate). Apart from industrial<br />

chemicals other known rodent hepatic peroxisome proliferators include<br />

herbicides, hypolipidaemic and other categories <strong>of</strong> therapeutic agents,<br />

certain steroids, food flavours and natural products.<br />

While peroxisome proliferators appear to be structurally diverse, at least<br />

for some compounds, similarities in their three-dimensional structures have<br />

been reported (Lake et al., 1988; Lake and Lewis, 1993). Many studies<br />

have demonstrated structure-activity relationships for various classes <strong>of</strong><br />

peroxisome proliferators including industrial chemicals (Lake and Lewis,<br />

1993). A characteristic feature <strong>of</strong> many, but not all, peroxisome<br />

proliferators is the presence <strong>of</strong> an acidic function (Lake et al., 1988; Lock<br />

et al., 1989). This acidic function is normally a carboxyl group, either<br />

present as a free carboxyl group in the parent structure or one that is<br />

unmasked by metabolism. Alternatively, the chemical may contain a<br />

chemical grouping which is a bioisostere <strong>of</strong> a carboxyl group (Thornber,


1979), such as tetrazole or a sulphonamide moiety (Eacho et al., 1986;<br />

Lock et al., 1989).<br />

It should be noted that rodent liver peroxisome proliferators exhibit<br />

marked compound potency differences. While potent peroxisome<br />

proliferators include compounds developed as hypolipidaemic agents (e.g.<br />

cipr<strong>of</strong>ibrate, Wy-14,643), plasticisers such as DEHP are less potent and<br />

chemicals such as acetylsalicylic acid are even less potent (Reddy et al.,<br />

1986; Barber et al., 1987; Lake and Lewis, 1993). For example, in a 30day<br />

feeding study, a similar magnitude <strong>of</strong> induction <strong>of</strong> palmitoyl-CoA<br />

oxidation was observed in rats fed 0.001 per cent cipr<strong>of</strong>ibrate and 0.5 per<br />

cent DEHP diets, whereas for DEHA a dietary level <strong>of</strong> >1.0 per cent but


226 PEROXISOME PROLIFERATION<br />

demonstrated to be effective in rat liver tumour promotion studies (Cattley<br />

and Popp, 1989; Bentley et al., 1993; Popp and Cattley, 1993).<br />

Mechanisms <strong>of</strong> hepatocarcinogenesis<br />

Several hypotheses have been proposed to account for why peroxisome<br />

proliferators can produce liver tumours in rodents. These mechanisms<br />

include:<br />

(a) Induction <strong>of</strong> sustained oxidative stress to hepatocytes (Reddy and<br />

Lalwani, 1983; Reddy and Rao, 1989).<br />

(b) A role <strong>of</strong> increased cell proliferation (Marsman et al., 1988; Popp and<br />

Marsman, 1991).<br />

(c) The promotion <strong>of</strong> spontaneously formed preneoplastic liver lesions<br />

(Schulte-Hermann et al., 1989; Cattley et al., 1991; Grasl-Kraupp et<br />

al., 1993).<br />

(d) A combination <strong>of</strong> two or all <strong>of</strong> the above factors.<br />

The oxidative stress hypothesis is based on the observation that the chronic<br />

administration <strong>of</strong> peroxisome proliferators produces a sustained oxidative<br />

stress in rodent hepatocytes due to an imbalance in the production and<br />

degradation <strong>of</strong> hydrogen peroxide (Reddy and Lalwani, 1983; Reddy and<br />

Rao, 1989). Peroxisome proliferators markedly induce the peroxisomal<br />

fatty acid β-oxidation cycle, but produce only a small increase in catalase<br />

activity. The first enzyme <strong>of</strong> the β-oxidation cycle, acyl-CoA oxidase,<br />

produces hydrogen peroxide and hence the cyclic oxidation <strong>of</strong> a single fatty<br />

acid molecule can result in the production <strong>of</strong> several molecules <strong>of</strong> hydrogen<br />

peroxide (Lazarow and DeDuve, 1976). Any excess hydrogen peroxide not<br />

destroyed by peroxisomal catalase can diffuse through the peroxisomal<br />

membrane into the cytosol where it will be a substrate for cytosolic<br />

selenium-dependent glutathione peroxidase. However, this enzyme activity<br />

and that <strong>of</strong> other enzymes including superoxide dismutase and glutathione<br />

S-transferases are <strong>of</strong>ten reduced by the administration <strong>of</strong> peroxisome<br />

proliferators to rodents (Reddy and Rao, 1989; Bentley et al., 1993; Lake,<br />

1993). These enzyme changes are postulated to result in increased<br />

intracellular levels <strong>of</strong> hydrogen peroxide which, either directly or via<br />

reactive oxygen species (e.g. hydroxyl radical), can attack membranes and<br />

DNA (Reddy and Lalwani, 1983; Reddy and Rao, 1989).<br />

A number <strong>of</strong> experimental observations have provided support for the<br />

involvement <strong>of</strong> oxidative stress in the hepatotoxicity <strong>of</strong> peroxisome<br />

proliferators (Reddy and Rao, 1989; Lake, 1993). For example,<br />

peroxisome proliferators have been reported in some studies to increase<br />

hepatic lipid peroxidation and lip<strong>of</strong>uscin deposition, to modulate levels <strong>of</strong><br />

hepatic antioxidants and to increase levels <strong>of</strong> 8-hydroxydeoxyguanosine in


B.G.LAKE AND R.J.PRICE 227<br />

hepatic DNA (Reddy and Rao, 1989; Bentley et al., 1993; Lake, 1993).<br />

However, the available data suggest that sustained oxidative stress is<br />

unlikely to be solely responsible for per oxisome proliferator-induced<br />

hepatocarcinogenesis in rodents. Although evidence <strong>of</strong> oxidative damage to<br />

hepatocytes has been observed in some studies, the magnitude <strong>of</strong> such<br />

effects does not correlate with the potency <strong>of</strong> the compound to produce<br />

tumours. For example, oxygen radical attack on DNA is known to result in<br />

a variety <strong>of</strong> modified DNA bases including 8-hydroxydeoxyguanosine.<br />

However, at bioassay dose levels both DEHP and DEHA produce similar<br />

increases in hepatic 8-hydroxydeoxyguanosine levels, but only DEHP<br />

produced liver tumours in male F344 rats (NTP, 1982a, b; Takagi et al.,<br />

1990; Lake, 1993).<br />

Many studies have demonstrated that cell proliferation is an important<br />

factor in the development <strong>of</strong> tumours by both genotoxic and nongenotoxic<br />

agents (Cohen and Ellwein, 1990, 1991). For example, an enhanced rate <strong>of</strong><br />

cell replication can increase the frequency <strong>of</strong> spontaneous lesions and the<br />

probability <strong>of</strong> converting DNA adducts from both endogenous and<br />

exogenous sources into mutations before they can be repaired (Cohen and<br />

Ellwein, 1990, 1991; Popp and Marsman, 1991). Peroxisome proliferators<br />

are known to produce a burst <strong>of</strong> cell replication in rodent hepatocytes<br />

during the first few days <strong>of</strong> administration (Reddy and Lalwani, 1983;<br />

Eacho et al., 1991). In some studies peroxisome proliferators have also<br />

been shown to produce a sustained stimulation <strong>of</strong> replicative DNA<br />

synthesis (Lake, 1993). Apart from intrinsic compound potency, dose is an<br />

important factor in determining whether a particular compound can<br />

produce either a transient or a sustained stimulation <strong>of</strong> replicative DNA<br />

synthesis in rodent hepatocytes. For example, low doses <strong>of</strong> nafenopin and<br />

Wy-14,643 do not produce a sustained stimulation <strong>of</strong> cell replication,<br />

whereas higher doses do produce this effect (Eacho et al., 1991; Price et al.,<br />

1992; Wada et al., 1992; Lake et al., 1993).<br />

Several studies have demonstrated the presence <strong>of</strong> numerous foci <strong>of</strong><br />

putative preneoplastic cells in the livers <strong>of</strong> untreated old rats and mice<br />

(Schulte-Hermann et al., 1983; Grasl-Kraupp et al., 1993). These lesions<br />

are considered to represent spontaneously initiated cells as they have<br />

similar biological characteristics to those <strong>of</strong> cells initiated by genotoxic<br />

carcinogens (Grasl-Kraupp et al., 1993). The ability <strong>of</strong> peroxisome<br />

proliferators to produce tumours in young compared to old rats has been<br />

investigated in studies with nafenopin (Kraupp-Grasl et al., 1991) and<br />

Wy-14,643 (Cattley et al., 1991). In both studies more adenomas and<br />

carcinomas were produced in old as against young rats.


228 PEROXISOME PROLIFERATION<br />

Species differences in response<br />

Many studies have examined species differences in hepatic peroxisome<br />

proliferation (Cohen and Grasso, 1981; Rodricks and Turnbull, 1987;<br />

Stott, 1988; Lock et al., 1989; Moody et al., 1991; Bentley et al., 1993).<br />

Based on both marker enzyme activities and ultrastructural examination<br />

the rat and mouse are clearly responsive species, the Syrian hamster<br />

appears to exhibit an intermediate response, whereas in most studies the<br />

guinea pig is either nonresponsive or refractory. For example, DEHP<br />

readily produces peroxisome proliferation in the rat and mouse, to a lesser<br />

extent in the Syrian hamster but not in the guinea pig (Osumi and<br />

Hashimoto, 1978; Lake et al., 1984b). Similar results have been obtained<br />

with more potent compounds including cipr<strong>of</strong>ibrate, clobuzarit, LY<br />

171883 and nafenopin (Orton et al., 1984; Eacho et al., 1986; Lake et al.,<br />

1989; Makowska et al., 1992).<br />

When assessing species differences in response a number <strong>of</strong> factors<br />

should be considered. These include the metabolism, disposition and dose<br />

<strong>of</strong> the test compound, sex differences, as well as intrahepatic differences in<br />

response. The importance <strong>of</strong> metabolism is illustrated by the industrial<br />

solvent trichloroethylene which produces peroxisome proliferation and<br />

liver tumours in the mouse but not in the rat (NCI, 1976; Elcombe, 1985).<br />

Metabolic studies demonstrated that the trichloroethylene was extensively<br />

metabolised to trichloroacetic acid in the mouse, whereas this was a minor<br />

saturable route <strong>of</strong> metabolism in the rat. That the difference in<br />

trichloroacetic acid formation was responsible for the observed species<br />

difference was demonstrated by the fact that this compound produced<br />

peroxisome proliferation in rat and mouse hepatocytes both in vivo and in<br />

vitro (Elcombe, 1985). An example <strong>of</strong> compound disposition is provided<br />

by DEHP which is known to be more extensively absorbed after oral<br />

administration in the rat than in the marmoset (Rhodes et al., 1986).<br />

However, the observed in vivo species differences in response are supported<br />

by the observation that metabolites <strong>of</strong> DEHP which produce peroxisome<br />

proliferation in rat hepatocytes in vitro have no significant effect in<br />

cultured marmoset hepatocytes (Elcombe and Mitchell, 1986). Generally,<br />

in vitro studies with primary hepatocyte cultures from the rat, mouse,<br />

Syrian hamster, guinea pig and marmoset have supported the results <strong>of</strong> in<br />

vivo studies in these species (Elcombe 1985; Elcombe and Mitchell, 1986;<br />

Lake et al., 1986; Bieri, 1993; Bentley et al., 1993; Foxworthy and Eacho,<br />

1994).<br />

Several studies have examined the ability <strong>of</strong> rodent peroxisome<br />

proliferators to produce effects in primates and humans. With respect to<br />

primates, studies with a number <strong>of</strong> compounds in both New (e.g.<br />

marmoset) and Old (e.g. Rhesus monkey) World monkeys have failed to<br />

provide any evidence <strong>of</strong> significant hepatic peroxisome proliferation


(Rodricks and Turnbull, 1987; Bentley et al., 1993). However, albeit at<br />

high doses two compounds, namely cipr<strong>of</strong>ibrate (Reddy et al., 1984) and<br />

DL-040 (Lalwani et al., 1985), have been reported to produce hepatic<br />

peroxisome proliferation in Cynomolgus and/or Rhesus monkeys. In<br />

humans, studies have been conducted in patients treated with several<br />

hypolipidaemic agents (all being rodent peroxisome proliferators) including<br />

cipr<strong>of</strong>ibrate, cl<strong>of</strong>ibrate, fen<strong>of</strong>ibrate and gemfibrozil (Bentley et al., 1993).<br />

While most studies have failed to detect any significant changes, cl<strong>of</strong>ibrate<br />

was reported to produce a small increase in the number <strong>of</strong> peroxisomes<br />

(Hanefeld et al., 1983) and cipr<strong>of</strong>ibrate to produce a small increase in the<br />

pro portion <strong>of</strong> the hepatocyte cytoplasm occupied by peroxisomes (cited in<br />

Bentley et al., 1993). However, owing to the large interindividual variation<br />

in peroxisome morphometrics observed in these studies, together with cell<br />

to cell variations and lobular variations, it is difficult to attach any clear<br />

biological significance to these findings (Bentley et al., 1993). Generally,<br />

peroxisome proliferators have not been reported to produce any significant<br />

effects on marker enzyme activities and/or peroxisomes in cultured primate<br />

and human hepatocytes (Bieri, 1993; Bentley et al., 1993; Foxworthy and<br />

Eacho, 1994).<br />

Some studies have also examined species differences in effects on cell<br />

replication. Both nafenopin and Wy-14,643 have been reported to<br />

stimulate replicative DNA synthesis in rat, but not in Syrian hamster,<br />

hepatocytes (Price et al., 1992; Lake et al., 1993). Although peroxisome<br />

proliferators can stimulate DNA synthesis in cultured rat hepatocytes,<br />

methylcl<strong>of</strong>enapate was reported to be ineffective in guinea pig, marmoset<br />

and human hepatocytes (Elcombe and Styles, 1989). Similarly, nafenopin<br />

has also been reported not to induce replicative DNA synthesis in human<br />

hepatocytes (Parzefall et al., 1991).<br />

Risk assessment <strong>of</strong> rodent liver peroxisome proliferators<br />

The key issues concerning the risk assessment <strong>of</strong> rodent liver peroxisome<br />

proliferators include:<br />

(a) Genotoxicity.<br />

(b) Likely human exposure.<br />

(c) Compound potency and no effect levels.<br />

(d) Precise mechanism(s) <strong>of</strong> liver tumour formation.<br />

(e) Species differences in response.<br />

B.G.LAKE AND R.J.PRICE 229<br />

Generally, peroxisome proliferators are considered to be non-genotoxic<br />

agents (Bentley et al., 1993; Budroe and Williams, 1993) and hence should<br />

be assessed differently from genotoxic carcinogens (Weisburger, 1994).<br />

Human exposure to rodent peroxisome proliferators depends on the


230 PEROXISOME PROLIFERATION<br />

intended usage <strong>of</strong> the particular compound. While hypolipidaemic agents<br />

are only administered to a restricted population <strong>of</strong> humans, exposure to<br />

industrial chemicals such as plasticisers is obviously far more widespread.<br />

For example, based on food surveillance surveys the daily human exposure<br />

to DEHA was reported to be 16 and 8.2 mg per person per day in 1987<br />

and 1990, respectively (MAFF 1987, 1990). In another study, where<br />

DEHA intake was assessed by measuring urinary levels <strong>of</strong> the major<br />

metabolite 2-ethylhexanoic acid, a median value <strong>of</strong> 2.7 mg per person per<br />

day was reported (L<strong>of</strong>tus et al., 1994).<br />

Apart from likely human exposure, consideration should be made <strong>of</strong> the<br />

relative potency <strong>of</strong> the particular compound to produce peroxisome<br />

proliferation and liver tumours in rodents. Plasticisers such as DEHP and<br />

DEHA are far less potent than certain therapeutic agents and<br />

experimentally used compounds (Reddy et al., 1986; Barber et al., 1987;<br />

Bentley et al., 1993; Lake and Lewis, 1993). Moreover rodent liver<br />

peroxisome proliferators exhibit clear no effect levels for both peroxisome<br />

proliferation and for tumour formation. For example, in the rat no effect<br />

levels for liver tumour formation have been observed in studies with<br />

several compounds including bezafibrate, cl<strong>of</strong>ibrate, DEHA and DEHP<br />

(Hartig et al., 1982; NTP 1982a, b). In addition, the threshold for tumour<br />

formation in rodents is appreciably greater than the threshold for<br />

peroxisome proliferation (Hartig et al., 1982; Reddy et al., 1986; Bentley<br />

et al., 1993).<br />

Several mechanisms have been proposed to account for why peroxisome<br />

proliferators produce tumours in rodent liver. If these various hypotheses<br />

are combined then a role for increased cell replication in peroxisome<br />

proliferatorinduced hepatocarcinogenesis may be readily identified. For<br />

example, if hepatocytes are transformed by either oxidative stress-induced<br />

damage or by alternative mechanisms, such initiated cells may be promoted<br />

to liver tumours by enhanced cell replication. Certainly peroxisome<br />

proliferators are effective promoters <strong>of</strong> certain populations <strong>of</strong> initiated cells<br />

and recent studies suggest that peroxisome proliferators can influence rates<br />

<strong>of</strong> both cell replication and cell death in particular populations <strong>of</strong><br />

hepatocytes (Grasl-Kraupp et al., 1993; Popp and Cattley, 1993; Marsman<br />

and Popp, 1994).<br />

With respect to species differences, rats and mice are clearly responsive<br />

species, whereas the majority <strong>of</strong> both in vivo and in vitro studies suggest<br />

that primates including man are either essentially refractory or certainly<br />

much less responsive to rodent peroxisome proliferators. However while<br />

effects on peroxisome morphology and marker enzyme activities have been<br />

extensively studied, few investigations have examined species differences in<br />

peroxisome proliferator-induced cell replication and liver tumour<br />

formation. As enhanced cell replication appears to play a role in peroxisome<br />

proliferator-induced hepatocarcinogenesis in rats and mice, it would


appear to be an important biomarker for assessing species differences in<br />

response. Rodent peroxisome proliferators do not appear to stimulate<br />

replicative DNA synthesis in vivo in Syrian hamster hepatocytes and in<br />

vitro in human hepatocytes (Elcombe and Styles 1989; Parzefall et al.,<br />

1991; Price et al., 1992; Lake et al., 1993). With respect to tumour<br />

formation, nafenopin and Wy-14,643 (two potent peroxisome<br />

proliferators) were reported not to produce liver lesions in the Syrian<br />

hamster although both compounds produced liver nodules and<br />

hepatocellular carcinoma after 60 weeks in the rat (Lake et al., 1993).<br />

Similarly, cl<strong>of</strong>ibrate was reported not to increase liver weight or produce<br />

liver tumours in marmosets after 6.5 years treatment (Tucker and Orton,<br />

1993) and in an ongoing study cipr<strong>of</strong>ibrate was found not to produce any<br />

morphological changes in marmoset liver after 3 years administration<br />

(Graham et al., 1994).<br />

In conclusion, the present literature suggests that rodent peroxisome<br />

proliferators are non-genotoxic agents which should be assessed differently<br />

from genotoxic compounds for human hazard (Weisburger, 1994).<br />

Assessment <strong>of</strong> likely human exposure and compound potency are also<br />

important factors together with information on compound no effect levels<br />

and evidence <strong>of</strong> species differences in response. Rodent liver peroxisome<br />

proliferators as a class <strong>of</strong> chemicals thus do not appear to pose any serious<br />

hazard for man. However, it would be desirable to elucidate further the<br />

mechanism(s) <strong>of</strong> peroxisome proliferator-induced hepatocarcinogenesis in<br />

susceptible species (i.e. the rat and mouse). From such studies the most<br />

appropriate biomarkers <strong>of</strong> liver tumour formation could be identified and<br />

examined in studies <strong>of</strong> species differences possibly including in vitro studies<br />

with human hepatocytes. Finally, further carcinogenicity studies in partially<br />

responsive (e.g. Syrian hamster) and non-responsive (e.g. guinea pig)<br />

species would strengthen the conclusion that peroxisome proliferators do<br />

not constitute any significant hazard to man.<br />

Acknowledgement<br />

We thank the UK Ministry <strong>of</strong> Agriculture, Fisheries and Food for financial<br />

support <strong>of</strong> BIBRA studies on hepatic peroxisome proliferation.<br />

References<br />

B.G.LAKE AND R.J.PRICE 231<br />

ALBRO, P.W., CORBETT, J.T., SCHROEDER, J.L. and JORDAN, S.T., 1983,<br />

Incorporation <strong>of</strong> radioactivity from labelled di-(2-ethylhexyl)phthalate into<br />

DNA <strong>of</strong> rat liver in vivo. Chemico-Biological Interactions, 44, 1–16.<br />

BARBER, E.D., ASTILL, B.D., MORAN, E.J., SCHNEIDER, B.F., GRAY, T.J. B.,<br />

LAKE, B.G. and EVANS, J.G., 1987, Peroxisome induction studies on seven<br />

phthalate esters, <strong>Toxicology</strong> and <strong>Industrial</strong> Health, 3, 7–22.


232 PEROXISOME PROLIFERATION<br />

BENTLEY, P., CALDER, I., ELCOMBE, C., GRASSO, P., STRINGER, D. and<br />

WIEGAND, H.-J., 1993, Hepatic peroxisome proliferation in rodents and its<br />

significance for humans, Food and Chemical <strong>Toxicology</strong>, 31, 857–907.<br />

BIEBER, L.L., KRAHLING, J.B., CLARKE, P.R.H., VALKNER, K.J. and<br />

TOLBERT, N.E., 1981, Carnitine acyltransferases in rat liver peroxisomes,<br />

Archives <strong>of</strong> Biochemistry and Biophysics, 211, 599–604.<br />

BIERI, F., 1993, Cultured hepatocytes: a useful in vitro system to investigate effects<br />

induced by peroxisome proliferators and their species specificity, in Gibson, G.<br />

and Lake, B. (Eds) Peroxisomes: Biology and Importance in <strong>Toxicology</strong> and<br />

Medicine, pp. 299–311, London: Taylor and Francis.<br />

BUDROE, J.D. and WILLIAMS, G.M. 1993, Genotoxicity studies <strong>of</strong> peroxisome<br />

proliferators, in Gibson, G. and Lake, B. (Eds) Peroxisomes: Biology and<br />

Importance in <strong>Toxicology</strong> and Medicine, pp. 525–68, London: Taylor and<br />

Francis.<br />

CATTLEY, R.C. and POPP, J.A., 1989, Differences between the promoting<br />

activities <strong>of</strong> the peroxisome proliferator WY-14,643 and phenobarbital in rat<br />

liver, Cancer Research, 49, 3246–51.<br />

CATTLEY, R.C., MARSMAN, D.S. and POPP, J.A., 1991, Age-related<br />

susceptibility to the carcinogenic effect <strong>of</strong> the peroxisome proliferator WY-14,<br />

643 in rat liver, Carcinogenesis, 12, 469–73.<br />

COHEN, S.M. and ELLWEIN, L.B., 1990, Cell proliferation in carcinogenesis,<br />

Science, 249, 1007–11.<br />

COHEN, S.M. and ELLWEIN, L.B., 1991, Genetic errors, cell proliferation and<br />

carcinogenesis, Cancer Research, 51, 6493–505.<br />

COHEN, A.J. and GRASSO, P., 1981, Review <strong>of</strong> the hepatic response to<br />

hypolipidaemic drugs in rodents and assessment <strong>of</strong> its toxicological<br />

significance to man, Food and Chemical <strong>Toxicology</strong>, 19, 585–605.<br />

DIRVEN, H.A.A.M., VAN DEN BROEK, P.H.H., PETERS, J.G.P.,<br />

NOORDHOEK, J. and JONGENEELEN, F.J., 1992, Microsomal lauric acid<br />

hydroxylase activities after treatment <strong>of</strong> rats with three classical cytochrome<br />

P-450 inducers and peroxisome proliferating compounds, Biochemical<br />

Pharmacology, 43, 2621–9.<br />

EACHO, P.I., FOXWORTHY, P.S., JOHNSON, W.D., HOOVER, D.M. and<br />

WHITE, S.L., 1986, Hepatic peroxisomal changes induced by a<br />

tetrazolesubstituted alkoxyacetophenone in rats and comparison with other<br />

species, <strong>Toxicology</strong> and Applied Pharmacology, 83, 430–7.<br />

EACHO, P.I., LANIER, T.L. and BRODHECKER, C.A., 1991, Hepatocellular<br />

DNA synthesis in rats given peroxisome proliferating agents: comparison <strong>of</strong><br />

Wy-14,643 to cl<strong>of</strong>ibric acid, nafenopin and LY 171883, Carcinogenesis, 12,<br />

1557– 61.<br />

ELCOMBE, C.R., 1985, Species differences in carcinogenicity and peroxisome<br />

proliferation due to trichloroethylene: a biochemical human hazard assessment,<br />

Archives <strong>of</strong> <strong>Toxicology</strong>, Supplement 8, 6–17.<br />

ELCOMBE, C.R. and MITCHELL, A.M., 1986, Peroxisome proliferation due to di<br />

(2-ethylhexyl)phthalate (DEHP): species differences and possible mechanisms,<br />

Environmental Health Perspectives, 70, 211–19.<br />

ELCOMBE, C.R. and STYLES, J.A., 1989, Species differences in peroxisome<br />

proliferation, Toxicologist, 9, 63.


B.G.LAKE AND R.J.PRICE 233<br />

FOXWORTHY, P.S. and EACHO, P.I., 1994, Cultured hepatocytes for studies <strong>of</strong><br />

peroxisome proliferation: methods and applications, Journal <strong>of</strong><br />

Pharmacological and Toxicological Methods, 31, 21–30.<br />

GIBSON, G.G., 1989, Comparative aspects <strong>of</strong> the mammalian cytochrome P450IV<br />

gene family, Xenobiotica, 19, 1123–48.<br />

GOEL, S.K., LALWANI, N.D., FAHL, W.E. and REDDY, J.K., 1985, Lack <strong>of</strong><br />

covalent binding <strong>of</strong> peroxisome proliferators nafenopin and WY-14,643 to<br />

DNA in vivo and in vitro, <strong>Toxicology</strong> Letters, 24, 37–43.<br />

GRAHAM, M.J., WILSON, S.A., WINHAM, M.A., SPENCER, A.J., REES, J.A.,<br />

OLD, S.L. and BONNER, F.W., 1994, Lack <strong>of</strong> peroxisome proliferation in<br />

marmoset liver following treatment with cipr<strong>of</strong>ibrate for 3 years, Fundamental<br />

and Applied <strong>Toxicology</strong>, 22, 58–64.<br />

GRASL-KRAUPP, B., HUBER, W. and SCHULTE-HERMANN, R., 1993, Are<br />

peroxisome proliferators tumour promoters in rat liver? in Gibson, G. and<br />

Lake, B. (Eds) Peroxisomes: Biology and Importance in <strong>Toxicology</strong> and<br />

Medicine, pp. 667–93, London: Taylor and Francis.<br />

GRAY, T.J.B., BEAMAND, J.A., LAKE, B.G., FOSTER, J.R. and GANGOLLI, S.D.,<br />

1982, Peroxisome proliferation in cultured rat hepatocytes produced by<br />

cl<strong>of</strong>ibrate and phthalate ester metabolites, <strong>Toxicology</strong> Letters, 10, 273–9.<br />

GUPTA, R.C., GOEL, S.K., EARLEY, K., SINGH, B. and REDDY, J.K., 1985, 32 P-<br />

Postlabelling analysis <strong>of</strong> peroxisome proliferator-DNA adduct formation in rat<br />

liver in vivo and hepatocytes in vitro, Carcinogenesis, 6, 933–6.<br />

HANEFELD, M., KEMMER, C. and KADNER, E., 1983, Relationship between<br />

morphological changes and lipid-lowering action <strong>of</strong> pchlorophenoxyisobutyric<br />

acid (CPIB) on hepatic mitochondria and<br />

peroxisomes in man, Atherosclerosis, 46, 239–46.<br />

HARTIG, F., STEGMEIER, K., HEBOLD, G.,ÖZEL, M. and FAHIMI, H.D.,<br />

1982, Study <strong>of</strong> liver enzymes: peroxisome proliferation and tumor rates in rats<br />

at the end <strong>of</strong> carcinogenicity studies with bezafibrate and cl<strong>of</strong>ibrate, Annals <strong>of</strong><br />

the New York Academy <strong>of</strong> Science, 386, 464–7.<br />

HAWKINS, J.M., JONES, W.E., BONNER, F.W. and GIBSON, G.G., 1987, The<br />

effect <strong>of</strong> peroxisome proliferators on microsomal, peroxisomal and<br />

mitochondrial enzyme activities in the liver and kidney, Drug Metabolism<br />

Reviews, 18, 441–515.<br />

HESS, R., STÄUBLI, W. and REISS, W., 1965, Nature <strong>of</strong> the hepatomegalic effect<br />

produced by ethyl chlorophenoxyisobutyrate in the rat, Nature, 208, 856–8.<br />

HINTON, R.H. and PRICE, S.C., 1993, Extrahepatic peroxisome proliferation and<br />

the extrahepatic effects <strong>of</strong> peroxisome proliferators, in Gibson, G. and Lake,<br />

B. (Eds) Peroxisomes: Biology and Importance in <strong>Toxicology</strong> and Medicine,<br />

pp. 487– 511, London: Taylor and Francis.<br />

HWANG, J.-J., HSAI, M.T.S. and JIRTLE, R.L., 1993, Induction <strong>of</strong> sister<br />

chromatid exchange and micronuclei in primary cultures <strong>of</strong> rat and human<br />

hepatocytes by the peroxisome proliferator Wy-14,643, Mutation Research,<br />

286, 123–33.<br />

ISHII, H., FUKUMORI, N., HORIE, S. and SUGA, T., 1980, Eifects <strong>of</strong> fat content<br />

in the diet on hepatic peroxisomes <strong>of</strong> the rat, Biochimica et Biophysica Acta,<br />

617, 1–11.


234 PEROXISOME PROLIFERATION<br />

KRAUPP-GRASL, B., HUBER, W., TAPER, H. and SCHULTE-HERMANN, R.,<br />

1991, Increased susceptibility <strong>of</strong> aged rats to hepatocarcinogenesis by the<br />

peroxisome proliferator nafenopin and the possible involvement <strong>of</strong> altered<br />

liver foci occurring spontaneously, Cancer Research, 51, 666–71.<br />

LAKE, B.G., 1993, Role <strong>of</strong> oxidative stress and enhanced cell replication in the<br />

hepatocarcinogenicity <strong>of</strong> peroxisome proliferators, in Gibson, G. and Lake, B.<br />

(Eds) Peroxisomes: Biology and Importance in <strong>Toxicology</strong> and Medicine, pp.<br />

595– 618, London: Taylor and Francis.<br />

LAKE, B.G. and LEWIS, D.F.V., 1993, Structure-activity relationships for<br />

chemically induced peroxisome proliferation in rodent liver, in Gibson, G. and<br />

Lake, B. (Eds) Peroxisomes: Biology and Importance in <strong>Toxicology</strong> and<br />

Medicine, pp. 313– 42, London: Taylor and Francis.<br />

LAKE, B.G., GRAY, T.J.B., PELS RIJCKEN, W.R., BEAMAND, J.A. and<br />

GANGOLLI, S.D., 1984a, The effect <strong>of</strong> hypolipidaemic agents on peroxisomal<br />

β-oxidation and mixed-function oxidase activities in primary cultures <strong>of</strong> rat<br />

hepatocytes. Relationship between induction <strong>of</strong> palmitoyl-CoA oxidation and<br />

lauric acid hydroxylation, Xenobiotica, 14, 269–76.<br />

LAKE, B.G., GRAY, T.J.B., FOSTER, J.R., STUBBERFIELD, C.R. and<br />

GANGOLLI, S.D., 1984b, Comparative studies on di-(2-ethylhexyl)phthalateinduced<br />

hepatic peroxisome proliferation in the rat and hamster, <strong>Toxicology</strong><br />

and Applied Pharmacology, 72, 46–60.<br />

LAKE, B.G., GRAY, T.J.B. and GANGOLLI, S.D., 1986, Hepatic effects <strong>of</strong><br />

phthalate esters and related compounds—in vivo and in vitro correlations,<br />

Environmental Health Perspectives, 67, 283–90.<br />

LAKE, B.G., LEWIS, D.F.V. and GRAY, T.J.B., 1988, Structure-activity<br />

relationships for peroxisome proliferation, Archives <strong>of</strong> <strong>Toxicology</strong> (Supplement<br />

12), 217–24.<br />

LAKE, B.G., EVANS, J.G., GRAY, T.J.B., KÖRÖSI, S.A. and NORTH, C.J., 1989,<br />

Comparative studies on nafenopin-induced hepatic peroxisome proliferation in<br />

the rat, Syrian hamster, guinea pig and marmoset, <strong>Toxicology</strong> and Applied<br />

Pharmacology, 99, 148–60.<br />

LAKE, B.G., EVANS, J.G., CUNNINGHAME, M.E. and PRICE, R.J., 1993,<br />

Comparison <strong>of</strong> the hepatic effects <strong>of</strong> nafenopin and Wy-14,643 on peroxisome<br />

proliferation and cell replication in the rat and Syrian hamster, Environmental<br />

Health Perspectives 101 (Supplement 5), 241–8.<br />

LALWANI, N.D., REDDY, M.K., GHOSH, S., BARNARD, S.D., MOLELLO, J. A.<br />

and REDDY, J.K., 1985, Induction <strong>of</strong> fatty acid β-oxidation and peroxisome<br />

proliferation in the liver <strong>of</strong> Rhesus monkeys by DL-040, a new hypolipidemic<br />

agent, Biochemical Pharmacology, 34, 3473–82.<br />

LAZAROW, P.B. and DEDUVE, C., 1976, A fatty acyl-CoA oxidizing system in<br />

rat liver peroxisomes: enhancement by cl<strong>of</strong>ibrate, a hypolipidaemic drug,<br />

Proceedings <strong>of</strong> the National Academy <strong>of</strong> Sciences USA, 73, 2043–6.<br />

LIN, L.I.-K., 1987, The use <strong>of</strong> multivariate analysis to compare peroxisome<br />

induction data on phthalate esters in rats, <strong>Toxicology</strong> and <strong>Industrial</strong> Health, 3,<br />

25–47.<br />

LOCK, E.A., MITCHELL, A.M. and ELCOMBE, C.R., 1989, Biochemical<br />

mechanisms <strong>of</strong> induction <strong>of</strong> hepatic peroxisome proliferation, Annual Reviews<br />

<strong>of</strong> Pharma-cology and <strong>Toxicology</strong>, 29, 145–63.


B.G.LAKE AND R.J.PRICE 235<br />

LOFTUS, N.J., WOOLLEN, B.H., STEEL, G.T., WILKS, M.F. and CASTLE, L.,<br />

1994, An assessment <strong>of</strong> the dietary uptake <strong>of</strong> di-2-(ethylhexyl)adipate (DEHA)<br />

in a limited population study, Food and Chemical <strong>Toxicology</strong>, 32, 1–5.<br />

MAKOWSKA, J.M., GIBSON, G.G. and BONNER, F.W., 1992, Species<br />

differences in cipr<strong>of</strong>ibrate induction <strong>of</strong> hepatic cytochrome P450 4A1 and<br />

peroxisome proliferation, Journal <strong>of</strong> Biochemical <strong>Toxicology</strong>, 7, 183–91.<br />

MARSMAN, D.S. and POPP, J.A., 1994, Biological potential <strong>of</strong> basophilic<br />

hepatocellular foci and hepatic adenoma induced by the peroxisome<br />

proliferator, Wy-14,643, Carcinogenesis, 15, 111–17.<br />

MARSMAN, D.S., CATTLEY, R.C., CONWAY, J.G. and POPP, J.A., 1988,<br />

Relationship <strong>of</strong> hepatic peroxisome proliferation and replicative DNA<br />

synthesis to the hepatocarcinogenicity <strong>of</strong> the peroxisome proliferators di(2ethylhexyl)phthalate<br />

and [4-chloro-6-(2,3-xylidino)-2-pyrimidinylthio]acetic<br />

acid (Wy-14,643) in rats, Cancer Research, 48, 6739–44.<br />

Ministry <strong>of</strong> Agriculture, Fisheries and Food (MAFF), 1987, Survey <strong>of</strong> Plasticiser<br />

Levels in Food Contact Materials and in Foods, Food Surveillance Paper No.<br />

21, London: HMSO.<br />

Ministry <strong>of</strong> Agriculture, Fisheries and Food (MAFF), 1990, Survey <strong>of</strong> Plasticiser<br />

Levels in Food Contact Materials and in Foods, Food Surveillance Paper No.<br />

30, London: HMSO.<br />

MOODY, D.E., REDDY, J.K., LAKE, B.G., POPP, J.A. and REESE, D.H., 1991,<br />

Peroxisome proliferation and nongenotoxic carcinogenesis: commentary on a<br />

symposium, Fundamental and Applied <strong>Toxicology</strong>, 16, 233–48.<br />

NCI, 1976, Carcinogenesis <strong>of</strong> trichloroethylene (CAS No. 79–01–6), NCI-CG-<br />

TR-2, National Cancer Institute.<br />

NTP (National <strong>Toxicology</strong> Program), 1982a, Carcinogenesis bioassay <strong>of</strong> di(2-ethylhexyl)phthalate<br />

(CAS No. 117–81–7) in F344 rats and B6C3F1 mice (feed<br />

study), Technical Report Series No. 217.<br />

NTP (National <strong>Toxicology</strong> Program), 1982b, Carcinogenesis bioassay <strong>of</strong> di(2ethyl-<br />

hexyl)adipate (CAS No. 103–23–1) in F344 rats and B6C3F1 mice (feed<br />

study), Technical Report Series No. 212.<br />

ORTON, T.C., ADAM, H.K., BENTLEY, M., HOLLOWAY, B. and TUCKER,<br />

M. J., 1984, Clobuzarit: species differences in the morphological and<br />

biochemical response <strong>of</strong> the liver following chronic administration, <strong>Toxicology</strong><br />

and Applied Pharmacology, 73, 138–51.<br />

OSUMI, T. and HASHIMOTO, T., 1978, Enhancement <strong>of</strong> fatty acyl-CoA<br />

oxidizing activity in rat liver peroxisomes by di(2-ethylhexyl)phthalate,<br />

Journal <strong>of</strong> Biochemistry, 83, 1361–5.<br />

PAGET, G.E., 1963, Experimental studies <strong>of</strong> the toxicity <strong>of</strong> Atromid with<br />

particular reference to fine structural changes in the livers <strong>of</strong> rodents, Journal<br />

<strong>of</strong> Atherosclerosis Research, 3, 729–37.<br />

PARZEFALL, W., ERBER, E., SEDIVY, R. and SCHULTE-HERMANN, R., 1991,<br />

Testing for induction <strong>of</strong> DNA synthesis in human hepatocyte primary cultures<br />

by rat liver tumor promoters, Cancer Research, 51, 1143–7.<br />

POPP, J.A. and MARSMAN, D.S., 1991, Chemically-induced cell proliferation in<br />

liver carcinogenesis, in Butterworth, B.E., Slaga, T.J., Farland, W. and<br />

McClain, M. (Eds) Chemically Induced Cell Proliferation: Implications for<br />

Risk Assessment, pp. 389–95, New York: Wiley-Liss.


236 PEROXISOME PROLIFERATION<br />

POPP, J.A. and CATTLEY, R.C., 1993, Peroxisome proliferators as initiators and<br />

promoters <strong>of</strong> rodent hepatocarcinogenesis, in Gibson, G. and Lake, B. (Eds)<br />

Peroxisomes: Biology and Importance in <strong>Toxicology</strong> and Medicine, pp. 653–<br />

65, London: Taylor and Francis.<br />

PRICE, R.J., EVANS, J.G. and LAKE B.G., 1992, Comparison <strong>of</strong> the effects <strong>of</strong><br />

nafenopin on hepatic peroxisome proliferation and replicative DNA synthesis<br />

in the rat and Syrian hamster, Food and Chemical <strong>Toxicology</strong> 30, 937–44.<br />

REDDY, J.K. and LALWANI, N.D., 1983, Carcinogenesis by hepatic peroxisome<br />

proliferators: evaluation <strong>of</strong> the risk <strong>of</strong> hypolipidemic drugs and industrial<br />

plasticisers to humans, CRC Critical Reviews in <strong>Toxicology</strong>, 12, 1–58.<br />

REDDY, J.K. and RAO, M.S., 1989, Oxidative DNA damage caused by persistent<br />

peroxisome proliferation: its role in hepatocarcinogenesis, Mutation Research,<br />

214, 63–8.<br />

REDDY, J.K., LALWANI, N.D., QURESHI, S.A., REDDY, M.K. and MOEHLE,<br />

C.M., 1984, Induction <strong>of</strong> hepatic peroxisome proliferation in non-rodent<br />

species, including primates, American Journal <strong>of</strong> Pathology, 114, 171–83.<br />

REDDY, J.K., REDDY, M.K., USMAN, M.I., LALWANI, N.D. and RAO, M. S.,<br />

1986, Comparison <strong>of</strong> hepatic peroxisome proliferative effect and its<br />

implication for hepatocarcinogenicity <strong>of</strong> phthalate esters, di(2-ethylhexyl)<br />

phthalate and di(2-ethylhexyl)adipate with a hypolipidemic drug,<br />

Environmental Health Perspectives, 65, 317–27.<br />

RHODES, C.ORTON, T.C., PRATT, I.S., BATTEN, P.L., BRATT, H.,<br />

JACKSON, S.J. and ELCOMBE, C.R., 1986, Comparative pharmacokinetics<br />

and subacute toxicity <strong>of</strong> di(2-ethylhexyl)phthalate (DEHP) in rats and<br />

marmosets: extrapolation <strong>of</strong> effects in rodents to man, Environmental Health<br />

Perspectives, 65, 299–308.<br />

RODRICKS, J.V. and TURNBULL, D., 1987, Interspecies differences in<br />

peroxisomes and peroxisome proliferation, <strong>Toxicology</strong> and <strong>Industrial</strong> Health,<br />

3, 197–212.<br />

SCHULTE-HERMANN, R., TIMMERMANN-TROSIENER, I. and<br />

SCHLUPPLER, J., 1983, Promotion <strong>of</strong> spontaneous preneoplastic cells in rat<br />

liver as a possible expla nation <strong>of</strong> tumor production by nonmutagenic<br />

compounds, Cancer Research, 43, 839–44.<br />

SCHULTE-HERMANN, R., KRAUPP-GRASL, B., BURSCH, W., GERBRACHT,<br />

U. and TIMMERMANN-TROSIENER, I., 1989, Effects <strong>of</strong> non-genotoxic<br />

hepatocarcinogens phenobarbital and nafenopin on phenotype and growth <strong>of</strong><br />

different populations <strong>of</strong> altered foci in rat liver, Toxicologic Pathology, 17,<br />

642–50.<br />

SHARMA, R., LAKE, B.G., FOSTER, J. and GIBSON, G.G., 1988a, Microsomal<br />

cytochrome P-452 induction and peroxisome proliferation by hypolipidaemic<br />

agents in rat liver. A mechanistic inter-relationship, Biochemical<br />

Pharmacology, 37, 1193–201.<br />

SHARMA, R., LAKE, B.G. and GIBSON, G.G., 1988b, Co-induction <strong>of</strong><br />

microsomal cytochrome P-452 and the peroxisomal fatty acid β-oxidation<br />

pathway in the rat by cl<strong>of</strong>ibrate and di-(2-ethylhexyl)phthalate. Dose-response<br />

studies, Biochemical Pharmacology, 37, 1203–6.


B.G.LAKE AND R.J.PRICE 237<br />

STOTT, W.T., 1988, Chemically induced proliferation <strong>of</strong> peroxisomes:<br />

implications for risk assessment, Regulatory <strong>Toxicology</strong> and Pharmacology, 8,<br />

125–59.<br />

TAKAGI, A., SAI, K., UMEMURA, T., HASEGAWA, R. and KUROKAWA, Y.,<br />

1990, Significant increase <strong>of</strong> 8-hydroxydeoxyguanosine in liver DNA <strong>of</strong> rats<br />

following short-term exposure to the peroxisome proliferators di(2-ethylhexyl)<br />

phthalate and di(2-ethylhexyl)adipate, Japanese Journal <strong>of</strong> Cancer Research,<br />

81, 213–15.<br />

THORNBER, C.W., 1979, Isosterism and molecular modification in drug design,<br />

Chemical Society Reviews, 8, 563–80.<br />

TUCKER, M.J. and ORTON, T.C., 1993, Toxicological studies in primates with<br />

three fibrates, in Gibson, G. and Lake, B. (Eds) Peroxisomes: Biology and<br />

Importance in <strong>Toxicology</strong> and Medicine, pp. 425–47, London: Taylor and<br />

Francis.<br />

VON DÄNIKEN, A., LUTZ, W.K. and SCHLATTER, C., 1981, Lack <strong>of</strong> covalent<br />

binding to rat liver DNA <strong>of</strong> the hypolipidemic drugs cl<strong>of</strong>ibrate and fen<strong>of</strong>ibrate,<br />

<strong>Toxicology</strong> Letters, 7, 311–19.<br />

VON DÄNIKEN, A., LUTZ, W.K., JÄCKH, R. and SCHLATTER, C., 1984,<br />

Investi-gation <strong>of</strong> the potential for binding <strong>of</strong> di(2-ethylhexyl)phthalate (DEHP)<br />

and di(2-ethylhexyl)adipate (DEHA) to liver DNA in vivo, <strong>Toxicology</strong> and<br />

Applied Pharmacology, 73, 373–87.<br />

WADA, N., MARSMAN, D.S. and POPP, J.A., 1992, Dose-related effects <strong>of</strong><br />

hepatocarcinogen, Wy-14,643 on peroxisomes and cell replication,<br />

Fundamental and Applied <strong>Toxicology</strong>, 18, 149–54.<br />

WEISBURGER, J.H., 1994, Does the Delaney Clause <strong>of</strong> the U.S. Food and Drug<br />

Laws prevent human cancers? Fundamental and Applied <strong>Toxicology</strong>, 22, 483–<br />

93.


18<br />

Neurotoxicity Testing <strong>of</strong> <strong>Industrial</strong> <strong>Compounds</strong>:<br />

in vivo Markers and Mechanisms <strong>of</strong> Action<br />

KORNELIS J.VAN DEN BERG, 1 JAN-BERT<br />

P.GRAMSBERGEN, 2 ELISABETH M.G.HOOGENDIJK, 1<br />

JAN H.C.M.LAMMERS, 1 WILLEM S.SLOOT 1 and<br />

BEVERLY M.KULIG 1<br />

1 TNO Nutrition and Food Research Institute, Rijswijk; 2<br />

Erasmus University, Rotterdam<br />

Introduction<br />

Neurotoxicity assessment is designed to provide an answer to the question<br />

<strong>of</strong> whether or not a particular chemical is able to evoke some form <strong>of</strong><br />

adverse effect specifically associated with the nervous system. The<br />

development <strong>of</strong> risk assessment procedures is a long-term goal in view <strong>of</strong><br />

the complexity <strong>of</strong> the target organ involved, e.g. the nervous system. As<br />

risk identification is a first step in the risk assessment paradigm <strong>of</strong><br />

chemicals (NAS, 1983), for neuro-toxicity this translates at present into<br />

procedures and methods aimed at characterization <strong>of</strong> altered<br />

neurobehaviour <strong>of</strong> exposed experimental animals and abnormalities in the<br />

morphology <strong>of</strong> the nervous system. It has been suggested that risk<br />

assessment procedures may take more the approach <strong>of</strong> ‘exposuredoseresponse’<br />

(Andersen, 1991) in which mechanisms play an important role at<br />

various levels, e.g. from disposition <strong>of</strong> the chemical through the body to<br />

target tissues, via biochemical interactions at the molecular level to a toxic<br />

response as altered behaviour. Understanding the mechanisms involved in<br />

this sequence <strong>of</strong> events is helpful to arrive in the future at quantitative risk<br />

assessment procedures, for instance biologically-based modelling (Borgh<strong>of</strong>f<br />

et al., 1991). In addition, a better knowledge <strong>of</strong> the mechanistic principles<br />

<strong>of</strong> neuro-toxic agents may lead to the development <strong>of</strong> specific biomarkers<br />

that would further improve the efficiency <strong>of</strong> procedures for neurotoxicity<br />

screening, given the magnitude <strong>of</strong> the number <strong>of</strong> potentially neurotoxic<br />

compounds (NRC, 1992).<br />

Only for a small select group <strong>of</strong> industrial chemicals are mechanisms <strong>of</strong><br />

neurotoxic action more or less characterized. Perhaps the oldest examples<br />

are the class <strong>of</strong> organophosphate (OP) pesticides where the molecular<br />

targets have been identified as acetylcholinesterase (AChE) and neuropathy<br />

target esterase (NTE), the latter being associated with organophosphate-


induced delayed neuropathy (OPIDN) (Cherniack, 1988). This has led to<br />

investigations into structure-activity relationships <strong>of</strong> OPs with NTE in vitro<br />

that have proved to be valuable in predicting OPIDN (Davis et al. 1985).<br />

Furthermore, certain tissue culture systems, such as neuroblastoma cell<br />

lines, contain NTE and AChE activity that may be suited for identification<br />

<strong>of</strong> OPs causing OPIDN (Veronesi, 1992). Biochemical assays for NTE and<br />

AChE, in conjunction with neurobehavioural observations and<br />

neuropathology, are currently incorporated into US and Japanese<br />

neurotoxicity testing guidelines.<br />

A second group <strong>of</strong> industrial chemicals for which indications exist on<br />

their mechanism <strong>of</strong> action include certain compounds causing peripheral<br />

neuro-pathy such as acrylamide, carbon disulphide and n-hexane. It is<br />

generally assumed that the primary action <strong>of</strong> these chemicals is the<br />

crosslinking <strong>of</strong> axonal proteins (Graham et al., 1982), a process that blocks<br />

axonal transport (Sickles, 1991) and may lead to degeneration <strong>of</strong> distal<br />

axons (Spencer and Schaumberg, 1980). The basic information thus<br />

collected is yet to be developed into a useful biomarker.<br />

The pyrethroid insecticides represent a third group <strong>of</strong> chemicals for<br />

which the neurotoxic mechanism <strong>of</strong> action has been elucidated. The major<br />

symptoms <strong>of</strong> pyrethroid intoxication, e.g. convulsions, tremors, paralysis,<br />

are primarily the result <strong>of</strong> interaction <strong>of</strong> the pyrethroids with sodiumchannels<br />

on nerve membranes (Lund and Narahashi, 1982). Whereas<br />

under normal circumstances a sodium channel is only opened during a<br />

depolarization event, pyrethroids prolong opening <strong>of</strong> these channels<br />

thereby causing repetitive excitation <strong>of</strong> nerve and nerve terminals. In tissue<br />

culture systems, e.g. neuroblastoma cell lines, direct electrophysiological<br />

studies on cells have been done to investigate the effectiveness <strong>of</strong> different<br />

pyrethroids for opening <strong>of</strong> sodium channels (Oortgiesen et al., 1989).<br />

Biomarkers <strong>of</strong> neurotoxicity<br />

It is clear that a mechanistic understanding <strong>of</strong> the neurotoxicity <strong>of</strong><br />

suspected chemicals is and will be proceeding at a slow pace. Eventual<br />

utilization <strong>of</strong> this knowledge in the form <strong>of</strong> biological markers for<br />

neurotoxicity risk assessment procedures is, necessarily, a long-term<br />

objective. In the mean time alternative approaches have been proposed<br />

recently that (a) may provide biomarkers that could aid to further define<br />

underlying mechanisms and (b) are directly linked to neurotoxic<br />

mechanisms <strong>of</strong> actions.<br />

Gliotypic and neurotypic proteins<br />

K.J.VAN DEN BERG ET AL. 239<br />

Insults to the brain by a large variety <strong>of</strong> agents or conditions, e.g. viral<br />

infection, auto-immune encephalitis, trauma and chemicals, evoke a fairly


240 NEUROTOXICITY TESTING OF INDUSTRIAL COMPOUNDS<br />

stereo typic response <strong>of</strong> astrocytes (Eng, 1988). The normally rather<br />

quiescent astrocytes change into reaction astrocytes with a characteristic<br />

morphological appearance, a process known as reactive gliosis or<br />

astrogliosis. It has been proposed that this fairly universal astrocytic<br />

reaction might be a useful parameter for risk identification purposes<br />

(O’Callaghan, 1992; Rosengren and Haglid, 1989). The hypertrophic<br />

response <strong>of</strong> astrocytes is biochemically characterized by a greatly enhanced<br />

synthesis <strong>of</strong> glial fibrillary acidic protein (GFAP), a major structural protein<br />

<strong>of</strong> intermediate filaments (Eng, 1988). Under a variety <strong>of</strong> experimentallyinduced<br />

insults to the central nervous system, increased GFAP<br />

immunoreactivity has consistently been found in association with neuronal<br />

damage (Eng, 1988; O’Callaghan and Miller, 1989). It must be kept in<br />

mind, however, that reactive astrogliosis represents an indirect indication<br />

<strong>of</strong> nerve cell damage or loss. A strategy has been proposed (Brock and<br />

O’Callaghan, 1987) to collect quantitative data on astrogliosis, for<br />

example for GFAP concentrations, in association with information on<br />

changes <strong>of</strong> neuron-specific proteins. e.g. synapsin I or synaptophysin. The<br />

general idea is that enhanced GFAP levels in combination with decreased<br />

synaptophysin concentrations in the brain would be strongly indicative <strong>of</strong> a<br />

neurotoxic event. Recent methodological developments have led to<br />

quantitative procedures for assessment <strong>of</strong> GFAP and synaptophysin in<br />

nerve tissues by dot-blot immunoassay and ELISA (Jahn et al. 1984;<br />

O’Callaghan, 1991).<br />

Cerebral calcium accumulation<br />

Nerve cells, like most animal cells, possess a complicated system<br />

comprising calcium gates, pumps and channels to maintain free cytosolic<br />

calcium ion levels within a low physiological range (Pounds, 1990). Under<br />

pathological conditions, including hypoxia-ischaemia and status<br />

epilepticus, calcium overload in vulnerable neurons has been observed that<br />

is thought to be associated with the process <strong>of</strong> cell death (both necrosis and<br />

apoptosis) (Boobis et al. 1989; Siesjo and Bengtsson, 1989). Cerebral<br />

calcium accumulation has, therefore, been proposed as a potential index <strong>of</strong><br />

brain pathology (Korf et al., 1986).<br />

Free radical formation<br />

Free radicals may be involved in mechanisms <strong>of</strong> toxicity, including neurotoxicity<br />

<strong>of</strong> chemicals and are gaining more attention as is obvious from a<br />

number <strong>of</strong> recent reviews on this topic (LeBel and Bondy, 1991; Halliwell<br />

et al. 1992; Aust et al., 1993). Basically, free radicals are molecules that<br />

contain one or more unpaired electrons, whereas most molecules are<br />

nonradicals. Because <strong>of</strong> the unpaired electron(s) free radicals are highly


eactive chemical intermediates. Once a free radical is formed it can initiate<br />

a chain <strong>of</strong> reactions as the free electron is passed from one molecule to the<br />

other. In vivo, free radicals involving oxygen species are continuously<br />

produced and evolution has also provided the body with defence<br />

mechanisms such as superoxide dismutase (SOD), catalase, and scavengers<br />

such as glutathione, ascorbate, etc.<br />

Oxidative stress by free radicals may be an important neuropathological<br />

mediator following exposure to a number <strong>of</strong> neurotoxic agents (LeBel and<br />

Bondy, 1991). Examples <strong>of</strong> neurotoxic chemicals suspected <strong>of</strong> causing free<br />

radicals include chlordecone, ethanol, methamphetamine, methyl mercury,<br />

toluene, triethyl lead and trimethyltin. Of special interest is a ‘designer’<br />

drug called MPTP 1 causing destruction <strong>of</strong> dopaminergic neurons <strong>of</strong> the<br />

basal ganglia with symptoms similar to Parkinson’s disease. It is the<br />

neurotoxic metabolite MPP +2 that has been found to induce cerebral<br />

oxygen radical formation in vitro. There is also suggestive evidence to<br />

indicate that free radical scavengers may provide protection <strong>of</strong> the basal<br />

ganglia against neuro-toxic effects <strong>of</strong> MPP + (LeBel and Bondy, 1991). The<br />

occurrence <strong>of</strong> these kinds <strong>of</strong> compounds has led, among other things, one<br />

to suspect possible involvement <strong>of</strong> environmental chemicals and factors<br />

associated with diet and/or lifestyle in Parkinson’s disease (Russell, 1992;<br />

Semchuk et al., 1993).<br />

Model neurotoxins<br />

In order to validate an approach based on changes in these proposed<br />

biomarkers experimental studies were performed using various model<br />

neuro-toxicants, including trimethyltin, kainic acid, heavy metals such as<br />

lead, methylmercury and manganese, and developmental neurotoxicants,<br />

e.g. polychlorinated biphenyls.<br />

Trimethyltin<br />

K.J.VAN DEN BERG ET AL. 241<br />

Trimethyltin (TMT) is known to cause in adult rats a rather selective<br />

neuronal degeneration in specific regions <strong>of</strong> the brain, notably in limbic<br />

structures such as the hippocampus where extensive loss <strong>of</strong> pyramidal cells<br />

in CA fields are observed by standard histopathological procedures<br />

(O’Callaghan, 1988). A single systemic dose <strong>of</strong> TMT (7.5 mg kg −1 ) given to<br />

adult rats caused, after a period <strong>of</strong> 3 weeks, an approximately three-fold<br />

enhanced level <strong>of</strong> GFAP in hippocampus (Figure 18.1, upper left panel). In<br />

a number <strong>of</strong> other brain regions, e.g. different parts <strong>of</strong> the cortex,<br />

thalamus, striatum, cerebellum and brain stem, no significant changes in<br />

GFAP were observed. Assessment <strong>of</strong> synaptophysin, a structural protein <strong>of</strong><br />

synaptic vesicles <strong>of</strong> neurons (Jahn et al. 1985) in the same brain regions is<br />

given in Figure 18.1 (lower left panel). TMT induced a significant


242 NEUROTOXICITY TESTING OF INDUSTRIAL COMPOUNDS<br />

Figure 18.1 Changes in cerebral glial fibrillary acidic protein (GFAP) and<br />

synaptophysin concentrations by trimethyltin (TMT) and kainic acid. Results are<br />

expressed as mean±SEM. Open bars, control; closed bars, 7.5 mg trimethyltin<br />

(TMT) per kg or 12 mg kainic acid per kg; HP, hippopcampus; AM, amygdala; *,<br />

p


Kainic acid<br />

K.J.VAN DEN BERG ET AL. 243<br />

Kainic acid (KA), a glutamate agonist, is a potent model neurotoxin that<br />

causes neurodegeneration in a number <strong>of</strong> limbic structures including<br />

hippocampus, amygdala, piriform cortex (Sperk et al. 1983; Ben-Ari,<br />

1985). It is generally assumed that the mechanism <strong>of</strong> action involves<br />

interaction <strong>of</strong> KA with a special class <strong>of</strong> glutamate receptors and release <strong>of</strong><br />

endogenous excitatory amino acids in levels that are detrimental to<br />

neurons (Meldrum and Garthwaite, 1990).<br />

Kainic acid did induce, in a single systemic dose (12 mg kg −1 ), highly<br />

increased concentrations <strong>of</strong> GFAP in a number <strong>of</strong> target structures. For<br />

instance in hippocampus and amygdala GFAP levels did rise to 650 and<br />

960 per cent <strong>of</strong> control levels (Figure 18.1, upper right panel). Recent<br />

results from this laboratory have revealed that in a time-course study (Van<br />

den Berg and Gramsbergen, 1993) maximum levels are obtained after 4<br />

weeks that remained highly elevated in most target brain regions for at<br />

least a period <strong>of</strong> 6 months (Figure 18.2). The quantitative data on GFAP<br />

concentrations were supported by increased GFAP immunoreactivity in<br />

hippocampal sections visualized by immunohistochemical procedures. In<br />

addition to the damage in the hippocampus, permanently enhanced GFAP<br />

levels were found in other brain regions, e.g. piriform cortex, septum<br />

(Gramsbergen and Van den Berg, 1994) known to be targets <strong>of</strong> KA<br />

neurotoxicity.<br />

Synaptophysin levels were significantly reduced by KA in hippocampus<br />

and amygdala to a comparable degree (Figure 18.1, lower right panel).<br />

These data indicate a decrease in synaptophysin content encountered in<br />

brain regions where neuronal elements are known to be lost by these<br />

model neurotoxins. The magnitude <strong>of</strong> the changes in synaptophysin<br />

concentrations were much smaller than those <strong>of</strong> GFAP in the same brain<br />

structures. This may be explained by the fairly selective neuronal loss in<br />

specific layers <strong>of</strong>, for instance, the hippocampus, by TMT and KA. The<br />

effect is thus rather diluted in a biochemical procedure. Once an effect is<br />

scored, more detailed biochemical analysis is possible by using a punch<br />

technique after microdissection <strong>of</strong> brain nuclei (Palkovits and Brownstein,<br />

1988). Alternatively, a follow-up by histopathological procedures, for<br />

example by the cupric silver degeneration stain, would provide further<br />

details <strong>of</strong> neuronal damage (O’Callaghan and Jensen, 1992). In the<br />

experiments described above there was no clear correlation between the<br />

graded regional GFAP response and decrease <strong>of</strong> synaptophysin<br />

concentration. This may suggest a differential region-specific response <strong>of</strong><br />

astrocytes towards neuronal injury.<br />

In our laboratory cerebral calcium accumulation was determined<br />

recently after a single systemic dose <strong>of</strong> KA (12 mg kg −1 , i.p.), given to adult<br />

rats. A rapid uptake <strong>of</strong> 45 Ca was observed in various regions <strong>of</strong> the brain. A


244 NEUROTOXICITY TESTING OF INDUSTRIAL COMPOUNDS<br />

Figure 18.2 Time-course <strong>of</strong> GFAP concentration in hippocampus by kainic acid. Rats<br />

were treated with a single systemic dose <strong>of</strong> kainic acid (12 mg kg −1 , i.p.). GFAP was<br />

determined by ELISA at the times indicated. Results are expressed as mean±SEM<br />

(from Van den Berg and Gramsbergen, 1993, with permission).<br />

time-course study, covering a period <strong>of</strong> 6 months, has indicated that in the<br />

hippocampus a peak <strong>of</strong> 45 Ca uptake occurred already after 4 days and<br />

normal values were reached only after another 2–4 months (Van den Berg<br />

and Gramsbergen, 1993). More recent results indicate that other limbic<br />

areas display similar kinetics, while 45 Ca uptake in striatum and cortex<br />

return faster to normal values, e.g. within 2 weeks. Only in the thalamus a<br />

long-term sustained 45 Ca uptake was present for a period <strong>of</strong> 6 months. In<br />

general a good correlation was found between the regions showing<br />

sustained 45 Ca accumulation and those known to be targets <strong>of</strong> KA<br />

neurotoxicity. When these data on 45 Ca accumulation were related to<br />

effects <strong>of</strong> KA on GFAP concentrations, also a good agreement was<br />

observed concerning the brain regions involved (Gramsbergen and Van den<br />

Berg, 1994). Histopathological examination <strong>of</strong> hippocampal sections<br />

revealed extensive neurodegeneration by KA in CA1, CA3 and CA4<br />

regions (Van den Berg and Gramsbergen, 1993).<br />

Heavy metals<br />

Lead may produce in children symptoms <strong>of</strong> acute encephalopathy after<br />

exposure to high doses and at lower dose levels learning disorders and


K.J.VAN DEN BERG ET AL. 245<br />

hyperactive behaviour. In adults neurotoxicity is more <strong>of</strong>ten encountered<br />

as peripheral neuropathy after chronic occupational exposure to lead<br />

(Marsh, 1985). A number <strong>of</strong> mechanisms have been implicated in lead<br />

neurotoxicity (Bressler and Goldstein, 1991), but as yet no central<br />

hypothesis has emerged.<br />

In order to investigate the effect <strong>of</strong> lead on CNS structural proteins, a<br />

subchronic dosing experiment with adult rats was performed in which<br />

animals received daily doses <strong>of</strong> lead acetate (4, 8, 12.5 mg kg −1 i.p.) for 28<br />

days. GFAP concentrations were subsequently determined in different brain<br />

regions. Already at the lowest dose level GFAP levels were found to be<br />

significantly increased in several brain regions, notably in different parts <strong>of</strong><br />

the cortex, hippocampus and striatum, while cerebellum and brain stem<br />

remained unaffected. Neurobehavioural assessment <strong>of</strong> animals, preceding<br />

the neurochemical analysis, also revealed significant alterations <strong>of</strong><br />

neuromuscular function, excitability and spontaneous activity.<br />

Methylmercury has caused a number <strong>of</strong> poisonings in man (Marsh, 1985)<br />

where it appears to affect in particular both the central and peripheral<br />

nervous system. In an animal experiment, adult rats were subchronically<br />

dosed with methylmercury (0.75 or 2 mg kg −1 ) for 28 days.<br />

Neurobehavioural assessment indicated that grip strength was significantly<br />

impaired. Neurochemical analysis <strong>of</strong> GFAP in the central nervous system<br />

was performed in selected brain regions and, in addition, in various<br />

segments <strong>of</strong> the spinal cord. Increased GFAP levels were observed in the<br />

cerebrum only in the frontal cortex, also in brain stem and in spinal cord. A<br />

further detailed analysis <strong>of</strong> brain stem sub-structures showed significantly<br />

enhanced GFAP levels in pons and medulla oblongata but not in midbrain.<br />

In spinal cord GFAP concentration was increased in specific<br />

sections, e.g. in the cervical and lumbar segments but not in the thoracic<br />

segment.<br />

The results with these particular examples <strong>of</strong> heavy metals have indicated<br />

the unsuspected presence <strong>of</strong> regions in the central nervous system with<br />

astrogliosis, as determined in a biochemical GFAP assay. The neuronal<br />

damage involved has not yet been confirmed independently, e.g. by<br />

assessment <strong>of</strong> synaptophysin. The possibilities remain, therefore, that<br />

reactive astrocytosis by these heavy metals may be indirectly a result <strong>of</strong><br />

breaching the integrity <strong>of</strong> the blood-brain barrier (Bressler and Goldstein,<br />

1991) or <strong>of</strong> a direct toxic action on astroglial cells (Selvin Testa et al.,<br />

1990; Stark et al., 1992).<br />

Manganese is a well recognized industrial neurotoxin associated with<br />

neurologic effects after prolonged exposure in occupational settings (Katz,<br />

1985). The clinical manifestations <strong>of</strong> manganism bear a large similarity to<br />

those <strong>of</strong> Parkinson’s disease (PD). The neurodegenerative disease PD is<br />

characterized by a selective loss <strong>of</strong> neurons in the basal ganglia.


246 NEUROTOXICITY TESTING OF INDUSTRIAL COMPOUNDS<br />

Experimental studies were done to investigate the vulnerability <strong>of</strong> the<br />

basal ganglia in manganism. Manganese (as Mn 2+ ) was applied<br />

intrastriatally to rats and region-specific brain damage was assessed by<br />

determining regional 45 Ca accumulation using quantitative<br />

autoradiographic procedures developed in this laboratory (Gramsbergen<br />

and Van der Sluijs-Gelling, 1993). It appeared that Mn 2+ induced a timedependent<br />

45 Ca accumulation in most <strong>of</strong> the regions constituting the basal<br />

ganglia, e.g. striatum and several other structures such as globus pallidus,<br />

entopeduncular nucleus, several thalamic nuclei and substantia nigra (Sloot<br />

et al., 1994).<br />

As monoaminergic neurotransmission plays a predominant role in the<br />

basal ganglia, recent studies have indicated that concentrations <strong>of</strong> biogenic<br />

amines such as dopamine and its major metabolites, serotonin and<br />

noradrenaline were also reduced in striatum by Mn 2+ . The kinetics <strong>of</strong> this<br />

process indicated that concentrations <strong>of</strong> most monoamines and metabolites<br />

were temporarily reduced except for dopamine and metabolites in striatum<br />

that remained at a permanently reduced level (>90 days) (Sloot et al.,<br />

1994). In order to demonstrate the specificity <strong>of</strong> the manganese effects,<br />

several other compounds were studied, including ferrous ions (Fe 2+ ). An<br />

equimolar dose <strong>of</strong> Fe 2+ , applied intrastriatally, produced, however, a much<br />

more extensive and widespread 45 Ca accumulation throughout the basal<br />

ganglia and, in addition, in nucleus accumbens and cerebral cortex.<br />

Ferrous ions were also three times more potent than manganese ions in<br />

causing depletion <strong>of</strong> dopamine in striatum (Sloot et al., 1994).<br />

The results based on both 45 Ca accumulation and biogenic amine levels<br />

are in concordance with the hypothesis that the basal ganglia, which are<br />

enriched in iron and iron-binding proteins, represent a selective target for<br />

manganese. The role and fate <strong>of</strong> endogenous iron in the brain, and the<br />

basal ganglia in particular, under toxic conditions including chronic<br />

manganese exposure, merits further investigations.<br />

Oxidative stress by free radical formation may play a role in these toxic<br />

events. Transition metals are known as strong promoters <strong>of</strong> reactive<br />

oxygen species. Especially iron, as the ferrous ion (Fe 2+ ), has been found to<br />

react with hydrogen peroxide to form the hydroxyl radical in the so-called<br />

Fenton reaction (Halliwell et al., 1992; Aust et al., 1993). It is thought that<br />

this mechanism plays an important direct role in iron poisoning (Aust et<br />

al., 1993). In an indirect way oxidative stress by iron may be initiated by<br />

chemicals that are able to ‘liberate’ iron from stores such as ferritin,<br />

transferrin, haemoglobin, etc. Recently, evidence has been obtained that a<br />

number <strong>of</strong> chemicals, including the pesticides paraquat and diquat, may<br />

release iron from ferritin in vivo as well as in vitro (Aust et al., 1993),<br />

involving organic radical and superoxide formation.<br />

These observations are particularly relevant for the interpretation <strong>of</strong> the<br />

observed neurotoxic effects <strong>of</strong> iron described above. Dopamine is relatively


easily subjected to a process <strong>of</strong> auto-oxidation. The decrease in dopamine<br />

levels by iron may have been caused by oxygen radicals. Circumstantial<br />

evidence suggests that a similar mechanism <strong>of</strong> action may underly<br />

manganese neurotoxicity. The decrease in dopamine in the basal ganglia by<br />

manganese is also thought to occur through catalysis <strong>of</strong> dopamine<br />

oxidation (LeBel and Bondy, 1991), possibly involving radical oxygen<br />

species. An open question is whether manganese might participate in the<br />

‘iron release’ hypothesis (Sloot and Gramsbergen, 1994). Current efforts<br />

are being made to determine free radical formation by iron and manganese<br />

and to relate this to dopamine depletion. For this purpose rats are given<br />

salicylic acid (SA) and subsequently the SA hydroxylation products are<br />

measured in cerebrospinal fluid and brain tissues as indices <strong>of</strong> hydroxyl<br />

radical formation.<br />

Developmental neurotoxins (PCBs)<br />

K.J.VAN DEN BERG ET AL. 247<br />

Concern has been raised about the long term consequences <strong>of</strong> low level<br />

intake <strong>of</strong> polychlorinated biphenyls (PCBs) with respect to neurotoxicity as<br />

it relates to nervous tissue development and intellectual performance in the<br />

juvenile and adult stages. Several epidemiological studies with infants have<br />

shown a negative correlation between PCB levels in cord blood and<br />

cognitive functions and a positive correlation between PCB levels and<br />

altered neurological parameters such as hypotonia and hyporeflexia (Rogan<br />

et al., 1988; Jacobson et al., 1990). Experimental studies in various species<br />

including primates have also provided arguments for neurotoxic effects in<br />

<strong>of</strong>fspring after perinatal exposure to PCBs (Tilson et al., 1990). In this<br />

laboratory evidence has recently been obtained to indicate dramatic<br />

reduction in sexual behaviour and reproduction in <strong>of</strong>fspring that was<br />

perinatally exposed to PCBs (Smits-van Proojie et al., 1993).<br />

In order to investigate eventual structural alterations in the CNS,<br />

pregnant Wistar WU rats were exposed to Aroclor 1254 on days 10–16 <strong>of</strong><br />

gestation. At a young age (3 weeks) and adult age (3 months) <strong>of</strong>fspring<br />

were sacrificed and various brain regions were dissected for assessment <strong>of</strong><br />

gliotypic and neurotypic proteins. In untreated control animals<br />

developmental aspects <strong>of</strong> astrocytes in the central nervous system were<br />

encountered. Both in hypothalamus and cerebellum <strong>of</strong> control animals<br />

GFAP levels were increased by 200–300 per cent between 3 weeks and 3<br />

months postnatally. A developmental GFAP increase was also found in<br />

brain stem, striatum and lateral olfactory tract, although to a lesser extent.<br />

In hippocampus and prefrontal cortex, GFAP levels remained virtually<br />

unchanged between 3 weeks and 3 months. These results, therefore,<br />

indicate that glial cell maturation and/or differentiation is not uniformly<br />

distributed over the whole brain. It appeared that glial cells at birth in rats<br />

were fully developed in brain regions dealing with cognitive functions, e.g.


248 NEUROTOXICITY TESTING OF INDUSTRIAL COMPOUNDS<br />

cortex and hippocampus, while in regions such as hypothalamus and<br />

cerebellum this process occurred entirely neonatally. It should be kept in<br />

mind that in the rat also neuronal maturation and differentiation are not<br />

completed at birth and continue to a large extent postnatally.<br />

Exposure <strong>of</strong> pregnant rats to Aroclor 1254 did lead to a number <strong>of</strong><br />

alterations in GFAP levels in various brain regions in <strong>of</strong>fspring as compared<br />

to control animals. The most striking differences were observed in brain<br />

stem. While in control animals <strong>of</strong> both sexes GFAP levels increased<br />

postnatally, the normal developmental increase in perinatally exposed<br />

animals was absent (Figure 18.3, upper panel). At 3 months the relative<br />

deficit in GFAP levels was 41 per cent for male and 30 per cent for female<br />

progeny. Already at the lowest dose <strong>of</strong> Aroclor 1254 (5 mg/kg) a maximum<br />

decrease was observed (Figure 18.3, upper panel). In addition to brain stem,<br />

a similar effect on GFAP levels was found in striatum although the relative<br />

deficit at 3 months was somewhat less. Quite an opposite pattern emerged<br />

in brain regions such as cerebellum, lateral olfactory tract and prefrontal<br />

cortex, where GFAP levels were increased relative to unexposed progeny.<br />

In hippocampus no significant changes in GFAP levels were encountered.<br />

The question arose whether the observed neurodevelopmental toxicity in<br />

rats by PCBs was specific for astroglial cells or also involved neuronal<br />

maturation, differentiation and death. For this purpose the same nervous<br />

tissues were used for quantitative assessment <strong>of</strong> a neuronal marker in the<br />

form <strong>of</strong> synaptophysin. In brains <strong>of</strong> untreated adult control rats, regionspecific<br />

differences were observed in synaptophysin concentrations, being<br />

high in the prefrontal cortex, striatum and hippocampus and relatively low<br />

in lateral olfactory tract, cerebellum and brain stem. Synaptogenesis for the<br />

brain as a whole largely takes place in the rat from birth until postnatal<br />

day 70 (Knaus et al., 1986). Regional differences in the speed <strong>of</strong> postnatal<br />

synaptogenesis from 3 weeks to 3 months were found in a number <strong>of</strong><br />

structures being most pronounced in cerebellum (190 per cent increase) and<br />

prefrontal cortex (170 per cent increase) while in brain stem little change was<br />

observed.<br />

As a result <strong>of</strong> perinatal exposure to Aroclor 1254, altered expression <strong>of</strong><br />

synaptophysin was observed. In most brain structures examined, including<br />

brainstem (Figure 18.3, lower panel) significant decreases in synaptophysin<br />

concentrations were found. This suggests that during development <strong>of</strong> the<br />

central nervous system, PCBs may interfere with the formation <strong>of</strong> synaptic<br />

vesicles, synaptogenesis, or formation <strong>of</strong> nerve terminals.<br />

A straightforward interpretation <strong>of</strong> the present results is not possible at<br />

this stage. However, what is clear is that perinatal exposure to PCBs may<br />

cause changes in the structural composition <strong>of</strong> the central nervous system<br />

both in the neuronal and the glial cell compartment. Apparently there are<br />

different effects on nerve cells <strong>of</strong> the CNS depending on the brain region<br />

involved. The brain stem and striatum are regions with decreased


K.J.VAN DEN BERG ET AL. 249<br />

Figure 18.3 Developmental effects on glial fibrillary acidic protein (GFAP) and<br />

synaptophysin concentrations in brain stem after perinatal exposure to Aroclor<br />

1254. Results are expressed as mean±SEM with the values <strong>of</strong> the control animals at<br />

day 21 set at 100 per cent. Open bars, control; shaded bars, 5 mg Aroclor 1254 per<br />

kg; closed bars, 25 mg Aroclor 1254 per kg; D 21 and D 90, postnatal days; *, p


250 NEUROTOXICITY TESTING OF INDUSTRIAL COMPOUNDS<br />

Table 18.1 Cerebral calcium accumulation<br />

a Systemic dose.<br />

b Intrastriatal dose.<br />

synaptophysin and increased GFAP in structures such as cerebellum, lateral<br />

olfactory tract, and prefrontal cortex. Such a situation could arise when<br />

neurons in these latter regions fail to receive proper input from developing<br />

neurons originating in brain stem and striatum. Further investigations into<br />

this interesting topic may provide further clues for the developmental<br />

toxicity <strong>of</strong> PCBs and related compounds that possibly could aid in the<br />

interpretation <strong>of</strong> neurobehavioural effects, for instance altered sexual<br />

behaviour and reproduction (Smits-van Prooije et al., 1993).<br />

The findings observed in cerebellum are consistent with a phase <strong>of</strong><br />

hypothyroidism during development but it is clear that a conclusive role<br />

for thyroid hormone remains to be further established. The present results<br />

furthermore suggest that alterations in synaptophysin/GFAP levels may be<br />

useful and sensitive parameters to study compounds suspected <strong>of</strong><br />

developmental neurotoxicity.<br />

Conclusion<br />

The results with various neurotoxins demonstrate that assessment <strong>of</strong><br />

gliotypic proteins such as GFAP may be a useful tool to identify and<br />

quantify persistent toxic insults <strong>of</strong> the CNS, especially when this is backed<br />

up by indications for loss <strong>of</strong> neuronal elements, e.g. decreased<br />

synaptophysin concentration. Also in circumstances <strong>of</strong> developmental<br />

neurotoxicity, caused by chemicals such as PCBs, an approach based on<br />

changes <strong>of</strong> gliotypic and neurotypic proteins may provide a promising<br />

biomarker. Of course, further investigations with various other compounds<br />

are required to substantiate these interesting findings.<br />

Cerebral calcium accumulation may be useful as an early indicator <strong>of</strong><br />

neurotoxicity on a more prospective basis as is suggested by various<br />

examples <strong>of</strong> neurotoxic compounds (summarized in Table 18.1). Because in<br />

unlesioned brain regions the background levels <strong>of</strong> calcium uptake remain


very low, areas where neurotoxic events take place are easily identified and<br />

quantified by autoradiographic or scintillation counting procedures.<br />

Finally, the role <strong>of</strong> oxidative stress by free radical formation in the<br />

nervous system merits further studies since it may open up possibilities for<br />

providing a biomarker that is linked to a mechanism <strong>of</strong> neurotoxic action.<br />

Acknowledgements<br />

The author is grateful for unpublished information provided by Dr Didema<br />

de Groot on neuropathology, by Dennis C.Morse, MSc and the students<br />

Wendelien Wesseling and Annemiek Plug, Agricultural University<br />

Wageningen, on developmental effects <strong>of</strong> PCBs; and for expert technical<br />

assistance by Alita van der Sluijs-Gelling.<br />

References<br />

K.J.VAN DEN BERG ET AL. 251<br />

ANDERSEN, M.E., 1991, Abstract, Eurotox Congress, Maastricht.<br />

AUST, S.D., CHIGNELL, C.F., BRAY, T.M., KALYANARAMAN, B. and<br />

MASON, R.P., 1993, Free radicals in toxicology, Toxicol. Appl. Pharmacol.,<br />

120, 168–78.<br />

BEN-ARI, Y., 1985, Limbic seizure and brain damage produced by kainic acid:<br />

mechanisms and relevance to human temporal lobe epilepsy, Neuroscience, 14,<br />

375–403.<br />

BOOBIS, A .R., FAWTHROP, D.J. and DAVIES, D.S., 1989, Mechanisms <strong>of</strong> cell<br />

death, Trends Pharmacol. Sci., 10, 275–80.<br />

BORGHOFF, S., GARGAS, M.L., ANDERSEN, M.E. and CONOLLY, R.B., 1991,<br />

Development <strong>of</strong> a biologically based response model for 2,4,4-trimethyl-2pentanol<br />

induced alfa-2u-globulin nephropathy, Toxicologist, 11, 138.<br />

BRESSLER, J.P. and GOLDSTEIN, G.W., 1991, Mechanisms <strong>of</strong> lead<br />

neurotoxicity, Biochem. Pharmacol., 41, 479–84.<br />

BROCK, T.O. and O’CALLAGHAN, J.P., 1987, Quantitative changes in the<br />

synaptic vesicle proteins synapsin I and p38 and the astrocyte-specific protein<br />

glial fibrillary acidic protein are associated with chemical-induced injury to the<br />

rat central nervous system, J. Neurosci., 7, 931–42.<br />

CHERNIACK, M.G., 1988, Toxicological screening for organophosphorusinduced<br />

delayed neurotoxicity: complications in toxicity testing,<br />

Neurotoxicology, 9, 249– 72.<br />

DAVIS, C.S., JOHNSON, M.K. and RICHARDSON, R.J., 1985,<br />

Organophosphorus compounds, in O’Donoghue, J.L. (Ed.) Neurotoxicity <strong>of</strong><br />

<strong>Industrial</strong> and Commercial <strong>Compounds</strong>, p. 1–24, Boca Raton, Florida: CRC<br />

Press.<br />

ENG, L.F., 1988, Regulation <strong>of</strong> glial intermediate filaments in astrogliosis, in<br />

Norenberg, M.D., Hertz, L. and Schoesboe, A. (Eds) The Biochemical<br />

Pathology <strong>of</strong> Astrocytes, pp. 79–90, New York: Alan R Liss.<br />

GRAHAM, D.G., ANTHONY, D.C., BOELKELHEIDE, K., MADSCHMANN,<br />

N.A., RICHARDS, R.G., WOLFRAM, J.W. and SHAW, B.R., 1982, Studies <strong>of</strong>


252 NEUROTOXICITY TESTING OF INDUSTRIAL COMPOUNDS<br />

the molecular pathogenesis <strong>of</strong> hexane neuropathy. II. Evidence that pyrrole<br />

derivatization <strong>of</strong> lysyl residues leads to protein crosslinking, Toxicol. Appl.<br />

Pharmacol, 64, 415–22.<br />

GRAMSBERGEN, J.B.P. and VAN DER SLUIJS-GELLING, A.J., 1993, Time- and<br />

dose-dependent 45 Ca 2+ accumulationinrat striatum and substantia nigra after<br />

an intrastriatal injection <strong>of</strong> quinolinic acid, Exp. Neurol., 121, 261–9.<br />

GRAMSBERGEN, J.B.P. and VAN DEN BERG, K.J., 1994, Regional and<br />

temporal pr<strong>of</strong>iles <strong>of</strong> calcium accumulation and glial fibrillary acidic protein<br />

levels in rat brain after systemic injection <strong>of</strong> kainic acid, Brain Res., 667, 216–<br />

28.<br />

HALLIWELL, B., GUTTERIDGE, J.M.C. and CROSS, C.E., 1992, Free radicals,<br />

antioxidants, and human disease: where are we now? J. Lab. Clin. Med., 119,<br />

598–620.<br />

JACOBSON, J.L., JACOBSON, S.W. and HUMPHREY, H.E., 1990, Effects <strong>of</strong> in<br />

utero exposure to polychlorinated biphenyls and related contaminants on<br />

cognitive functioning in young children, J. Pediatr., 116, 38–45.<br />

JAHN, R., SCHIEBLER, W. and GREENGARD, P., 1984, A quantitative<br />

dotimmunobinding assay for proteins using nitrocellulose membrane filters,<br />

Proc. Nat. Acad. Sci. USA, 81, 1684–7.<br />

JAHN, R., SCHIEBLER, W., OUIMET, C. and GREENGARD, P., 1985, A 38000<br />

dalton protein (p-38) present in synaptic vesicles, Proc. Nat. Acad. Sci. USA,<br />

82, 4137–41.<br />

KATZ, G.V., 1985, Metals and metalloids other than mercury and lead, in<br />

O’Donoghue, J.L. (Ed.) Neurotoxicity <strong>of</strong> <strong>Industrial</strong> and Commercial<br />

Chemicals, I, pp. 171–92, Boca Raton, Florida: CRC Press.<br />

KNAUS, P., BETZ, H. and REHM, H., 1986, Expression <strong>of</strong> synaptophysin during<br />

postnatal development <strong>of</strong> the mouse brain, J. Neurochem., 47, 1302–4.<br />

KORF, J., GRAMSBERGEN, J.B.P., PRENEN, G.H.M. and Go, K.G.,<br />

1986, Cation shifts and excitotoxins in Alzheimer and Huntington disease and<br />

experimental brain damage, in Swabb, D.F., Fliers, E., Mirmiran, M., Van<br />

Gool, W.A. and Van Harren, F. (Eds) Progress in Brain Research Vol. 70, pp.<br />

213–226. Amsterdam: Elsevier.<br />

LEBEL, C.P. and BONDY, S.C., 1991, Oxygen radicals: common mediators <strong>of</strong><br />

neurotoxicity, Neurotoxicol. Teratol., 13, 341–6.<br />

LUND, A.E. and NARAHASHI, T., 1982, Dose-dependent interaction <strong>of</strong> the<br />

pyrethroid isomers with sodium channels <strong>of</strong> squid axon membranes,<br />

Neurotoxicology, 3, 11–24.<br />

MARSH, D.O., 1985, The neurotoxicity <strong>of</strong> mercury and lead, in O’Donoghue, J.L.<br />

(Ed.) Neurotoxicity <strong>of</strong> <strong>Industrial</strong> and Commercial Chemicals, I, pp. 159–91,<br />

Boca Raton, Florida: CRC Press.<br />

MAYER, M.L. and MILLER, R.J., 1990, Excitatory amino acid receptors, second<br />

messengers and regulation <strong>of</strong> intracellular Ca in mammalian neurons, Trends<br />

Pharmacol Sci., 11, 254–60.<br />

MELDRUM, B. and GARTHWAITE, J., 1990, Excitatory amino acid<br />

neurotoxicity and neurodegenerative disease, Trends Pharmacol. Sci., 11, 379–<br />

87.


K.J.VAN DEN BERG ET AL. 253<br />

NATIONAL ACADEMY OF SCIENCES(NAS), 1983, Risk Assessment in Federal<br />

Government: Managing the Process, Washington, DC. National Academic<br />

Press.<br />

NATIONAL RESEARCH COUNCIL (NRC), 1992, Environmental<br />

Neurotoxicology, Washington, DC. National Academic Press.<br />

O’CALLAGHAN, J.P., 1988, Neurotypic and gliotypic proteins as biochemical<br />

markers <strong>of</strong> neurotoxicity, Neurotoxicol. Teratol, 10, 445–52.<br />

O’CALLAGHAN, J.P., 1991, Quantification <strong>of</strong> glial fibrillary acidic protein:<br />

comparison <strong>of</strong> slot-immunobinding assays with a novel sandwich ELISA,<br />

Neurotoxicol. Teratol., 13, 275–81.<br />

O’CALLAGHAN, J.P., 1992, Assessment <strong>of</strong> neurotoxicity using assays <strong>of</strong> neuroand<br />

glia-localized proteins: chronology and critique, in Tilson, H. and<br />

Mitchell, C. (Eds) Neurotoxicology, pp. 83–100. New York: Raven Press.<br />

O’CALLAGHAN, J.P. and MILLER, D.B., 1989, Assessment <strong>of</strong> chemically-induced<br />

alterations in brain development using assays <strong>of</strong> neuro- and glia-localized<br />

proteins, Neurotoxicology, 10, 393–406.<br />

O’CALLAGHAN, J.P. and JENSEN, K.F., 1992, Enhanced expression <strong>of</strong> glial<br />

fibrillary acidic protein and the cupric silver degeneration reaction can be used<br />

as sensitive and early indicators <strong>of</strong> neurotoxicity, Neurotoxicology, 13, 113–<br />

22.<br />

OORTGIESEN, M., VAN KLEEF, R. and VIJVERBERG, H.P.M., 1989, Effects Of<br />

pyrethroids on neurotransmitter operated ion channels in cultured mouse<br />

neuroblastoma cells, Pest. Biochem. Physiol, 34, 164–73.<br />

PALKOVITS, M. and BROWNSTEIN, M.J., 1988, Maps and Guide to<br />

Microdissection <strong>of</strong> the Rat Brain, New York: Elsevier.<br />

POUNDS, J.G., 1990, The role <strong>of</strong> cell calcium in current approaches to toxicology,<br />

Environm. Hlth Perspect., 84, 7–15.<br />

ROGAN, W.J., GLADEN, B.C., HUNG, K.L., KOONG, S.L., SHIH, L.Y.,<br />

TAYLOR, J.S., WU, Y.C. et al., (1988), Congenital poisoning by<br />

polychlorinated biphenyls and their contaminants in Taiwan, Science, 241,<br />

334–6.<br />

ROSENGREN, L.E. and HAGLID, K.G., 1989, Long term neurotoxicity <strong>of</strong> styrene.<br />

A quantitative study <strong>of</strong> glial fibrillary acidic protein (GFA) and S-100, Br. J.<br />

Ind. Med., 46, 316–20.<br />

RUSSELL, T., 1992, Environmental risk factors in Parkinson’s disease, Food<br />

Chem. Toxicol., 30, 343–7.<br />

SELVIN TESTA, A., LOPEZ COSTA, J.J., NESSI DE AVINON, A.C. and PECCI<br />

SAAVEDRA, J., 1991, Astroglial alterations in rat hippocampus during<br />

chronic lead exposure, GLIA, 4, 384–92.<br />

SEMCHUK, K.M., LOVE, E.J. and LEE, R.G., 1993, Parkinson’s disease: a test <strong>of</strong><br />

the multifactorial etiologic hypothesis, Neurology, 43, 1173–80.<br />

SICKLES, D.W., 1991, Toxic neur<strong>of</strong>ilamentous axonopathies and fast anterograde<br />

axonal transport. III. Recovery from single injections and multiple dosing<br />

effects <strong>of</strong> acrylamide and 2,5-hexanedione, Toxicol. Appl. Pharmacol, 108,<br />

390–6.<br />

SIESJO, B.K. and BENGTSSON, F., 1989, Calcium fluxes, calcium antagonists,<br />

and calcium-related pathology in brain ischemia, hypoglycemia, and spreading<br />

depression: a unifying hypothesis, J. Cereb. Blood Flow Metab., 9, 127–140.


254 NEUROTOXICITY TESTING OF INDUSTRIAL COMPOUNDS<br />

SLOOT, W.N. and GRAMSBERGEN, J.B.P., 1994, Axonal transport <strong>of</strong><br />

manganese and its relevance to selective neurotoxicity in the rat basal ganglia,<br />

Brain Res., 657, 124–32.<br />

SLOOT, W.N., VAN DER SLUIJS-GELLING, A.J. and GRAMSBERGEN, J.B.P.,<br />

1994, Selective lesions by manganese and extensive damage by iron after<br />

injection into rat striatum or hippocampus, J. Neurochem., 62, 205–16.<br />

SMITS-VAN PROOIJE, LAMMERS, J.H.C.M., WAALKENS-BERENDSEN,<br />

D.H., KULIG, B.M. and SNOEIJ, N.J., 1993, Effects <strong>of</strong> the PCB 3,4,5,3′,4′,5′hexachlorobiphenyl<br />

on the reproduction capacity <strong>of</strong> Wistar rats,<br />

Chemosphere, 27, 395– 400.<br />

SPENCER, P.S. and SCHAUMBERG, H.H., 1980, Experimental and Clinical<br />

Neurotoxicology, Baltimore: Williams and Wilkins.<br />

SPERK, G., LASSMAN, H., BARAN, H., KISH, S.J., SEITELBERGER, F. and<br />

HORNYKIEWICZ, O., 1983, Kainic acid induced seizures: neurochemical and<br />

histopathological changes, Neuroscience, 10, 1301–15.<br />

STARK, M., WOLFF, J.E. and KORBMACHER, A., 1992, Modulation <strong>of</strong> glial cell<br />

differentiation by exposure to lead and cadmium, Neurotoxicol. Teratol, 14,<br />

247–52.<br />

TILSON, H.A., JACOBSON, J.L. and ROGAN, W.J., 1990, Polychlorinated<br />

biphenyls and the developing nervous system: cross-species comparisons,<br />

Neuro-toxicol. Teratol, 12, 239–48.<br />

VAN DEN BERG, K.J. and GRAMSBERGEN, J.B.P., 1993, Long-term changes in<br />

glial fibrillary acidic protein and calcium levels in rat hippocampus after a<br />

single systemic dose <strong>of</strong> kainic acid, Ann. N.Y. Acad. Sci., 679, 394–401.<br />

VERONESI, B., 1992, In vitro screening batteries for neurotoxicants,<br />

Neurotoxicology, 13, 185–96.


19<br />

Endocrine <strong>Toxicology</strong> <strong>of</strong> the Thyroid for<br />

<strong>Industrial</strong> <strong>Compounds</strong><br />

CHRISTOPHER K.ATTERWILL and SAMUEL<br />

P.AYLWARD<br />

CellTox Centre, University <strong>of</strong> Hertfordshire, Hatfield, Herts<br />

General introduction<br />

Classification <strong>of</strong> endocrine toxicity<br />

Xenobiotic-induced endocrine dysfunction and toxicity is a common<br />

finding in safety studies and an increasingly important consideration in the<br />

riskassessment process. The effective identification <strong>of</strong> potential endocrine<br />

toxicological effects depends upon xenobiotic effect classification in<br />

relation to normal endocrine function and pathology.<br />

Classifications <strong>of</strong> different types <strong>of</strong> endocrine toxicity have been<br />

proposed in previous publications on this subject. Capen and Martin<br />

(1989) proposed a detailed classification based on clinical endocrine<br />

function and pathology. On the other hand, Baylis and Tunbridge (1985)<br />

proposed a simpler classification based on the adverse endocrine reactions<br />

<strong>of</strong> xenobiotics which are observed clinically. From a toxicological or<br />

preclinical safety testing point <strong>of</strong> view, Atterwill and Flack (1993) favoured<br />

classifying endocrine toxicology <strong>of</strong> xenobiotics in a manner which is<br />

similar in concept to that for classifying other toxicological phenomena<br />

(CIOMS, 1983) taking into account the unique nature <strong>of</strong> the endocrine<br />

system. This is as follows (see also Figure 19.1):<br />

Class 1 Effects which can be predicted from the endocrine pharmacology<br />

<strong>of</strong> compounds. An example would be the oestrogens and progestogens<br />

which have a plethora <strong>of</strong> effects on metabolic parameters in addition to their<br />

actions on oestrogen sensitive target sites when administered at<br />

pharmacological doses (the therapeutic dose levels). Further examples are<br />

certain dopamine and 5-HT antagonists acting on hypothalamic and<br />

pituitary receptors which may disrupt ‘downstream’ endocrine functions.<br />

Class 2 Effects which again can be predicted from the endocrine<br />

pharmacology <strong>of</strong> the compound when administered at doses well in excess<br />

<strong>of</strong> the therapeutic dose level. An example would be adrenal steroid<br />

suppression and general excessive catabolism observed with high dose and


256 ENDOCRINE TOXICOLOGY OF THE THYROID<br />

Figure 19.1 Classification <strong>of</strong> endocrine toxicity (Atterwill and Flack, 1993).<br />

prolonged use <strong>of</strong> glucocorticoids. In addition, the use <strong>of</strong> thyromimetic agents<br />

which may suppress pituitary thyrotroph function.<br />

Class 3 Effects which could not have been predicted from the<br />

pharmacology <strong>of</strong> the compound. This group can be subclassified into: (a)<br />

Effects which are direct or primary actions on an endocrine gland.<br />

Examples <strong>of</strong> this might be the action <strong>of</strong> ketoconazoles on adrenal and<br />

testicular function and the action <strong>of</strong> alloxan and streptozocin on the β-cell<br />

<strong>of</strong> the pancreas; and (b) effects on endocrine glands which are indirect or<br />

secondary to changes in other organs or control mechanisms<br />

(homeostasis). Examples here would be the actions <strong>of</strong> phenobarbitone and<br />

PCBs on the rat thyroid and the effects <strong>of</strong> lactose and polyols on the rat<br />

adrenal medulla.<br />

Class 4 Effects which cannot be predicted from pre-clinical studies<br />

because <strong>of</strong> idiosyncratic effects on the endocrine system.<br />

As indicated by Baylis and Tunbridge (1985) the adverse effects <strong>of</strong><br />

pharmaceutical agents on the endocrine system are generally due to normal


C.K.ATTERWILL AND S.P.AYLWARD 257<br />

or exaggerated pharmacological responses, that is Classes 1 and 2.<br />

Furthermore, it appears that endocrine toxicology can be detected reliably<br />

in pre-clinical studies. What is essential is that Class 3 toxic endocrine effects<br />

are classified appropriately. Class 3a effects would be expected to be seen<br />

across species, whereas there are numerous examples where Class 3b<br />

effects appear to be species-specific. There seems to be a paucity <strong>of</strong> clear-cut<br />

examples for Class 4 effects. There are several examples <strong>of</strong> compounds<br />

which have idiosyncratic immunotoxicological effects in susceptible humans<br />

which cannot be predicted from pre-clinical studies.<br />

Incidence <strong>of</strong> thyroid toxicity and tumours <strong>of</strong> the thyroid<br />

Most information on incidence is derived from pharmaceutical toxicity<br />

databases. Such data from Ribelin (1984) suggest that the endocrine system<br />

<strong>of</strong> the rats is particularly sensitive to toxicity from xenobiotics. This is also<br />

supported by Heywood (1984) in which he examined the target organ<br />

toxicity for 42 pharmaceutical compounds in the rat and dog. The<br />

endocrine system <strong>of</strong> the rat was only second to the liver as the most<br />

frequently affected target organ (38 per cent liver, 31 per cent endocrine).<br />

Ribelin (1984) reported that the most frequent endocrine lesion occurs in<br />

the adrenals followed by the testes but this analysis was conducted on<br />

chemicals and pharmaceuticals, and the data indicated that it was the<br />

cortical layers <strong>of</strong> the adrenal that were being predominantly effected,<br />

suggesting that the adrenal changes may be reflecting general stress<br />

responses rather than direct adrenal gland toxicity.<br />

In another analysis conducted in conjunction with the Centre <strong>of</strong><br />

Medicines Research this area was further explored. <strong>Toxicology</strong> data on<br />

124 compounds (all pharmaceuticals) were analysed. Just under 50 per<br />

cent (61/124) <strong>of</strong> these compounds have effects on one or more endocrine<br />

glands. Similar to Ribelin (1984) the adrenals were the most frequently<br />

affected, followed by the testes and the thyroid. An extensive survey <strong>of</strong> the<br />

different types <strong>of</strong> thyroid toxicity for both pharmaceutical agents and<br />

industrial chemicals was presented by Atterwill et al. (1993).<br />

Perturbation <strong>of</strong> thyroid function<br />

Thyroid function can be perturbed by agents affecting a number <strong>of</strong><br />

processes involved in the regulation <strong>of</strong> the hypothalamic-pituitary-thyroidliver<br />

(H-P-T-L) axis (Figure 19.2). These agents can affect function directly<br />

by interacting with thyroid cell receptors on their intracellular transduction<br />

mechanisms (see Figure 19.3). Alternatively thyroid function may be<br />

altered indirectly by agents affecting thyroid hormone metabolism and/or<br />

distribution—this event being followed by the release <strong>of</strong> thyrotrophic<br />

factors, or by xenobiotic-mediated alterations in the release <strong>of</strong> these factors


258 ENDOCRINE TOXICOLOGY OF THE THYROID<br />

Figure 19.2 Hypothalamic-pituitary-thyroid-liver (HPTL) axis.<br />

themselves from the hypothalamus or pituitary gland (see also review by<br />

Cavalieri and Pitt-Rivers (1981) and Atterwill et al. (1993)). For example,<br />

thyromimetics such as L-T 3 or D-T 3 can cause pituitary atrophy and<br />

thyroid ‘shutdown’ and toxicity due to suppressive effects on the<br />

thyrotrophs in the adenohypophysis (Atterwill, 1988).


Figure 19.3 Thyroid follicular epithelial cell function.<br />

C.K.ATTERWILL AND S.P.AYLWARD 259<br />

Control <strong>of</strong> thyroid function and pathobiology <strong>of</strong> thyroid<br />

lesions<br />

Xenobiotic toxic effects on the hypothalamic-pituitarythyroid-liver<br />

(H-P-T-L) axis<br />

The control <strong>of</strong> mammalian thyroid follicular function is shown<br />

schematically in Figure 19.2 together with the points at which agents may<br />

perturb function and cause toxicity and thyroid lesions. The most common<br />

classes <strong>of</strong> agent affecting function are discussed at the different loci in<br />

terms <strong>of</strong> the categories (1–4) <strong>of</strong> endocrine toxicity (Atterwill and Flack,<br />

1993).<br />

The various control mechanisms and factors influencing hormone<br />

synthesis, distribution and metabolism can be summarised as follows:<br />

thyroid hormone (T 3 and T 4) synthesis and secretion from thyroid gland<br />

are controlled by thyroid stimulating hormone (TSH) released from the<br />

pituitary gland. This in turn is under control by hypothalamic thyrotrophin<br />

releasing hormone (TRH) and circulating levels <strong>of</strong> the thyroid hormones.<br />

Thyroid hormones exist in the circulation in the free (free T 4 (FT 4) and<br />

free T 3 (FT 3)) and protein-bound forms (approx 99 per cent <strong>of</strong> total T 4-<br />

TT 4 and total T 3-TT 3) and it is the FT 3 hormone produced by deiodination<br />

from T 4 which has both a physiological action on T 3 nuclear receptors in<br />

target tissues and influences pituitary TSH output.


260 ENDOCRINE TOXICOLOGY OF THE THYROID<br />

Protein-binding <strong>of</strong> the hormone in the circulation takes place on several<br />

moieties, the pr<strong>of</strong>iles <strong>of</strong> which vary between different species (Dohler et al.,<br />

1973). The major proteins are thyroglobulin (TBG), thyroid binding<br />

pre albumin (TBPA) and albumin. Free and bound T 3 and T 4 are in<br />

dynamic equilibrium in the circulation and because <strong>of</strong> differences in affinity<br />

for the binding proteins there is more T 3 in the unbound form (approx 0.4<br />

per cent <strong>of</strong> total) than T 4 (approx 0.04 per cent <strong>of</strong> total).<br />

Thyroid hormone synthesis takes place at the apical membrane <strong>of</strong> the<br />

polarised follicular epithelial cells and depends on TSH-stimulated active<br />

iodide uptake under the influence <strong>of</strong> many extracellular and intracellular<br />

factors and signals (see Figure 19.3). Iodide is oxidised by peroxidase<br />

enzymes which mediate the incorporation <strong>of</strong> iodine into the tyrosyl<br />

residues <strong>of</strong> the colloidal glycoprotein, thyroglobulin. Colloid-bound<br />

thyroid hormone is stored in the follicular lumen and is released into the<br />

circulation by lysosomal action on the colloidal complex following<br />

endocytosis <strong>of</strong> colloid droplets into the cells. The incorporation and<br />

organification <strong>of</strong> iodide into thyroid hormone within thyroid follicles and<br />

the toxicological effects <strong>of</strong> xenobiotics can be studied in vivo using the<br />

‘perchlorate-discharge test’ (Atterwill et al., 1987) or in vitro using cultured<br />

thyrocytes (see Figure 19.4) from different species (Atterwill and Fowler,<br />

1990).<br />

Catabolic metabolism <strong>of</strong> the hormonal products <strong>of</strong> the thyroid gland, FT 3<br />

and FT 4, is achieved via two major pathways, deiodination and<br />

conjugation (glucuronidation and sulphation yielding a more water-soluble<br />

product for biliary excretion) and one minor route, deamination<br />

(decarboxylation, see Figure 19.9 later).<br />

The metabolic fate <strong>of</strong> thyroxine relies predominantly on its deiodination<br />

to T 3, only 20 per cent <strong>of</strong> circulating T 3 (the thyromimetic) is secreted by<br />

the thyroid (Engler and Burger, 1984). The remaining 80 per cent is derived<br />

from the deiodinative conversion <strong>of</strong> thyroxine (FT 4) to T 3. Deiodination<br />

can occur at several sites—the ones <strong>of</strong> major importance from the clearance<br />

aspect being liver and kidney whilst pituitary deiodination is essential for<br />

controlling responsiveness to circulating FT 4 levels. The deiodinases exist<br />

as three iso-zymes: Type I (5′-D; localised in liver, kidney, thyroid and<br />

central nervous system (CNS) tissue; and is propylthiouracil (PTU)<br />

sensitive); Type II (5'-D; localised exclusively in the CNS, brown adipose<br />

tissue, and pituitary; PTU-insensitive); and Type III (5-D, CNS; PTU<br />

insensitive) and have different affinities for T 4, different maturational<br />

patterns and different compensatory responses to hypothyroidism (see<br />

Kohrle et al., 1987).<br />

Bastomsky (1973), using Gunn rats, congenitally jaundiced due to UDPglucuronyl<br />

transferase deficiency (including T 4-glucuronyltransferase)<br />

demonstrated that the rate limiting step in hepatic thyroxine clearance (via


C.K.ATTERWILL AND S.P.AYLWARD 261<br />

the biliary excretion pathway) is the formation <strong>of</strong> the glucuronic acid<br />

conjugate, T 4-glucuronide.<br />

Hepatic conjugation, either sulphation (preferring FT 3) or<br />

glucuronidation (preferring FT 4) yields a more water soluble product,<br />

excreted in the bile/ biliary duct is a major pathway in T 4 excretion.<br />

Further deiodination via the deiodinase group <strong>of</strong> enzymes <strong>of</strong> T 4 T 3conjugates,<br />

T 4-amines and T 3 to thyromimetically inactive iodothyronines<br />

(rT 3 Triace, Tetrace, T 2 and T 1; see Figures 19.8 and 19.9) plays an<br />

important part in the T 4/T 3 biotransformation cascade and completes the<br />

thyroid hormone metabolic pr<strong>of</strong>ile.<br />

The key factor in maintaining correct thyroid follicular capability is an<br />

appropriate TSH output to TRH stimulation alongside circulating levels <strong>of</strong><br />

FT 4. Perturbation <strong>of</strong> this homeostatic control results in a classical thyroid<br />

response. The initial thyroid responses to increasing TSH levels are<br />

follicular cell hypertrophy, loss <strong>of</strong> colloid and vascular dilatation. In<br />

conventional animal toxicology studies performed for regulatory<br />

authorities one <strong>of</strong> the first indices <strong>of</strong> thyrotoxicity, therefore, is the<br />

observation <strong>of</strong> altered thyroid histopathology, primarily as follicular cell<br />

hypertrophy and/or diffuse hyperplasia, <strong>of</strong>ten leading to focal hyperplasia,<br />

thyroid adenomas and adenocarcinomas in longer term toxicity studies<br />

after longer term exposure.<br />

Pathobiology <strong>of</strong> thyroid follicular cell hyperplasia and<br />

neoplasia<br />

Thyroid neoplasia (see Figure 19.5) develops predictably in experimental<br />

species exposed to any procedure inducing prolonged and excessive TSH<br />

secretion (for example, the administration <strong>of</strong> chemical goitrogens, chronic<br />

iodine deficiency or subtotal thyroidectomy) although humans and mouse<br />

appear to be more resistant to TSH-induced thyroid neoplasia than rat.<br />

The number <strong>of</strong> cytogenetic abnormalities within the thyroid epithelium<br />

increases with duration <strong>of</strong> excess TSH exposure, with follicular cell<br />

hyperplasia potentially leading to neoplasia. The histopathological<br />

sequence <strong>of</strong> events is as follows (see Zbinden, 1987): following<br />

hypertrophy <strong>of</strong> the follicular epithelial cells focal hyperplasias appear in the<br />

gland which are distinct areas <strong>of</strong> papillary growth with enlarged epithelia.<br />

As these foci continue to grow they form nodules partly surrounded by<br />

collagenous fibres. These lesions are transition states between focal<br />

hyperplasias and adenomas. Adenomas are larger nodules that compress<br />

the surrounding tissue and have a distinct capsule. Follicular<br />

microcarcinomas (characterised by irregular gland-like structures,<br />

basophilia and nuclear crowding) may appear in some nodules. Larger<br />

carcinomas usually retain a follicular structure but sometimes consist <strong>of</strong><br />

solid sheets <strong>of</strong> polymorphous cells (Zbinden, 1987).


262 ENDOCRINE TOXICOLOGY OF THE THYROID<br />

Figure 19.4 Cultured porcine thyrocytes in vitro. These scanning electron<br />

micrographs show (a) individual cultured ‘inverted’ follicles, and (b) individual<br />

follicular epithelial cells at higher magnification (apical membrane facing upwards)<br />

displaying TSH-stimulated microvilli.


C.K.ATTERWILL AND S.P.AYLWARD 263<br />

Figure 19.5 Factors influencing the development <strong>of</strong> thyroid neoplasia.<br />

Two consecutive processes are thought to occur in the development <strong>of</strong><br />

thyroid tumours; first, initiation which occurs quickly and is irreversible<br />

and secondly, promotion which occurs slowly and is reversible (and for<br />

which cell proliferation may be a necessary but not sufficient condition).<br />

Initiators may be ionising radiation, chemical/biological agents or genetic<br />

factors, with TSH acting as a promotion agent. Spontaneous thyroid<br />

follicular cell tumours arise from unknown aetiologies and factors and ageinduced<br />

changes in the cell membrane, growth factor and signaltransduction<br />

mechanisms may be involved. Small subpopulations <strong>of</strong> hyperreactive<br />

epithelial cells retaining the high replication rate <strong>of</strong> the foetal stage<br />

have been identified (Peter et al., 1982; Smeds et al., 1987) which may<br />

enter clonal expansion following only slight elevations in TSH (such as<br />

those in handled or stressed animals) or other growth factors, leading to<br />

spontaneous nodular goitres (see Zbinden, 1987).<br />

Many experimental studies have confirmed the key role <strong>of</strong> TSH as a<br />

stimulator <strong>of</strong> thyroid growth. In a rat thyroid cell line (FRTL-5) TSHstimulated<br />

growth <strong>of</strong> the cells was found to be associated with a marked<br />

increase in c-fos and c-myc oncogene expression (Colletta et al., 1986).<br />

Another example <strong>of</strong> the tumour promoting capacity <strong>of</strong> TSH is given by


264 ENDOCRINE TOXICOLOGY OF THE THYROID<br />

studies where rats were given carcinogens such as N-methyl-N-nitrosourea<br />

(MNU) and then phenobarbital, or put on an iodine deficient diet. These<br />

treatments cause an early and increased incidence <strong>of</strong> thyroid follicular<br />

lesions and tumour formation (Hiasa et al., 1982; Oshima and Ward, 1984).<br />

The duration <strong>of</strong> exposure to high circulating TSH concentrations is also<br />

important in that intermittent administration <strong>of</strong> chemical goitrogens with<br />

TSH ‘normalisation’ does not appear to lead to follicular neoplasias.<br />

An elaborate series <strong>of</strong> studies have shown that a sustained elevation <strong>of</strong><br />

serum TSH in the rat leads to three phases <strong>of</strong> thyroid growth (Figure 19.6):<br />

(1) a phase <strong>of</strong> rapid growth lasting 1–2 months, followed by (2) a plateau<br />

phase <strong>of</strong> 3–6 months (growth desensitising mechanism (GDM) limiting<br />

epithelial cell mitotic response), followed eventually (3) by the appearance<br />

<strong>of</strong> multiple follicular cell tumours (loss <strong>of</strong> GDM; see Wynford-Thomas et<br />

al., 1982; Stringer et al., 1985; Smith et al., 1986). The reversibility <strong>of</strong> TSHinduced<br />

thyroid focal hyperplasia will evidently depend, therefore, on the<br />

stage during these ‘timed’ cellular changes in the first 6 months at which<br />

the TSH stimulus is withdrawn. Once the GDM is non-operative<br />

reversibility is not possible.<br />

Tumour progression seems to occur by a multi-stage process involving<br />

clonal ‘expansion’ and naturally occuring clones <strong>of</strong> cells have been<br />

demonstrated with high intrinsic proliferation potential in the mouse<br />

thyroid gland (Smeds et al., 1987), perhaps helping to explain the focal<br />

nature <strong>of</strong> hyperplastic and neoplastic lesions. The loss <strong>of</strong> a GDM within<br />

the follicular cells appears to be accompanied by an altered dependence or<br />

sensitivity to certain growth factors as well as the possible loss <strong>of</strong> an antioncogene<br />

which limits the follicular cells’s growth response to TSH. For<br />

example, the growth <strong>of</strong> normal cultured human thyroid cells requires TSH<br />

and insulin-like growth factor 1 (IGF1) in combination, whereas cells from<br />

adenomatous tissue in vitro proliferate in response to either TSH or IGF<br />

independently (Williams et al., 1987). This is due to the acquisition <strong>of</strong><br />

autocrine production <strong>of</strong> IGF 1 by the tumour cells themselves (see Thomas<br />

and Williams, 1991). Since the differentiation and growth <strong>of</strong> thyrocytes<br />

under TSH is regulated by cyclic-AMP-dependent mechanisms<br />

(Figure 19.2), tissue hyperplasia and hyperthyroidism might be expected to<br />

result when activation <strong>of</strong> the adenyl cyclase-cAMP cascade becomes<br />

unregulated. This can occur, for example, when somatic mutations impair<br />

the GTPase activity <strong>of</strong> G-protein coupled reactors, which may thus behave<br />

as proto-oncogenes. Such a mechanism is probably responsible for the<br />

development <strong>of</strong> a minority <strong>of</strong> monoclonal hyperfunctioning thyroid<br />

adenomas (Parma et al., 1993) (these also result in a silencing <strong>of</strong> normal<br />

thyroid function in extra-adenomatous tissue). Other non-genotoxic<br />

factors, such as agents affecting patterns <strong>of</strong> DNA methylation when<br />

coupled with a growth stimulus, should also be given consideration when


C.K.ATTERWILL AND S.P.AYLWARD 265<br />

Figure 19.6 Pathobiology <strong>of</strong> thyroid tumorigenesis.


266 ENDOCRINE TOXICOLOGY OF THE THYROID<br />

attempting to define mechanisms in thyroid carcinogenesis (Thomas and<br />

Williams, 1992).<br />

In summary, there is, therefore, good evidence that sustained TSH drive<br />

to the thyroid gland can lead to a de-regulation <strong>of</strong> thyroid function. When<br />

investigating xenobiotic or drug-induced thyroid tumour formation, the<br />

mechanisms whereby TSH drive is increased can be understood by<br />

undertaking a series <strong>of</strong> experimental studies using in vitro and in vivo<br />

techniques. Having delineated the mechanism <strong>of</strong> the thyrotoxic effect it<br />

may then be possible to determine whether a particular drug or compound<br />

elicits a similar response in different species (including humans) and to<br />

investigate the dose-response relationship for this effect.<br />

Investigative toxicological studies and examples <strong>of</strong><br />

xenobiotics causing thyroid toxicity via the H-P-T-L axis<br />

Introduction<br />

Atterwill et al., (1993) give extensive examples <strong>of</strong> both pharmaceutical and<br />

industrial compounds causing thyroid toxicity via the five main sites along<br />

the H-P-T-L axis as shown in Figure 19.7 and readers should refer to this<br />

for further and more detailed information. In this chapter, three <strong>of</strong> these<br />

five thyroid toxicity loci are described in relation to the endocrine effects<br />

produced, industrial xenobiotic examples, and investigative in vivo and in<br />

vitro tests to delineate mechanisms and species-specific effects. This<br />

information is further summarised in Figure 19.8.<br />

In terms <strong>of</strong> industrial compounds the most frequently cited examples<br />

causing thyroid toxicity appear to be in the categories <strong>of</strong>: (i) those<br />

potentially affecting the plasma protein binding <strong>of</strong> thyroid hormones—for<br />

example, the nitrile herbicide, ioxynil (Ogilvie and Ramsden, 1988); (ii)<br />

those acting directly on the thyroidal peroxidase enzyme as goitrogens, and<br />

blocking thyroid hormone synthesis and secretion—for example, the coal<br />

derived hydroxyphenol products (Lindsay et al., 1992); and (iii) those<br />

affecting the hepatic metabolism and elimination T 3 and T 4—for example,<br />

compounds such as β-naphth<strong>of</strong>lavone, PCBs and alachlor (Ogilvie and<br />

Ramsden, 1988). Tables 19.1–19.3 show examples <strong>of</strong> these three class<br />

effects, compounds producing the effects and some <strong>of</strong> the range <strong>of</strong><br />

investigative tests currently available.<br />

in vivo and in vitro studies <strong>of</strong> xenobiotics acting on the<br />

hepatic metabolism and clearance <strong>of</strong> thyroxine<br />

There is a growing list <strong>of</strong> agents, both pharmaceutical and industrial<br />

xenobiotics, which act in rodents by interfering with thyroid hormone


Figure 19.7 Toxicological loci in H-P-T-L axis.<br />

C.K.ATTERWILL AND S.P.AYLWARD 267<br />

metabolism, hepatic elimination and thus circulating TSH levels (see also<br />

Capen and Martin, 1989; McClain, 1989; Atterwill et al., 1993). The<br />

xenobiotics include phenobarbital (McClain, 1989), β-naphth<strong>of</strong>lavone<br />

(Johnson et al., 1993), the polychlorinated biphenyls (Bastomsky, 1974),<br />

diproteverine (a calcium antagonist; Flack et al., 1989), SC37211 (a Searle<br />

imidazole antimicrobial (Comer et al., 1985), L649923 (a leukotriene D 4<br />

antagonist; Saunders et al., 1988), a novel oxyacetamide-FOE 5043<br />

(Christenson et al., 1993), alachlor (Brewster et al., 1993), PCNB<br />

(pentachloronitrobenzene; Story et al., 1993), and hexachlorobenzene<br />

(Ogilvie and Ramsden, 1988).<br />

Most <strong>of</strong> these compounds have thus far been assumed to act in vivo via<br />

the induction <strong>of</strong> hepatic uridine diphosphate glucuronosyltransferase (UDP-<br />

GT) in the rat, with species-specific formation <strong>of</strong> thyroid tumours in<br />

carcinogenicity studies (see McClain, 1989). Indeed many <strong>of</strong> the<br />

compounds, including phenobarbital do lead to increased hepatic UDP-GT<br />

activity and appearance <strong>of</strong> glucuronidated T 4 in the bile, sometimes with<br />

elevated bile flow rates (see McClain, 1989). However, others such as the


268 ENDOCRINE TOXICOLOGY OF THE THYROID<br />

Figure 19.8 Investigative tests on H-P-T-L Axis.<br />

pharmaceutical temelastine increase predominantly the clearance <strong>of</strong> free<br />

T 4, though the bile product is not in conjugate form (Poole et al., 1989,<br />

1990). Other compounds such as the food dye FD&C Red No 3 (Capen<br />

and Martin, 1989) are able to lower circulating triiodothyronine (T 3) by<br />

altered deiodination suggesting the further existence <strong>of</strong> alternative<br />

mechanisms. Furthermore, not all chemicals inducing hepatic neoplasia in<br />

rodents cause thyroid neoplasia (McClain, 1989).<br />

We and others have reported that two SK&F histamine antagonists,<br />

temelastine (SK&F 93944) and lupitidine (SK&F 93479) produce ratspecific<br />

thyroid lesions via perturbation <strong>of</strong> the hepatic locus (Atterwill et


Table 19.1<br />

Table 19.2<br />

C.K.ATTERWILL AND S.P.AYLWARD 269<br />

al., 1989). Increased thyroxine clearance from the circulation, followed by<br />

elevated TSH ‘drive’ and increased thyroid follicular cell growth were<br />

observed. These com pounds act rapidly within minutes—hours <strong>of</strong> in vivo<br />

drug administration and are apparently able to increase the accumulation <strong>of</strong><br />

T 4 directly in vitro by cultured rat hepatocytes (Atterwill et al., 1989; Poole<br />

et al., 1990). Phenobarbital appears to share this property in vitro


270 ENDOCRINE TOXICOLOGY OF THE THYROID<br />

Table 19.3<br />

(Aylward et al., 1994) and increases the accumulation <strong>of</strong> thyroxine in<br />

treated rat liver in vivo (Oppenheimer et al., 1968).<br />

Thyroxine transport (Figure 19.9) is regulated by specific components<br />

located within the plasma membrane in various cell types including<br />

fibroblasts and hepatocytes and is an important prerequisite for both<br />

hormone metabo lism and nuclear hormonally-mediated events (Pliam and<br />

Goldfine, 1977; Krenning et al., 1981; Blondeau, 1986). Investigations<br />

indicate that there exist two distinct transport systems specific to<br />

thyroxine: a high-affinity, low capacity, energy-dependent ATP-ase linked<br />

transport system and a low-affinity, high capacity transport mechanism<br />

(Sorimachi and Robbins, 1978; Krenning et al., 1981, 1983; Blondeau,<br />

1986; Rao, 1991).<br />

Many <strong>of</strong> the compounds listed as indirect carcinogens in rat (due to an<br />

ability to induce liver microsomal enzymes and increase glucuronidated<br />

thyroxine elimination in the bile) have been shown to increase hepatic UDP-<br />

GT activity or cause liver hypertrophy indicative <strong>of</strong> following repeated<br />

dosing (Comer et al., 1985; McClain, 1989; Johnson et al., 1993).<br />

However, there have been no attempts to provide a definitive link between<br />

UDP-GT induction and thyroid pathology or to prove a primary<br />

endocrinological effect via UDP-GT. Our previous work with temelastine<br />

in the rat in vivo (Atterwill et al., 1989) was able to demonstrate that the<br />

increased clearance <strong>of</strong> T 4 from the rat circulation appeared within a few<br />

hours <strong>of</strong> a single compound dosing (Atterwill et al., 1989). Even


Figure 19.9 Hepatic events leading to hormone elimination.<br />

C.K.ATTERWILL AND S.P.AYLWARD 271<br />

phenobarbital is able to increase the thyroxine clearance in a relatively<br />

short timespan (Atterwill, unpublished observations). New in vitro data are<br />

not inconsistent with the time course <strong>of</strong> the in vivo phenomena where<br />

enhanced T 4 hepatocytic accumulation by cultured rat hepatocytes<br />

following compound exposure occurred as early as 60–90 min after<br />

exposure (Aylward et al., 1994). There was no membrane cytotoxic effect<br />

<strong>of</strong> the compounds at the threshold concentrations producing these effects in<br />

vitro. This shows a potential rapid direct effect <strong>of</strong> the xenobiotics on


272 ENDOCRINE TOXICOLOGY OF THE THYROID<br />

Table 19.4 Species and energy dependence <strong>of</strong> enhanced thyroxine accumulation in<br />

vitro<br />

Key: ↑T 4, increase; ↓T 4, decrease; ↔T 4, no change; Temperature? ATP?,<br />

temperature/ATP dependent; NA, not applicable.<br />

hepatocellular T 4 accumulation. The correlation between in vivo—in vitro<br />

species-specific toxicological effects is also evident (Figure 19.10 and<br />

Table 19.4). One <strong>of</strong> the features <strong>of</strong> temelastineinduced thyroid toxicology<br />

in vivo was the apparent species-specificity to the rat (Atterwill et al.,<br />

1989; Poole et al., 1989). Temelastine-mediated thyroid follicular<br />

hypertrophy and hyperplasia was not observed in dog, mouse or monkey<br />

following temelastine treatment (Figure 19.10). In vitro, no enhanced<br />

thyroxine accumulation in response to temelastine or phenobarbital was<br />

observed in guinea pig or dog hepatocytes (Aylward et al., 1994). In<br />

support <strong>of</strong> these findings, it has been demonstrated that the guinea pig is<br />

insensitive to thyroid pathological changes after phenobarbital or βnaphth<strong>of</strong>lavone<br />

administration in vivo (Johnson et al., 1993; Wyatt et al.,<br />

1993). In support <strong>of</strong>, and as an extension <strong>of</strong> these findings, we now present<br />

important new findings to demonstrate conclusively that some <strong>of</strong> the rapid<br />

‘effectors’ <strong>of</strong> thyroid toxicity via the liver, such as temelastine, do so<br />

independently <strong>of</strong> a primary action on UDP-GT, whereas other cytochrome<br />

P450 inducers such as phenobarbital may have a combined effect. This<br />

work was carried out using hepatocytes prepared from UDP-GT system<br />

deficient Gunn rats.<br />

Studies on Gunn rat hepatocytes in vitro<br />

Hepatocytes were prepared from the normal or Gunn rat (deficient in UDP-<br />

GT isozymes conjugating thyroxine) and exposed to either temelastine or<br />

phenobarbital (2 or 20 µM) for 3 h as before (Aylward et al., 1994). The<br />

results show (Figure 19.11) that whereas temelastine was able to enhance<br />

thyroxine accumulation in both types <strong>of</strong> hepatocytes, phenobarbital only<br />

produced alterations in hormone accumulation in normal cells, supporting


C.K.ATTERWILL AND S.P.AYLWARD 273<br />

Figure 19.10 Effect <strong>of</strong> temelastine on 125I-T4 clearance (from Atterwill et al.,<br />

(1989)).


274 ENDOCRINE TOXICOLOGY OF THE THYROID<br />

Figure 19.11 Effect <strong>of</strong> temelastine and phenobarbital on thyroxine accumulation in<br />

vitro by hepatocytes from control and Gunn rats.<br />

earlier in vivo findings where phenobarbital was toxicologically inactive in<br />

this strain <strong>of</strong> rat (Bastomsky, 1973).


C.K.ATTERWILL AND S.P.AYLWARD 275<br />

Conclusions<br />

The observations now lend further and strong support to the hypothesis<br />

that indirect xenobiotic-induced thyroid toxicology can arise from direct<br />

effects on hepatic membrane-located thyroxine transport proteins. It also<br />

suggests that the species-specificity <strong>of</strong> this toxic effect in vivo <strong>of</strong> some<br />

xenobiotics may be attributed to actual species differences in the sensitivity<br />

<strong>of</strong> these hepatic carriers to the compounds and not simply or primarily to<br />

changes in T 4 glucuronidation via UDP-GT induction. For the first time we<br />

have demonstrated the usefulness <strong>of</strong> Gunn rat hepatocytes in vitro for<br />

discriminating between the two ‘hepatic subclasses’ <strong>of</strong> xenobiotics causing<br />

thyroid toxicity in rodents.<br />

A number <strong>of</strong> practical in vivo and in vitro investigative tests are now<br />

available for delineating mechanisms <strong>of</strong> thyroid toxicity along the H-P-T-L<br />

axis, and which also provide screening tools for examining chemical series<br />

<strong>of</strong> potentially toxic molecules: (i) Direct block <strong>of</strong> thyroid function via<br />

peroxidase inhibition can be measured in vivo by the perchlorate discharge<br />

test (Atterwill et al., 1987); (ii) it can also be measured in vitro using<br />

cultured thyrocytes (Atterwill and Fowler, 1990); (iii) indirect effects on<br />

hepatic thyroxine clearance can be assessed in vivo (Atterwill et al., 1989);<br />

or (iv) in vitro using cultured hepatocytes from different species or Gunn<br />

rat (Aylward et al., 1994). Effects on receptors at the hypothalamic and<br />

pituitary levels can also now be studied extensively using both in vivo and<br />

in vitro approaches (Buckingham and Gillies, 1993). This battery <strong>of</strong><br />

technology now available will greatly advance the mechanistic<br />

understanding and screening <strong>of</strong> thyroid endocrine toxicants.<br />

References<br />

AKOSO, B.T., SLEIGHT, S.D., NACHREINER, R.F. and AUST, S.D., 1982,<br />

Effects <strong>of</strong> purified polybrominated biphenyl congeners on the thyroid and<br />

pituitary glands in rats, J. Am. College Toxicol., 3, 23–36.<br />

ATTERWILL, C.K. and FOWLER, K.F., 1990, A comparison <strong>of</strong> cultured rat<br />

FRTL-5 and porcine thyroid cells for predicting the thyroid toxicity <strong>of</strong><br />

xenobiotics, Toxicol. in vitro, 4, 369–74.<br />

ATTERWILL, C.K. and FLACK, J.D., 1993, Endocrine <strong>Toxicology</strong>, Cambridge:<br />

Cambridge University Press.<br />

ATTERWILL, C.K., COLLINS, P., BROWN, G.G. and HARLAND, R.F., 1987,<br />

The perchlorate discharge test for examining thyroid function in rats, J.<br />

Pharmacol. Methods, 18, 199–203.<br />

ATTERWILL, C.K., KENNEDY, S., JONES, C.A., LEE, D.M., DAVIES, S. and<br />

POOLE, A., Comparison <strong>of</strong> the toxicity <strong>of</strong> orally administered Ltriiodothyronine<br />

(T 3) in rat and cynomolgus monkey, paper presented at the<br />

British Toxicological Society Meeting, Oxford, March 1987.


276 ENDOCRINE TOXICOLOGY OF THE THYROID<br />

ATTERWILL, C.K., POOLE, A., JONES, R. and BROWN, R., 1989, Mechanistic<br />

investigation <strong>of</strong> species-specific thyroid lesions induced by treatment with the<br />

histamine H1 antagonist Temelastine (SK&F 93944) in rats, Food Chem.<br />

Toxicol., 27, 681–90.<br />

ATTERWILL, C.K., JONES, C. and BROWN, R., 1993, Thyroid Gland II—<br />

Mechanisms <strong>of</strong> species-dependent thyroid toxicity, hyperplasia and neoplasia<br />

induced by xenobiotics, in Atterwill, C.K. and Flack, J.D. (Eds) Endocrine<br />

<strong>Toxicology</strong>, pp. 137–82, Cambridge: Cambridge University Press.<br />

AYLWARD, S., WALKER, T. and ATTERWILL, C.K., 1994, Xenobiotic<br />

modulation <strong>of</strong> thyroxine uptake and efflux by cultured rat hepatocytes in vitro<br />

in relation to toxicological effects on the thyroid gland, Toxicol. in vitro (in<br />

press).<br />

BASTOMSKY, C.H., 1973, The biliary excretion <strong>of</strong> thyroxine and its glucuronic<br />

acid conjugate in normal and Gunn rats, Endocrinology, 92, 35–40.<br />

BASTOMSKY, C.H., 1974, Effects on a polychorinated biphenyl mixture<br />

(Arochlor 1254) and DDT on the biliary thyroxine excretion in rats,<br />

Endocrinology, 95, 1150–5.<br />

BAYLISS, P.H. and TUNNBRIDGE, W.M.G., 1985, Endocrine disorders, in<br />

Davies, D.M. (Ed.) Textbook <strong>of</strong> Adverse Drug Reactions, 3rd edn, pp. 335–51,<br />

Oxford: Oxford Medical Publications.<br />

BLONDEAU, J.P., 1986, Saturable binding <strong>of</strong> the thyroid hormone in isolated rat<br />

hepatocytes, FEBS Lett., 204, 41–7.<br />

BREWSTER, D.W., HOTZ, F., WARD, D.P., HEYDENS, W.E. and WILSON, A.<br />

G.E., 1993, Evidence for a hormonally-mediated non-genotoxic mechanism <strong>of</strong><br />

action <strong>of</strong> alachlor-induced rat thyroid tumours, The Toxicologist, 13(1), 1440.<br />

BUCKINGHAM, J. and GILLIES, J.G., 1991, Hypothalamus and pituitary gland<br />

— xenobiotic-induced toxicity and models for its investigation, in Atterwill,<br />

C.K. and Flack, J.M. (Eds), Endocrine <strong>Toxicology</strong>, pp. 83–115, Cambridge:<br />

Cambridge University Press.<br />

CAPEN, C.C. and MARTIN, S.L., 1989, The effects <strong>of</strong> xenobiotics on the structure<br />

and function <strong>of</strong> the thyroid follicular and C-cells, Toxicolog. Pathol, 17(2),<br />

266–92.<br />

CAVALIERI, R.R. and PITT-RIVERS, R., 1981, The effects <strong>of</strong> drugs on the<br />

distribu-tion and metabolism <strong>of</strong> thyroid hormones, Pharmacol. Rev., 33, 55–<br />

80.<br />

CHRISTENSON, W.R., BECKER, B.D., HONNG, H.D. and WAHLE, B.S., 1993,<br />

Investigations into the mechanisms <strong>of</strong> decline in serum thyroxine following the<br />

administration <strong>of</strong> a novel oxyacetamide, (FOE 5043), The Toxicologist, 13(1),<br />

1438.<br />

CI OMS, 1983, Safety requirements for the first use <strong>of</strong> new drugs and diagnostic<br />

agents in man. A review <strong>of</strong> safety issues in early clinical trials <strong>of</strong> drugs.<br />

Geneva: Council for International Organisations <strong>of</strong> Medical Sciences.<br />

COLLETTA, G., CIRAFICI, A.M. and VECCHIO, G., 1986, Induction <strong>of</strong> the c-fos<br />

oncogene by thyrotropic hormone in rat thyroid cells in culture, Science, 233,<br />

458– 60.<br />

COMER, C.P., CHENGELIS, C.P., LEVIN, S. and KOTSONIS, F.M., 1985,<br />

Changes in thyroidal function and liver UDP-glucuronosyltransferase activity


C.K.ATTERWILL AND S.P.AYLWARD 277<br />

in rats following administration <strong>of</strong> a novel imidazole, (SC-37211), Toxicol.<br />

Appl. Pharmacol., 80, 427–318.<br />

DOCTER, R., KRENNING, E.P., BERNARD, H.F. and HENNEMANN, G., 1987,<br />

Active transport <strong>of</strong> iodothyronines into human cultured fibroblasts, J. Clin.<br />

Endocrinol. and Metabolism, 64(4), 624–8.<br />

DOHLER, K.D., WONG, C.C. and MUHLEN, A.V., 1973, The rat as a model for<br />

the study <strong>of</strong> drug effects on thyroid function: consideration <strong>of</strong> methodological<br />

problems, Pharmac. Ther., 5, 205–318.<br />

ENGLER, D. and BURGER, A.C., 1984, The deiodination <strong>of</strong> iodothyrones and their<br />

derivatives in man, Endocrin. Rev., 5, 151–2.<br />

FLACK, J.D., HAKANSSON, S., JEFFREY, D.J., KELVIN, A.S., MAILE, P.A.,<br />

MCCURRDO, A.S. and PERKINS, C.I., 1989, Investigation <strong>of</strong> the effects <strong>of</strong><br />

diproterivene on the thyroid <strong>of</strong> the rat, Hum. Toxicol., 8, 411.<br />

HEYWOOD, R., 1984, Prediction <strong>of</strong> adverse drug reactions from animal safety<br />

studies, in Bostrun, H. and Ljungstedt, N. (Eds) Detection and Prevention <strong>of</strong><br />

Adverse Drug Reactions, pp. 177–89, Sweden: Almaqvuist & Wiskell Int.<br />

HIASA, Y., KITAHORI, Y., OSHIMA, M., FUJITA, T., YUASA, T., KONISHI, N.<br />

and MIYASHIRO, A., 1982, Promoting effects <strong>of</strong> phenobarbital and barbital<br />

on development <strong>of</strong> thyroid tumours in rats treated with N-bis (2hydroxypropyl)<br />

nitrosamine, Carcinogenesis, 3, 1187–90.<br />

JOHNSON, S., McKILLOP, D., MILLER, J. and SMITH, I.K., 1993, The effect on<br />

rat thyroid function <strong>of</strong> an hepatic microsomal enzyme inducer, Hum., Exp.<br />

Toxicol., 12, 153–8.<br />

JONES, C.A., BROWN, G.C., DICKENS, T.A. and ATTERWILL, C.K., 1988,<br />

Differential effects <strong>of</strong> D-a and l-isomers <strong>of</strong> triiodothyrmine on pituitary TSH<br />

secretion and peripheral deiodinase activity in the rat, <strong>Toxicology</strong>, 48, 273–84.<br />

KOHRLE, J., BRABANT, G. and HESCH, R.D., 1987, Metabolism <strong>of</strong> thyroid<br />

hormones, Hormone Res., 26, 58–78.<br />

KRENNING, E., DOCTER, R., BERNARD, B., VISSA, T. and HENNEMANN,<br />

G., 1981, Characteristics <strong>of</strong> active transport <strong>of</strong> thyroid hormones into rat<br />

hepatocytes, Biochim. Biophys. Acta., 676, 314–20.<br />

KRENNING, E., DOCTER, R., VISSER, T.J. and HENNEMANN, G., 1983,<br />

Plasma membrane transport <strong>of</strong> thyroid hormones: its possible<br />

pathophysiological significance, J. Endocrinol, 6, 59–65.<br />

LINDSAY, R.H., HILL, J.B., GAITAN, E., COOKSEY, R.C. and JOLLEY, R.L.,<br />

1992, Anti-thyroid effects <strong>of</strong> coal-derived pollutants, J Toxicol. Environm.<br />

Hlth., 37, 467–81.<br />

McCLAIN, R.M., 1989, The significance <strong>of</strong> microsomal enzyme induction and<br />

altered thyroid function in rats: implications for thyroid gland neoplasia,<br />

Toxicol. Pathol., 17(2), 294–306.<br />

MOSSMAN, T., 1983, Rapid colorimetric assay for cellular growth and survival:<br />

application to proliferation and cytotoxicity assay, J. Immunolog. Methods, 65,<br />

55–63.<br />

OGILVIE, L.M. and RAMSDEN, D.B., 1988, Ioxynil and 3,5,3'-triiodothyronine:<br />

comparison <strong>of</strong> binding to human plasma proteins, Toxicol. Lett., 44, 281–7.<br />

OPPENHEIMER, J.H., BERNSTEIN, G. and SURKS, M.L., 1968, Increase<br />

thyroxine turnover and thyroidal function after stimulation <strong>of</strong> hepatocellular<br />

binding <strong>of</strong> thyroxine by phenobarbital, J. Clin. Invest., 47, 1399–406.


278 ENDOCRINE TOXICOLOGY OF THE THYROID<br />

OSHIMA, M. and WARD, J.M., 1984, Promotion <strong>of</strong> N-methyl-N-nitrosoureainduced<br />

thyroid tumours by iodine deficiency in F334/NCr rats, J. Nat. Cancer<br />

Inst., 73, 289–96.<br />

PARMA, J. et al., 1993, Somatic mutations in the thyrotropin receptor gene cause<br />

hyperfunctioning thyroid adenomas, Nature, 365, 649–51.<br />

PETER, H.J., STUDER, H., FORSTER, R. and GERBER, H., 1982, The<br />

pathogenesis <strong>of</strong> ‘hot’ and ‘cold’ follicles in multinodular goitres, J. Clin.<br />

Endocrinol. Metal., 55, 941–6.<br />

PLIAM, N.B. and GOLDFINE, I.D., 1977, High affinity thyroid hormone binding<br />

sites on purified rat liver plasma membranes, Biochem. Biophys. Res. Comm.,<br />

19, 166–72.<br />

POOLE, A., BETTON, G.R., SALMON, G., SUTTON, T. and ATTERWILL,<br />

C.K., 1989, Comparative toxicology <strong>of</strong> temelastine; a novel non-sedating H 1<br />

antagonist in the dog, rat and the monkey, Fundam. Appl. Toxicol, 14, 71–83.<br />

14.<br />

POOLE, A., PRITCHARD, D., JONES, R.B., CATTO, L. and LEONARD, T.,<br />

1990, In vivo biliary excretion and in vitro cellular accumulation <strong>of</strong> thyroxine<br />

in rats or cultured rat hepatocytes treated with a novel histamine H 1 receptor<br />

antagonist, Arch. Toxicol, 64, 474–81.<br />

RAO, C.G., 1991, Mode <strong>of</strong> entry <strong>of</strong> steroid and thyroid hormones into cells, Mol.<br />

Cell Endocrinol., 21, 97–108.<br />

RIBELIN, W.E., 1984, The effects <strong>of</strong> drugs and chemicals upon the structure <strong>of</strong> the<br />

adrenal gland, Fundam. Appl. Toxicol, 4, 105–19.<br />

SAUNDERS, J.E., EIGENBURG, D.A., BRACHT, L.E., WANG, W.R. and VAN<br />

ZWEITEN, M.J., 1988, Thyroid and liver trophic changes in rat secondary to<br />

microsomal enzyme induction caused by an experimental leukotrien<br />

antagonist, (L-649,923), Toxicol. Appl. Pharmacol., 5, 378–87.<br />

SMEDS, S., PETERS, H.J., JORTSO, E., GERBER, H. and STUDER, H., 1987,<br />

Naturally occurring clones <strong>of</strong> cells with high intrinsic proliferation potential<br />

within the follicular epithelium <strong>of</strong> mouse thyroids, Cancer Res., 47, 1646–51.<br />

SMITH, P., WYNFORD-THOMAS, D., STRINGER, B.M.J. and WILLIAMS,<br />

E.D., 1986, Growth factor control <strong>of</strong> rat thyroid follicular cell proliferation,<br />

Endocrinology, 119, 1439–45.<br />

SORIMACHI, K. and ROBBINS, O., 1978, Uptake and metabolism <strong>of</strong> thyroid<br />

hormones by cultured monkey hepatocarcinoma cells. Effects <strong>of</strong> potassium<br />

cyanide and dinitrophenol, Biochim. Biophys. Acta, 542, 515–26.<br />

STORY, D.I., CARDONA, R.A. and LENGEN, M.R., 1993, Effect <strong>of</strong> dietary PCNB<br />

on circulating levels <strong>of</strong> T 3, T 4 and TSH in rats, The Toxicologist, 13(1), 1446.<br />

STRINGER, B.M.J., WYNFORD-THOMAS, D. and WILLIAMS, E.D., 1985, In<br />

vitro evidence for an intracellular mechanism limiting the thyroid follicular cell<br />

growth response to thyrotropin, Endocrinology, 116, 611–15.<br />

THOMAS, G.A. and WILLIAMS, E.D., 1991, Evidence for and possiblemechanisms<br />

<strong>of</strong> non-genotoxic carcinogenesis in the rodent thyroid, Mutat. Res., 248, 357–<br />

70.<br />

THOMAS, G.A. and WILLIAMS, E.D., 1992, Production <strong>of</strong> thyroid tumours in<br />

mice by demethylating agents, Carcinogenesis, 13, 1039–42.


C.K.ATTERWILL AND S.P.AYLWARD 279<br />

WILLIAMS, D.W., WYNFORD-THOMAS, D. and WILLIAMS, E.D., 1987,<br />

Human thyroid adenomas show escape from IGF-1 dependence for growth,<br />

Ann. Endocrinol., 48(2), 11.<br />

WYATT, I., GYTE, A., FOSTER, J.R., WILLIAMS, S.M. and ELCOMBE, C.R.,<br />

1993, Differences in the response <strong>of</strong> the rat and guinea pig liver and thyroid to<br />

hepatic enzyme inducers, Hum. Exp. Toxicol., 12(6), 561.<br />

WYNFORD-THOMAS, D., STRINGER, B.M. J. and WILLIAMS, E.D., 1982,<br />

Dissociation <strong>of</strong> growth and function in the rat thyroid during prolonged<br />

goitrogen administration, Acta Endocrinol., 101, 210–16.<br />

ZBINDEN, G. 1987, Assessment <strong>of</strong> hyperplastic and neoplastic lesions <strong>of</strong> the thyroid<br />

gland, TIPS (Dec., 1987), 8, 511–14.


20<br />

Testing and Evaluation for Reproductive Toxicity<br />

ANTHONY K.PALMER<br />

Huntingdon Research Centre, Huntingdon<br />

Introduction<br />

Most <strong>of</strong> the presentations at this meeting refer to high level, scientific<br />

investigations <strong>of</strong> one or two, highly important, high production volume<br />

chemicals, for which an adverse effect has been demonstrated. They are<br />

studies <strong>of</strong> characterisation, because they elaborate on known effects using a<br />

wealth <strong>of</strong> available information. But, how were the adverse effects <strong>of</strong> these<br />

few substances first discovered, what were the initial clues? Sadly, for<br />

many, the observation <strong>of</strong> adverse effects in humans was the trigger to<br />

intensive investigations, which is akin to ‘shutting the stable door after the<br />

horse has bolted’.<br />

This presentation is concerned with detecting effects <strong>of</strong> substances for<br />

which little or no information is available and, preferably, before they<br />

cause harm to humans. This requires a different kind <strong>of</strong> science, for which<br />

the main asset is the ability to predict, with reasonable accuracy, possible<br />

activity from minimal information. It requires wide experience and a<br />

balance between imagination and pragmatism. These attributes are<br />

especially important for toxicity to reproduction, which triggers instinctive<br />

reactions in even the coolest and most objective scientist.<br />

Identifying the cause <strong>of</strong> adverse effects on human reproduction has long<br />

been surrounded by controversy and uncertainty. In respect <strong>of</strong> the<br />

evaluation <strong>of</strong> substances for reproductive toxicity this state <strong>of</strong> affairs seems<br />

likely to persist for years to come. The main obstacle to any attempt to<br />

rationalise the situation is that any discussion on evaluation almost<br />

inevitably gravitates to the black hole <strong>of</strong> regulatory guidelines. All<br />

guidelines are flawed because science fact is compromised by bureaucracy<br />

and science fiction. For many reasons, but especially the unwillingness <strong>of</strong><br />

any establishment to change the status quo, guidelines provide the worst<br />

starting point for developing a strategy for evaluation.<br />

Most guidelines are concerned only with methods for gathering specified<br />

information. On its own this information (on hazard) is insufficient and<br />

needs to be supplemented with other information, from other sources, to


Table 20.1 Production volume triggers for industrial chemicals<br />

Table 20.2 EC Annex VII and VIII toxicity tests for industrial chemicals<br />

A.K.PALMER 281<br />

Notes:<br />

Tests for ecotoxicity are not included.<br />

Progression, by rote, from base set to level 2 would involve duplication <strong>of</strong> effort in<br />

several areas.<br />

The state <strong>of</strong> confusion regarding testing for toxicity to reproduction is portrayed by<br />

the failure to decide on requirements at base set level and the curious mixture <strong>of</strong> old<br />

and new terminology regarding tests.<br />

predict whether humans might be affected. Most guidelines are a watershed<br />

in a broader spectrum <strong>of</strong> testing and assessment. They represent a point at<br />

which it may be decided that the only way to gather more information is to<br />

take the final step <strong>of</strong> exposing humans. Exposure <strong>of</strong> humans provides the


282 TESTING AND EVALUATION FOR REPRODUCTIVE TOXICITY<br />

Figure 20.1 Overlap <strong>of</strong> toxicity.<br />

only certain way to determine whether reproduction would be affected, but<br />

we need to do the best we can before taking the chance.<br />

The paradox in this is that exposure <strong>of</strong> humans is facilitated by failure to<br />

demonstrate toxicity in animals. But, lack <strong>of</strong> activity, being negative, cannot<br />

be proven, only presumed. To make this presumption investigations must<br />

be extensive and comprehensive to convey reasonable assurance that failure<br />

was not due to deficiencies in methodology.<br />

There is an exception. For industrial chemicals, testing <strong>of</strong> all substances<br />

to these criteria would be a monumental task, therefore, less stringent<br />

testing is allowed according to production volume, which serves as an<br />

approximation for the extent <strong>of</strong> the population likely to be exposed<br />

(Table 20.1). It is a strategy based on population risk, the downside <strong>of</strong><br />

which is an increased risk to the individual. The strategy takes advantage<br />

<strong>of</strong> the fact that, in a small population, the chance <strong>of</strong> identifying cause and<br />

effect is poor. At the base set level and level 1, as outlined by the EC, all<br />

tests are equivalent only to the voluntary preliminary studies conducted for


medicines, agrochemicals and food additives. They may be sufficient to<br />

detect potent toxicity but not comprehensive enough to allow presumption<br />

<strong>of</strong> the absence <strong>of</strong> hazard, especially as the base set does not include tests<br />

for reproductive toxicity (Table 20.2).<br />

As production volume increases, more extensive testing should be<br />

undertaken. Often, this has been neglected, prompting the development <strong>of</strong><br />

the OECD guidelines 421 and 422. These tests were intended to recover a<br />

situation that never should have arisen.<br />

A better approach?<br />

There is no question that evaluation for reproductive toxicity could be<br />

improved considerably. The question is whether industry and agencies are<br />

willing to do so. It would require a change <strong>of</strong> attitude in industry and<br />

agencies alike. Industry’s ‘passive avoidance’ <strong>of</strong> testing would need to be<br />

replaced by ‘active participation’. For a new substance the first step should<br />

be an integrated assessment <strong>of</strong> commercial prospects and potential toxicity<br />

over a broad spectrum (Figure 20.1). Early identification <strong>of</strong> ‘serious bad<br />

actors’, which tend to effect many systems, can save time and effort. Given<br />

the prognosis <strong>of</strong> problems ahead, it may be better to devote resources to<br />

finding safer alternatives, or to risk management, rather than to endless<br />

testing. For materials with a high commercial potential, the aim should be<br />

to get to full scale tests by the quickest route. Following the EC levels by rote<br />

is very inefficient since there is duplication with successive steps.<br />

Methods<br />

With an active participation policy, a much broader scope <strong>of</strong> methodology<br />

can and should be considered, ranging from searches for structure-activity<br />

relationships, through various in vitro methods, whole animal tests, wild<br />

life surveys and human surveys (Table 20.3). Due to time constraints I will<br />

concentrate on whole animal test systems.<br />

Structure-activity databases<br />

Structure and activity relationships are an obvious place to start any<br />

evaluation, despite the fact that currently available databases are far from<br />

perfect. Their reliability could be improved dramatically by adding unused<br />

information currently hidden in industry and agency archives.<br />

Entire mammalian tests<br />

A.K.PALMER 283<br />

Tests in entire mammals provide the only way <strong>of</strong> assessing what effects a<br />

substance may evoke in the complex, integrated and dynamic process <strong>of</strong>


284 TESTING AND EVALUATION FOR REPRODUCTIVE TOXICITY<br />

Table 20.3 Methods for detecting effects on human reproduction<br />

reproduction. To detect the wide range <strong>of</strong> possible effects it is necessary to<br />

expose mammals to a substance from conception through sexual maturity.<br />

It is necessary to look for consequences <strong>of</strong> this exposure through at least<br />

one life cycle (Figure 20.2). This long observation period is required for<br />

detection <strong>of</strong> latent manifestations <strong>of</strong> developmental toxicity, such as those<br />

induced by lead, alcohol, diethylstilboestrol and other hormonally active<br />

substances. The only means <strong>of</strong> covering all these aspects is a two generation<br />

study or the equivalent in a combination <strong>of</strong> tests.<br />

Restricted test systems<br />

The use <strong>of</strong> lesser tests for industrial chemicals is a concession. Examination<br />

for some effects is omitted because they are not perceived to be important<br />

or because they would be difficult to detect, or because they occur very<br />

rarely. For example, first detection <strong>of</strong> effects in <strong>of</strong>fspring <strong>of</strong> second<br />

generations is rare so such activity does not have a high priority.<br />

With these restricted tests, emphasis should be on detecting effects and<br />

not on manipulating a no effect level. Tests that could be considered would<br />

include OECD 421 and 422, the OECD single generation study and the old<br />

FDA Segment I study for medicines. The latter two are restricted tests<br />

because they do not allow detection <strong>of</strong> latent manifestations <strong>of</strong><br />

developmental toxicity.<br />

The best return for effort is afforded by OECD 422, which combines<br />

examination for general or systemic toxicity, as well as reproductive<br />

toxicity. However, realising its potential requires an experienced laboratory<br />

team, the courage to modify the test and the conceptual ability to know<br />

how to interpret the results.


Figure 20.2 Cycle <strong>of</strong> life/reproduction.<br />

A.K.PALMER 285<br />

OECD 421 involves treatment <strong>of</strong> both sexes from about 2 weeks prior to<br />

mating through to termination, a few days after birth <strong>of</strong> <strong>of</strong>fspring<br />

(Figure 20.3). Assessment <strong>of</strong> male fertility is achieved in two parts. Males<br />

are paired with females for detection <strong>of</strong> effects unrelated to<br />

spermatogenesis, for example, effects on sexual behaviour, libido or<br />

ejaculation and functional maturation <strong>of</strong> sperm.<br />

For detecting effects on spermatogenesis, direct methods, particularly<br />

histopathological examinations <strong>of</strong> testes and epididymides are used. Sperm<br />

analysis (seminology) could be added, although it does not seem to be<br />

better than histopathology. These methods could be incorporated into<br />

systemic toxicity studies rather than in the reproduction study per se.<br />

In respect <strong>of</strong> fecundity, treatment and observations <strong>of</strong> females include<br />

most <strong>of</strong> those applied in full scale tests. An exception is the lack <strong>of</strong><br />

observations for delayed, post-natal manifestations. Detailed examination<br />

<strong>of</strong> foetuses for skeletal and s<strong>of</strong>t tissue abnormalities is not included. The<br />

potential for prenatal effects is deduced by observation <strong>of</strong> post-natal<br />

differences in numbers pregnant, litter size, litter and mean pup weight at<br />

birth and to day 4 post partum.<br />

As with any guideline, OECD 421 should be used with commonsense<br />

and flexibility. If pretesting prognosis suggests that prenatal effects are<br />

unlikely, extension <strong>of</strong> the study to weaning <strong>of</strong> the <strong>of</strong>fspring (Figure 20.3)<br />

provides added safeguards at little extra cost. Increase group size and it


286 TESTING AND EVALUATION FOR REPRODUCTIVE TOXICITY<br />

Figure 20.3 OECD 421 priority selection test.<br />

Figure 20.4 Fertility and embryotoxicity.<br />

provides the equivalent <strong>of</strong> the OECD single generation study. Conversely,<br />

if pretesting prognosis suggests a high probability <strong>of</strong> prenatal effects,<br />

including induction <strong>of</strong> malformation, then females could be killed just<br />

before delivery and foetuses examined for structural defects (Figure 20.4).<br />

This provides the equivalent <strong>of</strong> a fertility and embryotoxicity study we will<br />

see again later.<br />

OECD guideline 422 simply adds to OECD 421, elements for assessment<br />

<strong>of</strong> systemic and neurotoxicity. For those who have never conducted such a<br />

test it seems impossibly complex, but it is neither as difficult to perform,<br />

nor to interpret, as is feared. Its rejection by EC Officialdom makes it an<br />

even better proposition, since there need be no inhibitions about modifying<br />

the design according to circumstances.<br />

Some brief examples <strong>of</strong> results that may be encountered with positive<br />

materials are illustrated by the examples <strong>of</strong> Carbendazim (metabolite <strong>of</strong><br />

Benomyl), DEHP, Cyclophosphamide and ethylene glycol methyl ether<br />

(EGME, 2-methoxyethanol). With Carbendazim (Table 20.4) macroscopic<br />

and microscopic examinations show unequivocal effects on testes and<br />

epididymides indicating an effect on spermatogenesis. An effect on females<br />

and <strong>of</strong>fspring is indicated by an increased duration <strong>of</strong> pregnancy, reduction<br />

in the number <strong>of</strong> females with live young and lower values for litter size,


Table 20.4 OECD 422: carbendazim, tabular summary<br />

Notes:<br />

a Malformations included hydrocephaly and misaligned tails.<br />

pp=post partum<br />

Bold type indicates treatment effects including macroscopic and microscopic<br />

changes in testes and epididymides (an effect on spermatogenesis), an increased<br />

duration <strong>of</strong> pregnancy, reduction in the number <strong>of</strong> females with live young and lower<br />

values for litter size, litter weight and mean pup weight. The dosage related pattern<br />

<strong>of</strong> response provides added emphasis, as does the observation <strong>of</strong> malformed<br />

foetuses.<br />

A.K.PALMER 287<br />

litter weight and mean pup weight. The dosage related pattern <strong>of</strong> response<br />

provides added emphasis, as does the observation <strong>of</strong> malformed foetuses.<br />

With DEHP (Table 20.5) an effect on spermatogenesis is evident at 2000<br />

mg kg −1 . Treatment at this dosage had to be withdrawn shortly after<br />

mating to avoid further mortalities <strong>of</strong> the more susceptible females. There<br />

is a marked reduction in the number <strong>of</strong> pregnancies. At lower dosages an<br />

increased duration <strong>of</strong> pregnancy, reduced litter size and litter weight,<br />

indicates an effect on the female and/or <strong>of</strong>fspring. The higher mean pup<br />

weights are consequent to the longer duration <strong>of</strong> pregnancy.<br />

With cyclophosphamide (Table 20.6) female deaths at 3 and 4.5 mg kg −1<br />

are attributable to treatment as, at 6.7 mg kg −1 , all females died. There was<br />

a reduction in the number <strong>of</strong> females with young, litter size, litter weight<br />

and mean pup weight, providing clear evidence <strong>of</strong> effects on the female and


288 TESTING AND EVALUATION FOR REPRODUCTIVE TOXICITY<br />

Table 20.5 OECD 422: DEHP, tabular summary<br />

Notes:<br />

[] Treatment at 2000 mg kg −1 was withdrawn after mating (4 weeks <strong>of</strong> treatment)<br />

due to loss <strong>of</strong> condition and mortality <strong>of</strong> females.<br />

pp=post partum.<br />

Bold type indicates treatment effects on spermatogenesis at 2000 mg kg −1 . There is<br />

a marked reduction in the number <strong>of</strong> pregnancies. At lower dosages an increased<br />

duration <strong>of</strong> pregnancy, reduced litter size and litter weight, indicates an effect on<br />

the female and or <strong>of</strong>fspring. The higher mean pup weights are consequent to the<br />

longer duration <strong>of</strong> pregnancy.<br />

<strong>of</strong>fspring. No effects on spermatogenesis were reported but, if a dosage<br />

inducing effects on the male had been selected, all the females would have<br />

died.<br />

With EGME (Table 20.7), dosages were based on acute toxicity and<br />

limited repeat dose toxicity studies only. This provided a more realistic<br />

representation <strong>of</strong> the testing <strong>of</strong> a new substance. The first consequence was<br />

the occurrence <strong>of</strong> systemic toxicity at 500 and 1000 mg kg −1 . Treatment at<br />

the high dosages had to be withdrawn prior to mating, investigating<br />

recovery became a new objective. At 100 mg kg −1 pr<strong>of</strong>ound effects on<br />

spermatogenesis were evident. Some pregnancies were obtained, but no live<br />

young were born. For the high dosage groups, effects on males remained<br />

evident several weeks after withdrawal <strong>of</strong> treatment. The duration <strong>of</strong><br />

pregnancy was increased. A consequence <strong>of</strong> this is the higher mean pup<br />

weight. Values for numbers <strong>of</strong> implantations, live young and litter weight<br />

were lower than control values. So, not only did the test show the<br />

anticipated effects on reproduction, it also demonstrated that recovery <strong>of</strong>


Table 20.6 OECD 422: cyclophosphamide, tabular summary<br />

Notes:<br />

pp=post partum.<br />

Bold type indicates treatment effect. Female deaths at 3 and 4.5 mg kg −1 are<br />

attributable to treatment as, at 6.7 mg kg −1 , all females died. There was a reduction<br />

in the number <strong>of</strong> females with young, litter size, litter weight and mean pup weight.<br />

No effects on spermatogenesis were reported but, if a dosage inducing effect on the<br />

male had been selected, all the females would have died.<br />

females was slow. As far as I know this has not been mentioned in the<br />

extensive literature on EGME.<br />

These examples and others show that the OECD tests are capable <strong>of</strong><br />

detecting substances with marked effects on reproduction. With such<br />

results it would be foolhardy to consider higher level guideline tests. If the<br />

substance is not abandoned any further testing would require case by case<br />

designs to characterise the detected effects more completely.<br />

Full scale testing<br />

If pretesting prognosis suggests that a substance is unlikely to present<br />

problems <strong>of</strong> toxicity, fast track progression to level 2 testing should be<br />

considered (Table 20.2) to avoid unnecessary duplication <strong>of</strong> step by step<br />

testing. Expected tests include a two generation study in rats and an<br />

embryotoxicity study in rats and rabbits. In the harmonised guideline for<br />

medicines this same strategy is just one <strong>of</strong> several options and the same or<br />

greater flexibility should be made available for testing industrial<br />

chemicals.<br />

Tests for embryotoxicity<br />

A.K.PALMER 289<br />

An anomaly in this strategy is the specification for embryotoxicity studies<br />

in two species, when only one species is required for all other aspects <strong>of</strong>


290 TESTING AND EVALUATION FOR REPRODUCTIVE TOXICITY<br />

Table 20.7 OECD 422: EGME, tabular summary<br />

Notes:<br />

[] Treatment at 500 and 1000 mg kg −1 withdrawn prior to mating due to loss <strong>of</strong><br />

condition and mortality. Animals in withdrawal phase.<br />

NE not examined<br />

pp=post partum<br />

Bold type indicates treatment effect. At 100 mg kg −1 pr<strong>of</strong>ound effects on<br />

spermatogenesis were evident. Some pregnancies were obtained, but no live young<br />

were born indicating effects on females and the conceptus.<br />

For the high dosage groups, effects on testes and epididymides remained evident<br />

several weeks after withdrawal <strong>of</strong> treatment. The duration <strong>of</strong> pregnancy was<br />

increased with a consequent increase in mean pup weight. Values for numbers <strong>of</strong><br />

implantations, live young and litter weight were reduced indicating slow recovery.<br />

This does not appear to have been mentioned in the extensive literature on EGME.<br />

reproductive toxicity. A more sensible strategy would be to identify a<br />

relevant species before testing. It is pointless to conduct a test in an<br />

unsuitable species and doubly pointless to conduct tests in two irrelevant<br />

species.<br />

The requirement for detailed examination <strong>of</strong> foetuses for abnormalities<br />

is based on an exaggerated perception <strong>of</strong> risk prompted by fear. For many<br />

reasons the risk <strong>of</strong> inducing abnormalities is extremely low. Those same<br />

reasons make detection by direct observation <strong>of</strong> malformations unreliable.


Table 20.8 Post-natal detection <strong>of</strong> prenatal effects<br />

A.K.PALMER 291<br />

Notes:<br />

Prenatal effects include any s<strong>of</strong>t tissue or skeletal changes (variants, anomalies or<br />

malformations) or altered foetal weight. Post-natal effects include reduced litter size<br />

at birth, reduced mean foetal weight or increased post natal mortality.<br />

In the few studies in which post-natal effects were not detected, prenatal changes<br />

were <strong>of</strong> a ‘minor’ nature (e.g. reduced ossification) and sometimes not conclusive.<br />

The lower number <strong>of</strong> litters reared in the Japanese Experiment 2 design would<br />

contribute to the slightly higher rate <strong>of</strong> ‘failures’.<br />

Where more serious prenatal effects such as the observation <strong>of</strong> malformations or<br />

prenatal death were observed a post-natal effect was always observed.<br />

The dimensions <strong>of</strong> the tests we conduct are too small and dosage regimes<br />

are contradictory to the basic principle that malformations are induced by<br />

application <strong>of</strong> a precise dosage at a precise time. Whilst direct observation<br />

<strong>of</strong> malformations is unreliable the saving grace is that induced<br />

malformations always occur within a wider spectrum <strong>of</strong> embryotoxicity.<br />

This wider spectrum provides a more reliable, if indirect, means <strong>of</strong><br />

detecting substances that might cause malformations. Also, these effects are<br />

important in their own right.<br />

Major effects such as altered <strong>of</strong>fspring weight and prenatal death can be<br />

observed postnatally. For example both EC Segment I and Japanese<br />

Experiment 2 studies include examinations for foetal abnormalities as well<br />

as postnatal observations. A survey <strong>of</strong> such studies shows that, when<br />

malformations were observed, a post-natal effect was always observed<br />

(Table 20.8). Surveys such as this should have been conducted or<br />

sponsored by agencies and industry before formulating guidelines. Why<br />

have they not done so?<br />

The obsession with abnormalities is based on the fear <strong>of</strong> another<br />

thalidomide. The great contradiction is that a considerable number <strong>of</strong> rat<br />

embryotoxicity studies with thalidomide failed to provided convincing<br />

demonstration <strong>of</strong> teratogenicity. Conversely, reproduction studies in rats<br />

provided unequivocal, post-natal evidence <strong>of</strong> effect in the form <strong>of</strong> a marked<br />

reduction in the number <strong>of</strong> females with young and a marked reduction in


292 TESTING AND EVALUATION FOR REPRODUCTIVE TOXICITY<br />

Table 20.9 Thalidomide—rat reproduction studies<br />

Notes:<br />

Thalidomide was administered in the diet to provide a daily intake <strong>of</strong> 200 mg kg −1<br />

bodyweight from 60 days prior to mating (US FDA two litter test). Later studies<br />

demonstrated that the reduced percentage <strong>of</strong> females with young and the lower live<br />

litter size in females with young was associated with embryolethality.<br />

litter size <strong>of</strong> the few that had young. Two studies, each containing several<br />

matings showed the reproducibility <strong>of</strong> the results (Table 20.9).<br />

Two generation studies<br />

The emphasis on structural abnormalities detracts from examination for<br />

other, important and more likely manifestations <strong>of</strong> reproductive toxicity<br />

(Figure 20.5). For example, current and proposed test guidelines for nonmedicines<br />

lack procedures for detection <strong>of</strong> developmental neurotoxicity (or<br />

behaviour). This is a curious contradiction given the current fashion for<br />

investigation <strong>of</strong> adult neurotoxicity.<br />

For detecting other effects on reproduction, all regulatory versions <strong>of</strong> the<br />

two generation study leave something to be desired. Even more<br />

disappointing is that newer versions proposed by the US FDA, the US EPA,<br />

the EC and OECD have recycled many <strong>of</strong> the old flaws. Truly, there has<br />

been a great deal <strong>of</strong> activity but very little progress.<br />

All current and proposed guidelines continue to require a prolonged<br />

premating treatment period for the F0 or parent generation. The claim that<br />

it is necessary to treat males for a full spermatogenic cycle is pure science<br />

fiction. Spermatogenesis is not a cycle but a sequence <strong>of</strong> overlapping batch<br />

processes. At any one time, all stages <strong>of</strong> spermatogenesis are present and a<br />

short dosing period is sufficient to cause effects.<br />

To detect these effects, direct histopathological methods can be applied<br />

shortly after treatment. Direct methods are quicker and more certain than<br />

mating to females. Mating trials are inefficient and lack sensitivity due to


Figure 20.5 Manifestations <strong>of</strong> developmental toxicity<br />

A.K.PALMER 293<br />

the high sperm production capacity <strong>of</strong> animals compared with humans.<br />

Interim results from an ongoing survey show that, where data are<br />

available, direct methods are effective and that a combination <strong>of</strong> direct<br />

methods with a premating dosing time <strong>of</strong> 2 weeks or less is even more<br />

effective (Table 20.10). The combination compares favourably with<br />

prolonged premating treatment and mating. Why have agencies and<br />

industry failed to conduct or sponsor such surveys?<br />

For non-medicines, use <strong>of</strong> a prolonged premating treatment period for the<br />

parent generation is an unnecessary duplication as treatment is continued<br />

into the F1 generation; this cannot be mated until animals have reached<br />

sexual maturity. Using science facts, a more efficient design for a two<br />

generation study (Figure 20.6) would include the following features:<br />

– One control and 2–4 test groups with dosages set at 2–5 fold descending<br />

intervals from a high dosage.<br />

– The high dosage should be a limit dose (1000 mg kg −1 ) or one inducing<br />

a minimal systemic effect on adults.<br />

– A short 2–4 week premating treatment period for both sexes <strong>of</strong> the<br />

parent (F0) generation.<br />

– A greater group size for the F0 generation to allow balanced selection <strong>of</strong><br />

the F1 generation.


294 TESTING AND EVALUATION FOR REPRODUCTIVE TOXICITY<br />

Table 20.10 Detecting effects on males<br />

Notes:<br />

a Unknown=data not available or not examined<br />

A, organ weights, histopathology, serum chemistry.<br />

B, sperm analysis, count, motility, morphology.<br />

Data derived from an ongoing survey <strong>of</strong> 150 substances for which an effect on<br />

males has been claimed, mostly from human studies. To date 80 <strong>of</strong> the substances<br />

have been evaluated. Results indicate that use <strong>of</strong> a prolonged premating treatment<br />

period (>2 wks) has not been helpful for indicating effects on humans and is no<br />

better than use <strong>of</strong> a short premating period alone (


A.K.PALMER 295<br />

For example, a simple division <strong>of</strong> a two generation study will provide a<br />

developmental toxicity study in which pregnant females are treated from<br />

implantation through lactation or beyond and an F1 generation reared<br />

through to sexual maturity (Figure 20.8). The counterpart to this is a study<br />

<strong>of</strong> fertility in which both sexes are treated from about 2 weeks prior to<br />

Figure 20.6 A two generation study.


296 TESTING AND EVALUATION FOR REPRODUCTIVE TOXICITY<br />

Figure 20.7 Reproductive toxicity—selecting studies.<br />

Figure 20.8 Pre- and post-natal (developmental toxicity) study.<br />

mating through to termination <strong>of</strong> males after a minimum <strong>of</strong> 4 weeks <strong>of</strong><br />

treatment overall (Figure 20.9). Treatment <strong>of</strong> females continues to<br />

implantation and they may be killed and examined at about day 13–15 <strong>of</strong><br />

pregnancy.<br />

Alternatively, treatment <strong>of</strong> females can be continued beyond closure <strong>of</strong><br />

the palate or even through pregnancy (Figure 20.4). Foetuses can be


Figure 20.9 Fertility<br />

delivered and examined for abnormalities according to procedures used in<br />

embryotoxicity studies. (Note that this study is almost identical to a<br />

modified OECD 421 study.) Combination with the ICH developmental<br />

toxicity study provides the equivalent <strong>of</strong> a two generation and<br />

embryotoxicity study.<br />

Interpretation <strong>of</strong> studies<br />

A.K.PALMER 297<br />

For regulatory agencies, one <strong>of</strong> the purposes <strong>of</strong> these tests is to gather<br />

information for labelling. By common consensus, a substance is labelled as<br />

a reproductive toxicant only if it induces effects at dosages below those<br />

causing systemic toxicity. Such labelling, however, can be very misleading<br />

especially with industrial chemicals. Even if the animal species can be<br />

shown to be a good surrogate for humans by means <strong>of</strong> kinetic and other<br />

studies, it is necessary to take into account the relationship between<br />

exposures causing effects and those likely to be encountered by humans.<br />

For example, a material can be labelled as a reproductive toxicant but<br />

present little or no real risk to humans because the effects are induced at<br />

exposures well in excess <strong>of</strong> those encountered by humans (Figure 20.10).<br />

Conversely, a substance not labelled as a reproductive toxicant could cause<br />

reproductive effects in humans if these are induced at exposures<br />

encountered by humans. In other words toxicity is relative, a matter <strong>of</strong><br />

dosage and situation. Labelling without reference to exposures is<br />

incomplete.


298 TESTING AND EVALUATION FOR REPRODUCTIVE TOXICITY<br />

Figure 20.10 Interpretation/extrapolation <strong>of</strong> reproductive toxicology.<br />

Conclusions<br />

In conclusion I should mention that time constraints enforce superficial<br />

mention <strong>of</strong> many important aspects. I would like to emphasise that<br />

improved testing and evaluation for toxicity to reproduction can be<br />

achieved with methodology that exists within and without regulatory<br />

guidelines. This has been illustrated by the special cases presented at this<br />

meeting.<br />

There is no necessity to be restricted to specific guidelines, there never<br />

was. We should make use <strong>of</strong> any and all test methods available as<br />

appropriate for the substance being investigated. Whatever the type <strong>of</strong><br />

substance, testing involves looking for the same hazards. Having identified<br />

a hazard, methods <strong>of</strong> assessing risk, essentially, are the same (although<br />

PBPK models for reproductive toxicity would be more complex than those<br />

used for systemic toxicity).<br />

The methodology is available, what is required is the willingness and<br />

wisdom to use it effectively and efficiently (Palmer, 1993a, b). The goal<br />

should be to investigate a specific substance to the extent necessary, no<br />

more and no less. For this there needs to be a change in attitude by<br />

industry, agencies and academia. Therein is the greatest problem since<br />

‘Change is not made without inconvenience, even from worse to better’ and<br />

people are very unwilling to change their prejudices and habits.


References<br />

A.K.PALMER 299<br />

PALMER, A.K., 1993a, Identifying environmental factors harmful to reproduction,<br />

Environm. Hlth Perspect. Supplements, 101(2), 19–25.<br />

PALMER, A.K., 1993b, Introduction to (pre)screening methods, Reproduct.<br />

Toxicol., 7, 95–8.


PART SIX<br />

Toxicity <strong>of</strong> selected classes <strong>of</strong> industrial<br />

chemicals


21<br />

Special Points in the Toxicity Assessment <strong>of</strong><br />

Colorants (Dyes and Pigments)<br />

HERMANN M.BOLT<br />

Institut für Arbeitsphysiologie an der Universität Dortmund,<br />

Dortmund<br />

Introduction<br />

Colorants (dyes and pigments) are very important industrial chemicals. A<br />

special point in the toxicological assessment <strong>of</strong> such compounds is their<br />

bioavailability upon inhalation. From the technological point <strong>of</strong> view<br />

pigments are colorants which are insoluble whereas dyes are soluble in the<br />

application mixture.<br />

Biologically, the most relevant route <strong>of</strong> potential exposure <strong>of</strong> humans to<br />

colorants is by inhalation. If a pigment is biologically insoluble, it may<br />

finally be removed from the airways by clearance mechanisms. However, in<br />

practice the situation is much more complicated. For instance, chromates<br />

are technically important pigments which are well investigated. Biochemical<br />

and toxicological research has shown that the common toxicological<br />

principle <strong>of</strong> chromates which penetrate the cell membrane and, after<br />

intracellular transformation, exert genotoxic effects, is the chromate anion<br />

(CrO ). In terms <strong>of</strong> inhalatory carcinogenicity, the very water-soluble<br />

alkali chromates and the practically insoluble lead chromate have the<br />

lowest potency. Pigments <strong>of</strong> an intermediate solubility, e.g. calcium<br />

chromate, zinc chromate, strontium chromate, have a high carcinogenic<br />

potency on the respiratory tract. Local storage <strong>of</strong> chromate particles in the<br />

airways with a slow but continuous local release <strong>of</strong> CrO seems therefore<br />

to be an important factor in respiratory carcinogenesis induced by<br />

chromates. But also lead chromate, which is technically regarded as<br />

insoluble, is bioavailable to some extent; this is visualized by practical cases<br />

<strong>of</strong> occupational lead chromate exposure which display markedly elevated<br />

blood lead levels.<br />

The question <strong>of</strong> systemic bioavailability, upon inhalation, became <strong>of</strong><br />

recent regulatory importance for azo colorants based on carcinogenic<br />

aromatic amines. This problem has already been addressed in detail<br />

(Myslak and Bolt, 1988).


302 SPECIAL POINTS IN THE TOXICITY ASSESSMENT OF COLORANTS<br />

Table 21.1 Number <strong>of</strong> azo colorants based on cancerogenic aromatic amines (2napthylamine,<br />

benzidine and its derivatives) listed in Colour Index (3rd edn, 3rd<br />

Rev., 1987)<br />

The problem <strong>of</strong> carcinogenic azo colorants<br />

In the past, azo colorants based on benzidene, 3,3′-dichlorobenzidine, 3,3′dimethylbenzidine<br />

(o-tolidine), and 3,3'-dimethoxybenzidine (odianisidine)<br />

have been synthesized in large amounts and numbers,<br />

especially in the German chemical industry. The Colour Index (1987) lists<br />

a total number <strong>of</strong> more than 2000 azodyes, 452 <strong>of</strong> them being based on 2naphthylamine,<br />

benzidine, or benzidine derivatives (Table 21.1).<br />

Azo colorants have a number <strong>of</strong> properties that have made them<br />

invaluable for dyeing a wide variety <strong>of</strong> materials, including natural and<br />

artificial fibres, plastics, resins, textiles, leather, paper, glass, ceramics,<br />

cement, inks, printing inks, chalks, crayons and carbon papers, as well as<br />

cosmetics, food and beverages. Interesting with respect to potential<br />

exposure <strong>of</strong> painters is the use <strong>of</strong> azo colorants in the coloring <strong>of</strong> oil-,<br />

resins-, emulsion-, lime-, and other aqueousbased paints, distempers,<br />

transparent laquers, spirit and oil wood stains, and varnishes (Colour<br />

Index, 1987). In all these fields, particularly benzidine-based azo colorants<br />

have found widespread use (Gregory, 1984).<br />

In the UK, the Carcinogenic Substances Regulation led in 1967 to<br />

discontinuation <strong>of</strong> the use <strong>of</strong> benzidine in the production <strong>of</strong> azo colorants<br />

(Martin and Kennelly, 1985). The US government in 1974 promulgated<br />

regulations to control benzidine at the workplace (Gregory, 1984).<br />

Nevertheless, in the period <strong>of</strong> 1972–4, more than 150000 persons in the<br />

USA were potentially occupationally exposed to benzidine-based colorants<br />

(Gregory, 1984); in 1978, approximately 1.7 million US pounds <strong>of</strong><br />

benzidine-based azo colorants were manufactured, and a further 1.6<br />

million pounds were imported into the USA (Lynn et al., 1980).<br />

In Germany, over 30 different benzidine-based azo colorants were<br />

manufactured in the early 1960s. The manufacture <strong>of</strong> these colorants was<br />

stopped in 1971, with the exception <strong>of</strong> one dye (Direct Black 4; C.I. No.<br />

30245); the manufacture <strong>of</strong> the latter was continued until 1973. Azo


H.M.BOLT 303<br />

colorants based on carcinogenic congeners <strong>of</strong> benzidine (e.g. 3,3′dimethoxybenzidine;<br />

3,3′-dimethylbenzidine) are most likely still being<br />

manufactured in some countries. The case <strong>of</strong> pigments based on 3,3′dichlorobenzidine<br />

is discussed below.<br />

Azo colorants are biologically active through their metabolites.<br />

Azoreduction <strong>of</strong> these compounds occurs in vivo (Radomski and<br />

Mellinger, 1962; Rinde and Troll, 1975; Robens et al., 1980) by an<br />

enzyme-mediated reaction. Azoreductases are found in mammalian tissues,<br />

particularly in liver (Fouts et al., 1957; Walker, 1970; Martin and Kennelly,<br />

1981; Kennelly et al., 1982) and also in gut bacteria (Yoshida and<br />

Miyakawa, 1973; Chung et al., 1978; Hartmann et al., 1978; Cerniglia et<br />

al., 1982; Bos et al., 1986). The result <strong>of</strong> this azoreduction is the release <strong>of</strong><br />

the (carcinogenic) aromatic amine from the colorant (Martin and Kennelly,<br />

1985). Studies performed on exposed workers have demonstrated that the<br />

azoreduction <strong>of</strong> benzidine-based colorants occurs in man (Genin, 1977;<br />

Boeninger, 1978; Lowry et al., 1980; Meal et al., 1981; Dewan et al.,<br />

1988). Studies <strong>of</strong> Lynn et al., (1980) and Bowman et al. (1983) have<br />

demonstrated that the metabolic conversion <strong>of</strong> benzidine-, 3,3′dimethylbenzidine-<br />

and 3,3′-dimethoxybenzidine-based colorants to their<br />

(carcinogenic) amine precursors in vivo is a general phenomenon that must<br />

be expected for each member <strong>of</strong> this class <strong>of</strong> chemicals.<br />

However, in contrast to water-soluble dyes, the question <strong>of</strong> biological<br />

azoreduction <strong>of</strong> (practically insoluble) pigments was a matter <strong>of</strong> discussion<br />

in the recent years. One study has claimed the presence <strong>of</strong> 3,3'dichlorobenzidine<br />

in the urine both <strong>of</strong> experimental animals fed with<br />

Pigment Yellow 12 and <strong>of</strong> exposed workers (Akiyama, 1970). However,<br />

other experimental studies, using more modern analytical tools, did not<br />

confirm these results (DHEW, 1978; Leuschner, 1978; Mondino et al.,<br />

1978; Nony et al., 1980).<br />

Several epidemiological studies have demonstrated that the use <strong>of</strong> the<br />

benzidine-based dyes has caused bladder cancer in humans. In a Japanese<br />

study, the risk <strong>of</strong> bladder cancer among dye applicators (kimono painters)<br />

was 6.8 times the expected rate (Yoshida et al., 1971). In a British study,<br />

workers performing the dyeing <strong>of</strong> textiles (and not exposed to benzidine<br />

itself) had a higher risk <strong>of</strong> bladder cancer (RR=3.4) than expected<br />

(Anthony, 1974). In a USSR study, an increased incidence <strong>of</strong> bladder<br />

cancer was found in workers who dried or ground benzidine-based dyes<br />

(Genin, 1977).<br />

In our own study on bladder cancer in painters (Myslak et al., 1991), the<br />

time <strong>of</strong> first exposure (painters with bladder tumors) dated mostly back to<br />

the first half <strong>of</strong> the century. Two factors may have been relevant: (1) at<br />

that time, a large number <strong>of</strong> benzidine-based azodyes was in manufacture,<br />

especially in Germany; (2) during that time it was usual for painters in<br />

Germany to prepare their paints themselves. This work included grinding


304 SPECIAL POINTS IN THE TOXICITY ASSESSMENT OF COLORANTS<br />

and mixing <strong>of</strong> the dyes and preparation <strong>of</strong> the coloring mixture by addition<br />

<strong>of</strong> solvents. All painters we had interviewed reported that this type <strong>of</strong> work<br />

had been regularly associated with considerable occurrence <strong>of</strong> dye dust in<br />

the atmosphere, up to the end <strong>of</strong> the 1950s.<br />

The very long latency period may explain why an enhanced risk <strong>of</strong> bladder<br />

cancer in German painters (due to previous exposure to azo dyes) is<br />

observed even today. Similar arguments have also been put forward for<br />

other occupational groups associated with an increased risk <strong>of</strong> bladder<br />

cancer, and where a causal connection with benzidine-based azo dyes had<br />

been proven or suggested, e.g. for textile dyers (Jenkins, 1978), leather<br />

dyers and shoeworkers (Decouflé, 1979), hairdressers (Guberan et al.,<br />

1985), and tailors (Anthony and Thomas, 1970). The results <strong>of</strong> our own<br />

survey <strong>of</strong> painters are very probably not relevant for the present working<br />

conditions in Germany and other highly industrialized countries, because<br />

<strong>of</strong> different materials, working methods, and hygienic standards introduced<br />

in recent years. They are, however, quite relevant for matters <strong>of</strong><br />

compensation <strong>of</strong> persons who are now diseased.<br />

Regulatory aspects (FRG)<br />

The arguments described above have led the German Commission for<br />

Investigation <strong>of</strong> Health Hazards <strong>of</strong> Chemical <strong>Compounds</strong> in the Work<br />

Area (MAK-Commission) to include the following chapter in the MAKlist,<br />

since 1988 (DFG, 1988):<br />

Azo colorants are characterized by the azo group -N=N-. They are<br />

made by the coupling <strong>of</strong> singly and multiply diazotized aryl amines.<br />

Of particular toxicological importance are colorants from double<br />

diazotized benzidine and from benzidine derivatives (3,3′dimethylbenzidine,<br />

3,3′-dimethoxybenzidine, 3,3′-dichlorobenzidine).<br />

In addition, aminoazo-benzene, aminonaphthalene and monocyclic<br />

aromatic amines are encountered. Reductive fission <strong>of</strong> the azo group,<br />

either by intestinal bacteria or by azo reductases <strong>of</strong> the liver and<br />

extrahepatic tissues, can cause these compounds to be released. Such<br />

breakdown products have been detected in animal experiments as<br />

well as in man (urine). Mutagenicity, which has been observed with<br />

numerous azo colorants in in-vitro test systems, and the<br />

carcinogenicity in animal experiments are attributed to the release <strong>of</strong><br />

amines and their subsequent metabolic activation. There are now<br />

epidemiological indications that occupational exposure to benzidinebased<br />

azo colorants can increase the incidence <strong>of</strong> bladder carcinomas.<br />

Thus, all azo colorants whose metabolism can liberate a carcinogenic<br />

aryl amine are suspected <strong>of</strong> having carcinogenic potential. Due to the<br />

large number <strong>of</strong> such dyes (several hundred) it seems neither possible


nor justifiable to substantiate this suspicion in each individual case by<br />

means <strong>of</strong> animal experimentation according to customary<br />

classification criteria. Instead, scientifically justifiable models have to<br />

be relied on. Therefore, as a preventive measure to avoid putting<br />

exposed persons at risk, it is recommended that the substances be dealt<br />

with as if they were classified in the same categories as the<br />

corresponding carcinogenic or suspected carcinogenic amines (A1, A2,<br />

B)<br />

If there are indications that the colorant itself (e.g. a pigment) or<br />

any carcinogenic breakdown products are not biologically available,<br />

the exclusion <strong>of</strong> risk should be experimentally proven or<br />

substantiated by biomonitoring. Suitable animal experiments can also<br />

rule out suspicion <strong>of</strong> carcinogenic potential.<br />

On the basis <strong>of</strong> this general view, which had been endorsed by the German<br />

Ministry <strong>of</strong> Labor (TGS 900, Bundesarbeitsblatt 1/1990, p. 63), the<br />

identification <strong>of</strong> the aromatic amine component <strong>of</strong> azo colorants is <strong>of</strong> key<br />

importance. A suitable compilation <strong>of</strong> azo colorants, according to their<br />

aromatic amine components, has been published by Myslak (1990).<br />

Azo pigments<br />

H.M.BOLT 305<br />

The postulate <strong>of</strong> further research on the bioavailability and/or<br />

carcinogenicity <strong>of</strong> azo pigments, especially those based on 3,3′dichlorobenzidine<br />

(v.s.), has focused interest on this particular problem.<br />

Azo pigments based on 3,3′-dichlorobenzidine (e.g. Pigment Yellow 12,<br />

Pigment Yellow 13, Pigment Yellow 14) have been orally administered to<br />

rats, hamsters, rabbits and monkeys, at doses up to 400 mg pigment kg −1<br />

b.w. (Leuschner, 1978; Mondino et al., 1978; DHEW, 1978; Nony et al.,<br />

1980; Decad et al., 1983; Sagelsdorff et al., 1990; Hoechst AG,<br />

unpublished data). These authors could not find 3,3'-dichlorobenzidine in<br />

the urine <strong>of</strong> animals treated with 3,3'-dichlorobenzidine pigments. Decad<br />

et al. (1983) demonstrated that 14 C-labelled Pigment Yellow 12, orally<br />

administered to rats, was completely excreted in the faeces. On the basis <strong>of</strong><br />

these investigations <strong>of</strong> disposition <strong>of</strong> 3,3′-dichlorobenzidine-based<br />

pigments, it is clear why none <strong>of</strong> the long-term animal carcinogenicity<br />

studies performed so far (Leuschner, 1978; DHWE 1978; ICI, unpublished<br />

data) has demonstrated a carcinogenic effect <strong>of</strong> a diaryl pigment.<br />

It therefore appears that the aromatic amine components from azo<br />

pigments are practically not bioavailable, as demonstrated for several<br />

pigments on the basis <strong>of</strong> 3,3′-dichlorobenzidine (ETAD, 1990; see also Bolt<br />

and Golka, 1993). Hence, it is now very unlikely that occupational<br />

exposure to insoluble azo pigments would be associated with a substantial<br />

risk <strong>of</strong> (bladder) cancer in man.


306 SPECIAL POINTS IN THE TOXICITY ASSESSMENT OF COLORANTS<br />

References<br />

AKIYAMA, T., 1970, The investigation on the manufacturing plant <strong>of</strong> organic<br />

pigment, Jikei Med. J., 17, 1–9.<br />

ANTHONY, H.M., 1974, <strong>Industrial</strong> exposure in patients with carcinoma <strong>of</strong> the<br />

bladder, J. Soc. Occup. Med., 24, 110–16.<br />

ANTHONY, H.M. and THOMAS, G.M., 1970, Tumors <strong>of</strong> the urinary bladder:<br />

An analysis <strong>of</strong> the occupations <strong>of</strong> 1030 patients in Leeds, England, J. Nat.<br />

Cancer Inst., 45, 879–95.<br />

BOENINGER, H., 1978, An investigation <strong>of</strong> the metabolic reduction <strong>of</strong> benzidine<br />

azo dyes to benzidine and its metabolites and their possible relationship to<br />

carcinoma <strong>of</strong> the bladder in man. Unpublished data (cited by Gregory, 1984).<br />

BOLT, H. M and GOLKA, K., 1993, Zur früheren Exposition von Malern<br />

gegenüber Az<strong>of</strong>arbst<strong>of</strong>fen, Arbeitsmed., Sozialmed., Umweltmed., 28, 417–21.<br />

Bos, R.P., VAN DER KRIEKEN, W., SMEIJSTERS, L., KOOPMAN, J.P.,<br />

DEJONGE, H.R., THEUWS, J.L.G. and HENDERSON, P.T., 1986, Internal<br />

exposure <strong>of</strong> rats to benzidine derived from orally administered benzidine-based<br />

dyes after intestinal azo reduction, <strong>Toxicology</strong>, 40, 207–13.<br />

BOWMAN, M.C., NONY, C.R., BILLEDEAU, S.M., MARTIN, J.L. and<br />

THOMPSON, H.C., 1983, Metabolism <strong>of</strong> nine benzidine-congener-based azo<br />

dyes in rats based on gas chromatographic assays <strong>of</strong> the urine for potentially<br />

carcinogenic metabolites, J. Anal. Toxicol. 7, 55–60.<br />

CERNIGLIA, C.E., FREEMAN, J.P., FRANKLIN, W. and PACK, L.D., 1982,<br />

Metabolism <strong>of</strong> azo dyes derived from benzidine, 3,3'-dimethylbenzidine and 3,<br />

3'-dimethoxybenzidine to potentially carcinogenic aromatic amines by<br />

intestinal bacteria, Carcinogenesis, 3, 1255–60.<br />

CHUNG, K.T., FULK, G.E. and EGAN, M., 1978, Reduction <strong>of</strong> azo dyes by<br />

intestinal anaerobes, Appl. Environ. Microbiol. 35, 55–62.<br />

Colour Index, 3rd edn., 3rd rev., 1987, Bradford: Society <strong>of</strong> Dyers and Colourists.<br />

Vols. 1–8.<br />

DECAD, G.M., SNYDER, C.D. and MITONA, C., 1983, Fate <strong>of</strong> water-insoluble<br />

and water-soluble dichlorobenzidine-based pigments, J. Toxicol. Environm.<br />

Hlth, 11, 455–65.<br />

DECOUFLÉ, P., 1979, Cancer risk associated with employment in the leather and<br />

leather products industry, Arch. Environm. Hlth, 34, 33–7.<br />

DFG (Deutsche Forschungsgemeinschaft), 1988, List <strong>of</strong> MAK and BAT Values<br />

1988, Weinheim: VCH Publishers.<br />

DEWAN, A., JANI, J.P., PATEL, J.S., GANDHI, D.N., VARIYA, M.R. and<br />

GHODSARA, N.B., 1988, Benzidine and its acetylated metabolites in the urine<br />

<strong>of</strong> workers exposed to Direct Black 38, Arch. Environm. Hlth, 43, 269–72.<br />

DHEW, 1978, Bioassay <strong>of</strong> diarylanilide yellow for possible carcinogenicity, DHEW<br />

Publication No. (NIH) 78–830, US Dept <strong>of</strong> Health, Education and Welfare,<br />

Public Health Service, National Cancer Institute, Carcinogens Testing Program.<br />

ETAD (Ecological and Toxicological Association <strong>of</strong> the Dyestuffs Manufacturing<br />

Industry, 1990, Zum kanzerogenen Potential von Diaryl-Azopigmenten auf<br />

Basis von 3,3′-Dichlorbenzidin, ETAD-Bericht T 2028-BB (D), ETAD,<br />

CH-4005, Basel 5.


H.M.BOLT 307<br />

FOUTS, J.R., KAMM, J.J. and BRODIE, B.B., 1957, Enzymatic reduction <strong>of</strong> prontosil<br />

and other azo dyes, J. Pharmacol, Exp. Ther., 110, 291–300.<br />

GENIN, W.A., 1977, Formation <strong>of</strong> clastogenic diphenylamine derivates as a result<br />

<strong>of</strong> the metabolism <strong>of</strong> direct azo dyes, Vopr. Oncol, 23, 50–2 (in Russian).<br />

GREGORY, A., 1984, The carcinogenic potential <strong>of</strong> benzidine-based dyes, J.<br />

Environm. Pathol. Toxicol. Oncol, 5, 243–59.<br />

GUBERAN, R., RAYMOND, L. and SWEETNAM, P.M., 1985, Increased risk for<br />

male bladder cancer among a cohort <strong>of</strong> male and female hairdressers from<br />

Geneva, Int. J. Epidemiol, 14, 549–54.<br />

HARTMANN, C.P., FULK, G.E. and ANDREWS, A.W., 1978, Azo reduction <strong>of</strong><br />

trypan blue to a known carcinogen by a cellfree extract <strong>of</strong> a human<br />

intestinal anaerobe, Mutat. Res., 58, 125–32.<br />

JENKINS, C.L., 1978, Textile dyes are potential hazards, J. Environm. Hlth, 40,<br />

279– 84.<br />

KENNELLY, J.C., HERTZOG, P.J. and MARTIN, C.N., 1982, The release <strong>of</strong> 4,4′diaminobiphenyls<br />

from azo dyes in the rat, Carcinogenesis, 3, 947–51.<br />

LEUSCHNER, F., 1978, Carcinogenicity studies on different diarylide yellow<br />

pigments in mice and rats, Toxicol. Lett., 2, 253–60.<br />

LOWRY, L.K., TOLOS, W.P., BOENINGER, M.F., NONY, C.R. and<br />

BOWMAN, M.C., 1980, Chemical monitoring <strong>of</strong> urine from workers<br />

potentially exposed to benzidine-derived azo dyes, Toxicol. Lett., 7, 29–36.<br />

LYNN, R.K., DONIELSON, D.W., ILIAS, A.M., KENNISH, J.M., WONG, K. and<br />

MATHEWS, H.B., 1980, Metabolism <strong>of</strong> bisazobiphenyl dyes derived from<br />

benzidine, 3,3′-dimethylbenzidine or 3,3′-dimethoxybenzidine to carcinogenic<br />

aromatic amines in the dog and rat, Toxicol Appl. Pharmacol, 56, 248–58.<br />

MARTIN, C.N. and KENNELLY, J.C., 1981, Rat liver microsomal azoreductase<br />

activity on four azo dyes derived from benzidine, 3,3′-dimethylbenzidine or 3,3′dimethoxybenzidine,<br />

Carcinogenesis, 2, 307–12.<br />

MARTIN, C.N. and KENNELLY, J.C., 1985, Metabolism, mutagenicity and DNA<br />

biding <strong>of</strong> biphenyl-based azo dyes, Drug Metab. Rev., 16, 89–117.<br />

MEAL, P.F., COCKER, J., WILSON, H.K. and GILMOUR, J.M., 1981, Search for<br />

benzidine and its metabolites in urine <strong>of</strong> workers weighing benzidine-derived<br />

dyes, Br. J. Ind. Med., 38, 191–3.<br />

MONDINO, A., ACHARI, R., DUBINI, M., MARCHISIO, M.A., SILVESTRI, S.<br />

and ZANOLO, G., 1978, Absence <strong>of</strong> dichlorobenzidine in the urine <strong>of</strong> rats,<br />

rabbits and monkeys treated with C.I. Pigment Yellow 13, Med. Lav., 69, 693–<br />

7.<br />

MYSLAK, Z.W., 1990, Az<strong>of</strong>arbmittel auf der Basis krebserzeugender und<br />

verdächtiger aromatischer Amine. Identification, Verwendungsbereiche,<br />

Herstellungszeiträume. Schriftenreihe der Bundesanstalt für Arbeitsschutz, GA<br />

35, Bremerhaven: Wissenschaftsverlag NW.<br />

MYSLAK, Z.W. and BOLT, H.M., 1988, Berufliche Exposition gegenüber<br />

Az<strong>of</strong>arbst<strong>of</strong>fen und Harnblasenkarzinom-Risiko, Zbl. Arbeitsmed., 10, 310–<br />

21.<br />

MYSLAK, Z.W., BOLT, H.M. and BROCKMANN, W., 1991, Tumors <strong>of</strong> the<br />

urinary bladder in painters: a case-control study, Am. J. Ind. Med., 19, 705–13.<br />

NONY, C.R., BOWMAN, M.C., CAIRNS, T., LOWRY, L.K. and TOLOS, W.P.,<br />

1980, Metabolism studies <strong>of</strong> an azo dye and pigment in the hamster based on


308 SPECIAL POINTS IN THE TOXICITY ASSESSMENT OF COLORANTS<br />

analysis <strong>of</strong> the urine for potentially carcinogenic aromatic amine metabolites.<br />

J. Anal. Toxicol, 4, 132–40.<br />

RADOMSKI, J.L. and MELLINGER, T.J., 1962, The absorption, fate and<br />

excretion in rats <strong>of</strong> the water-soluble azo dyes, FD&C Red No. 2. FD&C Red<br />

No. 4 and FD&C Yellow No. 6, J. Pharmacol. Exp. Ther., 136, 259–66.<br />

RINDE, E. and TROLL, W., 1975, Metabolic reduction <strong>of</strong> benzidine azo dyes to<br />

benzidine in the Rhesus monkey, J. Nat. Cancer Inst., 55, 181–2.<br />

ROBENS, J.F., DILL, G.S., WARD, J.M., JOINER, J.R., GRIESEMER, R.A. and<br />

DOUGLAS, J.F., 1980, Thirteen-week subchronic toxicity studies <strong>of</strong> Direct<br />

Blue 6, Direct Black 38 and Direct Brown 95 dyes, Toxicol. Appl. Pharmacol.,<br />

54, 431–42.<br />

SAGELSDORFF, P., JOPPICH-KUHN, M. and JOPPICH, M., 1990,<br />

Biomonitoring for the bioavailability <strong>of</strong> dichlorobenzidine from<br />

dichlorobenzidine-based dyes, J. Cancer Res. Clin. Oncol, 116, 79 (abstract).<br />

WALKER, R., 1970, The metabolism <strong>of</strong> azo compounds, a review <strong>of</strong> the literature,<br />

Food Cosmet. Toxicol, 8, 659–76.<br />

YOSHIDA, O., HARADA, T., MIYAKAWA, M. and KATO, T., 1971, Bladder<br />

cancer among dyers in the Kyoto area, Igaku Ayumi, 79, 421–2 (in Japanese).<br />

YOSHIDA, O. and MIYAKAWA, M., 1973, Etiology <strong>of</strong> bladder cancer: metabolic<br />

aspects, in Nakahara, W., Hirayama, T., Nishioka, K. and Sugano, H. (Eds)<br />

Analytical and Experimental Epidemiology <strong>of</strong> Cancer, Proc. <strong>of</strong> the 3rd Int.<br />

Symp. <strong>of</strong> Princess Takamatsu Cancer Research Fund, pp. 31–9, Baltimore:<br />

University Park Press.


22<br />

<strong>Toxicology</strong> <strong>of</strong> Textile Chemicals<br />

DIETER SEDLAK<br />

EnviroTex GmbH, Augsburg<br />

The former main aspects <strong>of</strong> textiles like fashion or usefulness seem to be<br />

pushed back by a new phenomenon, the ecological and toxicological aspect<br />

<strong>of</strong> textiles. Although everybody is talking about textiles in this context,<br />

everybody means the textile chemicals (and dye-stuffs) on the textile. With<br />

respect to this situation we have to consider new developments in Germany<br />

(Figure 22.1).<br />

For two years now textile finishing plants have to be approved within<br />

the German federal immission control act. This means that all sorts <strong>of</strong><br />

immissions to the working place or the surroundings <strong>of</strong> the plant must be<br />

defined in quantity and quality. In the meantime so-called emission factors<br />

have been developed. But this project is not yet finished. Even for insiders<br />

it was surprising how many textile chemicals show unexpected behaviour<br />

during their application. The reasons are:<br />

– unknown byproducts or impurities,<br />

– unknown reactivities between components,<br />

– unknown interactions with substrate,<br />

– unknown dependencies <strong>of</strong> process parameters.<br />

Today producers and users realise that the inherent toxicological properties<br />

<strong>of</strong> many textile chemicals evaluated within a typical safety data sheet<br />

represent only a part <strong>of</strong> the whole knowledge you need for safe handling<br />

and processing.<br />

A further interesting development undoubtedly is the discussion around<br />

the ecolabelling <strong>of</strong> textiles. There definitely is some danger so that different<br />

labels based on different commercial interests should be evaluated. A<br />

positive step could be the fact that MST and Ecotex have joined. But other<br />

societies like TÜV or GSF create their own labels including statements that<br />

they use the better (right) label criteria. This leads quite clearly to total<br />

confusion for the consumer. However, what the textile industry needs<br />

today is confidence. The developments discussed above will not help. From<br />

a technical point <strong>of</strong> view we have the same problem as discussed with<br />

respect to the immissions. Too little is known about the real properties and


310 TOXICOLOGY OF TEXTILE CHEMICALS<br />

Figure 22.1 New developments in Germany regarding toxicology <strong>of</strong> textile<br />

chemicals.<br />

behaviour <strong>of</strong> textile chemicals to define the absolute label criteria. These<br />

uncertainties can be solved only by cooperation <strong>of</strong> the different groups and<br />

not by aggressive competition. How can we handle both developments in a<br />

proper way?<br />

At first we have to be clear about the interactions <strong>of</strong> both problem fields.<br />

Let us start with the textile chemical delivered to a finishing plant<br />

(Figures 22.2 and 22.3). Following the directives for dangerous substances<br />

or safety data sheets the product is exactly labelled and its toxicological


Figure 22.2 Possible levels to discuss the toxicology <strong>of</strong> textile chemicals.<br />

D.SEDLAK 311<br />

properties are well described in the SDS. This is only as good as the<br />

information given to the product safety manager responsible for the<br />

possible ways <strong>of</strong> handling and processing. However, in most cases textile<br />

chemicals can be described as harmless.<br />

Nearly all show an acute oral toxicity greater than 2000 mg kg −1<br />

although they may contain toxic substances in diluted form. Only a few <strong>of</strong><br />

them possess an irritant character or even CMT properties. Nevertheless,<br />

many <strong>of</strong> them have impurities with these properties which are given more<br />

and more, even in low concentrations. This is a very important process


312 TOXICOLOGY OF TEXTILE CHEMICALS<br />

Figure 22.3 Toxicological pr<strong>of</strong>ile <strong>of</strong> textile chemicals.<br />

because these low concentrations may also lead to high vapour<br />

concentrations at the workplace if these substances are volatile. This is<br />

mostly the case.<br />

For example, the carcinogenic substance acrylonitrile has a product label<br />

only giving a concentration <strong>of</strong> 0.2 percent and more in the corresponding<br />

polymer dispersion. Even 0.02 percent may lead to workplace<br />

concentrations a hundred fold higher than the TLV allows. Product data<br />

sheets with these ‘properties’ still exist with no additional information<br />

because it is not needed!<br />

Another problem is that nearly all textile chemicals are exposed to<br />

temperatures between 100°C and 230°C during application which leads to<br />

many only poorly defined substances which may all be set free in the<br />

workplace and the surroundings. Here we are also confronted with a<br />

typical juristic phenomenon. Many suppliers do not take any responsibility<br />

regarding these questions if the user defines his process parameters himself,<br />

especially when using recipe components from different suppliers. But<br />

everybody knows that the user has not the means to evaluate the<br />

toxicological pr<strong>of</strong>ile <strong>of</strong> his chemicals mixture and process. This lack <strong>of</strong><br />

responsibility should be clarified.<br />

Now let us observe the result <strong>of</strong> such chemical treatment <strong>of</strong> textiles.<br />

Normally you will only discuss the summarised toxicological pr<strong>of</strong>iles <strong>of</strong> the<br />

used chemicals in an additive way. This means that the combination <strong>of</strong><br />

different products—all with no acute toxicity—will also show no acute


Figure 22.4 Typical composition <strong>of</strong> a flame retardant recipe.<br />

D.SEDLAK 313<br />

toxicity. Furthermore the concentration <strong>of</strong> the active ingredients in the new<br />

matrix textile is much lower than in the matrix water most textile<br />

chemicals are based on. This strategy seems to be appropriate. On the<br />

other hand take the irritant property <strong>of</strong>, for example, fatty amine based<br />

emulsifier. This property is lost during formulation <strong>of</strong> the emulsifier in the<br />

textile chemical by homogeneous dilution. The average concentration after<br />

application to the textile is surely lower. But who has information about<br />

the actual form <strong>of</strong> this fatty amine in the textile. Is it distributed<br />

homogeneously, is it more located at the surface, does it interact with other<br />

substances and even lose its irritant character? Many questions seem to be<br />

unanswered.<br />

Both complex questions—the toxicology <strong>of</strong> the handling and processing<br />

<strong>of</strong> textile chemicals and the toxicology <strong>of</strong> the result <strong>of</strong> the processing on<br />

textiles— will be discussed by means <strong>of</strong> an admittedly drastic example, a<br />

flame retardant process. This example is rather representative because an


314 TOXICOLOGY OF TEXTILE CHEMICALS<br />

Figure 22.5 Total composition <strong>of</strong> a flame retardant recipe.<br />

EEC-directive is ‘under construction’ just now which demands the finishing<br />

<strong>of</strong> all upholstery covers with flame retardant chemicals.<br />

The composition <strong>of</strong> this recipe, shown in Figure 22.4, doesn’t seem to be<br />

too complex. No special toxicological properties are apparent. The<br />

corrosive character <strong>of</strong> the phosporic acid vanishes during the application


D.SEDLAK 315<br />

Figure 22.6 Release <strong>of</strong> chemical substances during/after a flame retardant process.<br />

by neutralizing the finished textile with alkalis. Complex reactions <strong>of</strong> the<br />

different components are expected which are supposed to form not well<br />

defined polycondensates fixed to the textile fibre. On this basis there is no<br />

apparent reason to think <strong>of</strong> any toxicological side effects.<br />

In Figure 22.5, however, you will get a good idea about the real<br />

situation. But this detailed composition should not be deceptive about the<br />

fact that this is the ‘wanted’ technical composition verified by a few<br />

analytical data. In this case about 500 actual substances can be expected.<br />

Even if we should have available all the necessary toxicological data for the<br />

components it would not help, because there is poor information about the<br />

result <strong>of</strong> the finishing process itself. This is the only reality.<br />

A first step in the right direction would be the analysis <strong>of</strong> the process<br />

<strong>of</strong>fgas. The result reflects approximately the activities on the textile during


316 TOXICOLOGY OF TEXTILE CHEMICALS<br />

Figure 22.7 Needs for the textile toxicology assessment.<br />

processing. Figure 22.6 shows in which way this knowledge can be used to<br />

perform a proper risk assessment based on defined substances. However, in<br />

the case <strong>of</strong> evaluation <strong>of</strong> the skin toxicity we are confronted with a lack <strong>of</strong><br />

data concerning the bio-availability <strong>of</strong> the named substances. Not before<br />

having cleared these additional questions can the toxicologist seriously<br />

start his routine activities, the risk assessment. Anyway this seems to be<br />

much easier than to discover the chemical basis for this assessment.<br />

With respect to the toxicology <strong>of</strong> textile chemicals there is a lot to do<br />

mostly in the field <strong>of</strong> understanding the chemicals and processes used.<br />

After this we recommend the development <strong>of</strong> models for evaluating the<br />

bioavailability <strong>of</strong> substances and test kits to characterize the toxicological<br />

properties <strong>of</strong> textiles as a whole. Last but not least we hope for much<br />

better communication between the industries involved (Figure 22.7).


23<br />

Antioxidants and Light Stabilisers : Toxic Effects<br />

<strong>of</strong> 3,5-Di-alkyl-4hydroxyphenyl Propionic Acid<br />

Derivatives in the Rat and their Relevance for<br />

Human Safety Evaluation<br />

HELMUT THOMAS, PETER DOLLENMEIER, ELKE<br />

PERSOHN, HANSJÖRG WEIDELI and FELIX WAECHTER<br />

Ciba-Geigy Limited, Basle<br />

Introduction<br />

Antioxidants and light stabilisers are important additives for a wide range<br />

<strong>of</strong> plastic materials used for industrial as well as for medicinal and food<br />

packaging purposes. Technical efficiency requires that these compounds are<br />

mobile within the polymer network. This implies that humans may be<br />

exposed to such compounds not only during the manufacturing process but<br />

also in the course <strong>of</strong> the decay <strong>of</strong> polymers, by direct contact with<br />

substances that have migrated to the surface <strong>of</strong> plastic materials or by<br />

ingestion <strong>of</strong> diffusion-contaminated food. It is <strong>of</strong> considerable interest,<br />

therefore, to be aware <strong>of</strong> the toxicological properties <strong>of</strong> these compounds<br />

and the relevance <strong>of</strong> these properties for the safety assessment in humans.<br />

Sterically hindered phenolic antioxidants<br />

Phenolic antioxidants have been widely used as food preservatives and<br />

their almost classical representatives, 2,6-di-tert-butyl-4-methyl phenol<br />

(BHT) and 2-tert-butyl-4-methoxyphenol (BHA) have been characterised in<br />

extenso with respect to their biochemical and toxicological properties<br />

(Søndergaard and Olsen, 1982; Conning and Phillips, 1986; Ito et al.,<br />

1986a; Williams et al., 1990a,b; Verhagen et al., 1991). Being nongenotoxic,<br />

these compounds were found upon oral administration to<br />

rodents to cause slightly increased liver weight, to induce mainly epoxide<br />

hydrolase and phase II drug metabolising enzymes and to exert some anticarcinogenic<br />

effect (Benson et al., 1979; Choe et al., 1984; Gregus and<br />

Klaassen, 1988; Prochaska and Talalay, 1988; Perchellet and Perchellet,<br />

1989; Rodrigues et al., 1991). Pulmonary toxicity and carcinogenicity<br />

particularly <strong>of</strong> BHT in the mouse and formation <strong>of</strong> forestomach carcinoma<br />

and papilloma in the rat and Syrian hamster, respectively, have been<br />

reported and are considered to be largely species-specific effects (Abraham<br />

et al., 1986; Anon, 1986; Ito et al., 1986a, b; Verhagen et al., 1989).<br />

<strong>Industrial</strong> phenolic antioxidants as used in the polymer technology, by


318 ANTIOXIDANTS AND LIGHT STABILISERS: TOXIC EFFECT<br />

contrast, although related to BHT, require higher molecular weights and<br />

hydrophobicity in order to retain them in the polymer matrix. This can be<br />

achieved, for example, by introducing alkyl chains or (esterified)<br />

carboxyalkyl moieties in the para-position to the phenolic hydroxyl group<br />

with variations in the aliphatic substitution pattern <strong>of</strong> the 2- and 6positions.<br />

With these modifications the questions arose, whether or not the<br />

toxicology <strong>of</strong> the chemically modified <strong>of</strong>fspring would still be related to<br />

that <strong>of</strong> the phenolic core <strong>of</strong> the BHT ancestor or completely different,<br />

although a common metabolic degradation pathway to structural entities<br />

resembling BHT could be anticipated. Clarification <strong>of</strong> this question was<br />

expected to contribute to the understanding <strong>of</strong> the toxicology <strong>of</strong> an entire<br />

class <strong>of</strong> phenolic antioxidants. A number <strong>of</strong> differentially esterified 4hydroxy-3,<br />

5-dialkyl-phenylpropionic acid derivatives were subsequently<br />

subjected to subchronic toxicity testing in rats with the result that the<br />

effects encountered were largely dependent on the alcohol moiety used for<br />

esterification and the size <strong>of</strong> the alkyl-substituent in the 3- and 5-positions:<br />

most frequently, increased liver weights were encountered in compounds<br />

with bulky 3,5-substituents such as tert-butyl. In some instances, however,<br />

initial hepatomegaly was followed after several weeks or months <strong>of</strong><br />

treatment by increases in thyroid weights and a proliferation <strong>of</strong> the thyroid<br />

follicular epithelium. The mechanism leading to the latter effect has been<br />

investigated in detail using the di-ester <strong>of</strong> 3-tert-buty1–4-hydroxy-5-methylphenylpropionic<br />

acid with ethylene glycol, Compound B, as a model<br />

compound (Table 23.1).<br />

Blood kinetics and blood metabolites<br />

Compound B is a symmetrical di-ester compound (Table 23.1). When<br />

administered at a single oral dose <strong>of</strong> about 10 mg kg −1 body weight to male<br />

rats, 14 C-phenyl-labelled Compound B was readily adsorbed, and maximal<br />

blood radioactivities were reached after 1 h. Thereafter, blood radioactivity<br />

declined rapidly and only minute amounts were detected 48 h after<br />

treatment. At any time point investigated Compound A, the free carboxylic<br />

acid <strong>of</strong> Compound B, was the dominating blood metabolite, whereas the<br />

parent compound constituted a minor component only (Table 23.2). These<br />

findings are indicative <strong>of</strong> an efficient first pass hydrolysis and suggest that<br />

the carboxylic acid metabolite, Compound A, might be responsible for the<br />

toxicological pr<strong>of</strong>ile <strong>of</strong> this antioxidant in the rat.<br />

Liver enzyme induction<br />

Compound B was administered in the feed to male rats at dose levels <strong>of</strong> 50,<br />

150, 500 and 1000 ppm. After treatment for 14 days, absolute liver<br />

weights were dose-dependently increased. Biochemically, this


Table 23.1 3.5-Substituted 4-hydroxyphenyl propionic acid esters<br />

H.THOMAS ET AL. 319<br />

hepatomegaly was accompanied by an induction <strong>of</strong> total microsomal<br />

cytochrome P450, microsomal epoxide hydrolase and UDPglucuronosyltransferase,<br />

cytosolic glutathione S-transferase and<br />

peroxisomal β-oxidation. The observed induction <strong>of</strong> the cytochrome P450<br />

content was reflected in increased activities <strong>of</strong> ethoxycoumarin O-deethylase<br />

as well as lauric acid 11- and 12-hydroxylases (Table 23.3). This<br />

suggests an induction <strong>of</strong> cytochrome P450 isoenzymes <strong>of</strong> the subfamilies<br />

CYP2B and CYP4A. Indeed, increased amounts <strong>of</strong> CYP2B and CYP4A<br />

proteins were found in liver microsomes from treated animals by means <strong>of</strong><br />

Western-blotting with monoclonal antibodies specific for these iso-enzymes<br />

(data not shown). Within 28 days after cessation <strong>of</strong> a 14-day treatment at


320 ANTIOXIDANTS AND LIGHT STABILISERS: TOXIC EFFECT<br />

Table 23.2 Parent equivalents and blood metabolites <strong>of</strong> [ 14 C]-labelled Compound B<br />

after single oral administration <strong>of</strong> 9.5 mg kg −1 body weight to male rats<br />

Note:<br />

bld: below the limit <strong>of</strong> detection.<br />

Blood was taken at the indicated time intervals and extracted with ethyl acetate for<br />

analysis. Compound B and its free carboxylic acid metabolite (Compound A,<br />

Table 23.1) were identified by thin-layer co-chromatography with authentic<br />

reference samples. Quantification <strong>of</strong> <strong>Compounds</strong> A and B was accomplished by<br />

radiometric scanning <strong>of</strong> the plates following thin-layer chromatography <strong>of</strong> the<br />

respective blood extracts.<br />

1000 ppm, liver weights as well as the investigated biochemical parameters<br />

returned to control levels. Therefore, Compound B may be addressed as a<br />

reversible barbiturate- and peroxisome proliferator-type inducer in the rat,<br />

as characterised by its liver enzyme induction pr<strong>of</strong>ile.<br />

Effects on serum thyrotropin and thyroid hormones<br />

When male rats were fed Compound B admixed in the diet for 14 days at<br />

dose levels <strong>of</strong> 50, 150, 500 and 1000 ppm, liver and thyroid weights were<br />

increased in a dose-dependent manner. Histopathological examination <strong>of</strong><br />

the thyroid gland revealed hypertrophy <strong>of</strong> the follicular epithelium and<br />

thinning <strong>of</strong> colloid at 150, 500 and 1000 ppm. Morphological changes in<br />

the pituitary gland comprised enlarged thyrotropin (TSH)-producing cells<br />

with foamy or vacuolated cytoplasm. In addition, treatment resulted in<br />

markedly increased serum TSH and reverse triiodothyronine (rT 3)<br />

concentrations, whereas serum thyroxine (T 4) and triiodothyronine (T 3)<br />

levels were found slightly decreased. The effects observed at 1000 ppm<br />

were found to be reversible after a 28-day recovery period (Muakkassah-<br />

Kelly et al., 1991).<br />

In additional experiments, male rats were rendered hypothyroid, fed<br />

Compound B at 1000 ppm for 21 days and infused for the last 7 days with<br />

slightly supraphysiological concentrations <strong>of</strong> T 4. The observed changes in


H.THOMAS ET AL. 321<br />

Table 23.3 The effect <strong>of</strong> Compound B on selected biochemical parameters in the<br />

male rat liver<br />

Notes:<br />

Values are means <strong>of</strong> 9 (14 days treatment) or 4 (14 days treatment/28-days<br />

recovery) animals.<br />

Standard deviations are given in parentheses.<br />

0/0: Control recovery. 1000/0: High-dose recovery.<br />

* p


322 ANTIOXIDANTS AND LIGHT STABILISERS: TOXIC EFFECT<br />

metabolism has been observed with various deiodinase inhibitors (Hill et<br />

al., Liang et al., 1993) and hepatic enzyme inducers (McClain, 1989;<br />

Curran and DeGroot, 1991; Barter and Klaassen, 1992; Visser et al.,<br />

1993).<br />

Induction <strong>of</strong> thyroid neoplasia<br />

In a long-term feeding study, administration <strong>of</strong> Compound B to rats at a<br />

dose level <strong>of</strong> 1000 ppm was associated with an increased incidence <strong>of</strong><br />

thyroid gland follicular adenoma and carcinoma (Muakkassah-Kelly et al.,<br />

1991). Compound B was shown to be devoid <strong>of</strong> mutagenic and clastogenic<br />

activity (Muakkassah-Kelly et al., 1991). Therefore, it is likely that thyroid<br />

tumour induction by Compound B was not the result <strong>of</strong> a direct, genotoxic<br />

effect on this organ, but rather a consequence <strong>of</strong> the hormonal imbalance<br />

induced by this antioxidant in the rat.<br />

An intact hypothalamic-pituitary-thyroid axis is able to respond to a<br />

chemically induced alteration in peripheral hormone metabolism with<br />

increased hormone production by the hypertrophic gland. However, it is<br />

known that chronic, excessive stimulation <strong>of</strong> the gland can lead to<br />

follicular hyperplasia and ultimately progress to thyroid neoplasia (Paynter<br />

et al., 1988; McClain, 1989; Curran and DeGroot, 1991; Johnson et al.,<br />

1993). For Compound B, this hypothesis is in agreement with the doseresponse<br />

characteristics obtained in the long-term study, where thyroid<br />

tumours were induced exclusively at a dose-level sufficiently high to cause<br />

hormonal imbalance (Muakkassah-Kelly et al., 1991).<br />

Implications for human risk assessment<br />

For human risk assessment, it is <strong>of</strong> critical importance to identify the<br />

mechanism by which Compound B caused thyroid neoplasia in the rat, a<br />

species most <strong>of</strong>ten used for carcinogenic hazard identification. Hyperplastic<br />

changes in the thyroid are frequently observed in rat carcinogenicity<br />

studies, and this species appears to be very sensitive to compounds which<br />

interfere with thyroid hormone synthesis and/or catabolism. They evoke an<br />

immediate stimulation <strong>of</strong> the gland upon short-term treatment as a<br />

consequence <strong>of</strong> an increased pituitary TSH secretion (Zbinden, 1987;<br />

Paynter et al., 1988; McClain, 1989).<br />

However, the effects <strong>of</strong> xenobiotics on the pituitary-thyroid axis in<br />

rodents cannot necessarily be extrapolated to man since rodents and man<br />

are distinguished by many important physiological and biochemical<br />

differences (Gopinath, 1991): e.g. the amount <strong>of</strong> thyroxin-binding<br />

globulin, the half-life <strong>of</strong> T 4 and its biliary excretion as well as plasma THSlevels<br />

and their response to thyrotropin releasing hormone. These<br />

differences render the rat very sensitive to small changes in the plasma T 4


level, whilst humans are essentially insensitive (Hill et al., 1989; Grasso et<br />

al., 1991).<br />

Therefore, in contrast to rats, there is no conclusive evidence for a critical<br />

role <strong>of</strong> TSH in thyroid stimulation and carcinogenesis in humans (Hill et<br />

al., 1989). In cultured human thyroid cells, for example, THS was unable<br />

to induce proliferation whereas a stimulation <strong>of</strong> growth was observed in<br />

rat thyroid cells (Westermark et al., 1985). Clinical data are available for<br />

some compounds such as the anticonvulsants phenobarbitone,<br />

diphenylhydantoin and carbamazepine as well as the antibiotic rifampicin,<br />

which are known liver microsomal enzyme inducers in man. They increase<br />

thyroid hormone metabolism and excretion and eventually decrease serum<br />

thyroid hormone levels. There is also evidence, that administration <strong>of</strong> these<br />

drugs leads to thyroid stimulation, however, largely in the absence <strong>of</strong><br />

increased TSH levels (Curran and DeGroot, 1991). In addition,<br />

epidemiological data are not in favour <strong>of</strong> a link between human use <strong>of</strong> such<br />

compounds with an increased incidence <strong>of</strong> thyroid tumours (Curran and<br />

DeGroot, 1991), nor have increased rates <strong>of</strong> thyroid cancers been reported<br />

in areas <strong>of</strong> endemic iodine deficiency (McClain, 1989). Therefore, the<br />

currently available data do not support the idea, that thyroid stimulation<br />

as a response to chemically induced increases in circulating TSH<br />

concentrations significantly contributes to thyroid tumour formation in<br />

man.<br />

Compound B has been shown to cause thyroid tumours in the rat. In a<br />

series <strong>of</strong> speciality studies, the compound was identified as an enzyme<br />

inducer and a 5′-deiodinase inhibitor in the rat liver. These findings argue<br />

for a rodentspecific, indirect mechanism leading to the formation <strong>of</strong> thyroid<br />

tumours. Moreover, the observed dose-response characteristics are<br />

indicative <strong>of</strong> a threshold process, e.g. liver enzyme induction and inhibition<br />

<strong>of</strong> 5′-deiodination are irrevocable prerequisities for thyroid tumour<br />

formation in this species.<br />

Benzotriazole-based light stabilisers<br />

H.THOMAS ET AL. 323<br />

Ester derivatives <strong>of</strong> the 3-[3-(2H-benzotriazole-2-yl)-5-tert-butyl-4hydroxy-phenyl]<br />

propionic acid represent potent UV-light absorbers and<br />

constitute an important class <strong>of</strong> industrial plastic additives and light<br />

stabilisers (<strong>Compounds</strong> C-F, Table 23.1). Toxicologically, this class <strong>of</strong><br />

chemicals is characterised by generally low acute oral or dermal toxicity<br />

and the lack <strong>of</strong> genotoxicity in the commonly employed battery <strong>of</strong><br />

bacterial and cellular mutagenicity tests. Irrespective <strong>of</strong> the alcohol moiety,<br />

however, all compounds, when administered subchronically to rats,<br />

displayed very similar predominantly hepatotrophic effects, with spleen and<br />

kidney weights in addition being only slightly affected: pronounced<br />

hepatomegaly, hepatocyte hypertrophy, and concomitantly increased


324 ANTIOXIDANTS AND LIGHT STABILISERS: TOXIC EFFECT<br />

plasma transaminase activities. Upon electron microscopical examination,<br />

a striking peroxisome proliferation was the major finding.<br />

Compound F, a di-ester ‘product by process’ obtained upon esterification<br />

<strong>of</strong> 3-[3-(2H-benzotriazole-2-yl)-5-tert-buty1–4-hydroxyphenyl] propionic<br />

acid with polyethyleneglycol 300, when tested for its toxicity to rat<br />

reproduction in a Segment I study, gave rise to increased numbers <strong>of</strong> stillborn<br />

pups, decreased pup survival, decreased weight gain <strong>of</strong> surviving pups, and<br />

dark discoloured abdominal skin regions in a number <strong>of</strong> pups at higher<br />

dose levels.<br />

The common nature <strong>of</strong> general toxicology findings with all investigated<br />

derivatives <strong>of</strong> the addressed benzotriazole-based light stabilisers suggested a<br />

common basis <strong>of</strong> action and, depending upon this action, perhaps a very<br />

similar behaviour and extent <strong>of</strong> potency as foetotoxic agents. In order to<br />

investigate these interrelationships a series <strong>of</strong> mechanistic studies were<br />

conducted focusing on the kinetics, primary metabolism <strong>of</strong> the parent<br />

compounds in vitro and in vivo and their effect on selected biochemical<br />

liver parameters in rats. Compound F was selected as a model compound<br />

to investigate the mechanism <strong>of</strong> toxicity in pregnant female rats and<br />

foetuses.<br />

In vitro hydrolysis<br />

Compound D, the methyl ester <strong>of</strong> 3-[3-(2H-benzotriazole-2-yl)-5-tertbutyl-4-hydroxyphenyl]<br />

propionic acid was readily hydrolysed in vitro by<br />

rat serum as well as rat liver homogenate while a homogenate <strong>of</strong> rat small<br />

intestine when compared on a gram tissue basis, appeared to be less<br />

efficient by three orders <strong>of</strong> magnitude than the liver. Increasing the sterical<br />

hindrance around the ester bond by formation <strong>of</strong> the di-ester with a short<br />

chain alcohol reduced the rate <strong>of</strong> in vitro hydrolysis considerably as<br />

demonstrated for Compound F, the diester <strong>of</strong> hexane-l,6-diol with<br />

Compound C. When <strong>of</strong>fered at a test concentration <strong>of</strong> 0.2 mM, essentially<br />

no hydrolysis <strong>of</strong> this compound was observed with rat serum, and the<br />

hydrolysis by liver and small intestine homogenate was estimated to<br />

proceed at least two and one orders <strong>of</strong> magnitude slower, respectively, than<br />

calculated for Compound D (Table 23.1 and 23.4).<br />

Blood kinetics and blood metabolites<br />

Assuming comparable extents <strong>of</strong> intestinal absorption in vivo, the observed<br />

differences in the in vitro hydrolysis rates might as well suggest<br />

significantly different in vivo hydrolysis rates and consequently quite<br />

different residence times for both parent compounds in the rat in vivo.<br />

Different residence times, on the other hand, may eventually allow not only<br />

for additional routes <strong>of</strong> metabolism but also for an intensification <strong>of</strong> toxic


Table 23.4 Kinetic parameters for the in vitro hydrolysis <strong>of</strong> Compound D and E by<br />

rat serum and organ homogenates<br />

Notes:<br />

a The apparent Vmax value for serum is given in µmol min −1 ml −1.<br />

H.THOMAS ET AL. 325<br />

b Initial velocity <strong>of</strong> hydrolysis at 0.2 mM ester concentration.<br />

Hydrolysis <strong>of</strong> Compound D was determined in 50 mM Tris/phosphate buffer, pH 7.<br />

5, containing either 1 per cent (v/v) rat serum, or 1.25 per cent (w/v) rat liver<br />

homogenate or 10 per cent (w/v) small intestine homogenate. Similarly, hydrolysis<br />

<strong>of</strong> Compound E was assessed using 98 per cent (v/v) rat serum or in the presence <strong>of</strong><br />

10 mM Tris/HCl buffer, pH 7.5, containing 250 mM sucrose and either 24.5 per<br />

cent (w/v) rat liver homogenate or 19.6 per cent (w/v) small intestine homogenate.<br />

effects. This question was addressed in a pharmacokinetic study under<br />

conditions <strong>of</strong> single oral administration <strong>of</strong> <strong>Compounds</strong> D and E at a dose<br />

level <strong>of</strong> 10 mg kg −1 each (Table 23.5).<br />

14 C-Phenyl-labelled Compound D was readily absorbed from the<br />

gastrointestinal tract. Maximal blood radioactivity was reached between 1<br />

and 2 h and subsequently eliminated with an apparent half-life <strong>of</strong> 10.0–11.<br />

8 h. After 48 h only minute amounts <strong>of</strong> radioactivity equalling about 3 per<br />

cent <strong>of</strong> the blood levels in T max were detectable. Analysis <strong>of</strong> the resulting<br />

blood metabolite pattern largely confirmed the findings <strong>of</strong> the preceding in<br />

vitro investigation <strong>of</strong> enzymatic Compound D hydrolysis: particularly<br />

during periods <strong>of</strong> high parent equivalent concentrations in blood as<br />

recorded between 30 min and 6 h after dosing, hydrolysis appeared to be<br />

the major metabolic pathway as evidenced by the high concentrations <strong>of</strong><br />

the carboxylic acid, Compound C (34–77 per cent), and an unidentified<br />

metabolite (17–36 per cent) which was regarded to have evolved from<br />

Compound C by an additional metabolic step (Table 23.5).<br />

Quite surprisingly, Compound E was found to be absorbed to a much<br />

lower extent than Compound D, with a C max after 1 h <strong>of</strong> less than one<br />

tenth <strong>of</strong> the value seen with the latter. Elimination with an apparent halflife<br />

<strong>of</strong> 12.0 h and slightly higher residual radioactivity <strong>of</strong> approximately 4.8<br />

per cent <strong>of</strong> the blood levels recorded at T max after 48 h indicated only a<br />

slightly reduced elimination rate as compared to Compound D. Also, quite<br />

different from the initially anticipated result, hydrolysis contributed<br />

substantially to the rapid metabolism <strong>of</strong> Compound E. The 24 h AUC<br />

values revealed a 23 per cent and 36 per cent contribution <strong>of</strong> the carboxylic


326 ANTIOXIDANTS AND LIGHT STABILISERS: TOXIC EFFECT<br />

Table 23.5 Summary <strong>of</strong> pharmacokinetic parameters obtained after single oral<br />

administration <strong>of</strong> 10 mg kg −1 Compound D and E to two male rats each<br />

Notes:<br />

a Below the limit <strong>of</strong> reliable quantification.<br />

pe: Parent equivalent.<br />

acid, Compound C, and unidentified metabolites, respectively, to the total<br />

AUC. It is assumed that the majority <strong>of</strong> unidentified metabolites have<br />

arisen from further biotransformation <strong>of</strong> the carboxylic acid. The<br />

significant contribution <strong>of</strong> hydrolysis products to the total AUC is still<br />

evident after 168 h although low blood radioactivity levels 24 h after<br />

dosing generally prevented accurate quantitation <strong>of</strong> the metabolites and<br />

thus slightly diminished their apparent overall share (Table 23.5).<br />

Liver enzyme induction<br />

Subchronic oral (gavage) administration <strong>of</strong> single daily doses <strong>of</strong> Compound<br />

C for 14 days, Compound D for 14 days, Compound E for 13 weeks, and<br />

Compound F for 114 days to male rats (Tables 23.6 and 23.7) was<br />

correlated with a dose-dependent massive increase in absolute liver weight<br />

up to about 190 per cent <strong>of</strong> control at the highest dose level irrespective <strong>of</strong><br />

the treatment period and the test compound. This pronounced<br />

hepatomegaly was paralleled by a comparably small two-fold elevation <strong>of</strong><br />

the microsomal cytochrome P450 contents and an about 50 per cent<br />

decrease in total UDP- glucuronosyltransferase activity. Essentially no<br />

changes were recorded for the cytochrome P450 dependent<br />

ethoxycoumarin O-de-ethylase activity while microsomal epoxide<br />

hydrolase activities appeared to vary, with slight increases in Compound C<br />

and Compound D treated animals, no changes in Compound E treated<br />

animals and even a dose-dependent reduction to 46 per cent <strong>of</strong> control in<br />

rats treated with Compound F at 100 mg kg −1 (Table 23.6).<br />

Strongly induced peroxisomal fatty acid β-oxidation activities for all<br />

model compounds as well as lauric acid 12-hydroxylase activities tested for<br />

<strong>Compounds</strong> E and F were accompanied by significant dose-dependent<br />

decreases in glutathione S-transferase activities to 32, 51, 40 and 15 per<br />

cent <strong>of</strong> control at the highest dose level tested for Compound C, D, E and


Table 23.6 The effect <strong>of</strong> various benzotriazole-based light stabilisers on selected biochemical parameters related to and indicative<br />

for a barbiturate and/or polycyclic aromatic hydrocarbon type enzyme induction in male rat liver<br />

H.THOMAS ET AL.<br />

327<br />

Notes:<br />

dnt: dose level not tested,<br />

nd: not determined.<br />

Microsomal epoxide hydrolase and total microsomal UDP-glucuronosyltransferase activities were determined with styrene oxide<br />

and 3-methyl-2-nitrophenol as substrate, respectively.<br />

Values are means±standard deviation <strong>of</strong> 10 control and 5 treated animals (a, b) or 8 animals (c) or 5 animals per group (d).<br />

Asterisks indicate results significantly different (two-sided Dunnett’s test) from control: * p


328 ANTIOXIDANTS AND LIGHT STABILISERS: TOXIC EFFECT<br />

F, respectively. For Compound F, which appeared to be the most potent<br />

inducer <strong>of</strong> peroxisomal β-oxidation and lauric acid 12-hydroxylase<br />

activities, a concomitant strong 90 per cent reduction <strong>of</strong> morphine UDPglucuronosyltrasferase<br />

and marked 2.5-fold increase in bilirubin UDPglucuronosyltransferase<br />

activity was recorded (Table 23.7). Electron<br />

microscopy confirmed what had already been indicated by changes in the<br />

investigated enzyme levels, a striking proliferation <strong>of</strong> peroxisomes with the<br />

same appearance <strong>of</strong> these organelles regardless <strong>of</strong> the compound tested:<br />

vigorous increase in number, a striking number <strong>of</strong> markedly enlarged<br />

peroxisomes frequently containing matrical inclusions (matrical plates) and<br />

peroxisomes forming arrays or clusters (polyperoxisomes) in the virtual<br />

absence <strong>of</strong> any significant proliferation <strong>of</strong> smooth endoplasmic reticulum<br />

(data not shown).<br />

Consequently, the hepatotrophic effects <strong>of</strong> the tested benzotriazole-based<br />

light stabilisers were clearly assigned to their action as peroxisome<br />

proliferators in rat liver. Mindful <strong>of</strong> the different durations <strong>of</strong> treatment<br />

their potency was found to rank in the order Compound E


Table 23.7 The effect <strong>of</strong> various benzotriazole-based light stabilizers on selected biochemical parameters related to and indicative<br />

for a peroxisome proliferator type enzyme induction in male rat liver<br />

H.THOMAS ET AL. 329<br />

Notes:<br />

dnt: dose level not tested.<br />

nd: not determined.<br />

Cyanide-insensitive peroxisomal fatty acid -oxidation and cytosolic glutathione S-transferase activities were determined with<br />

[l-14C]-palmitoyl-CoA and1-chloro2,4-dinitrobenzene as substrate, respectively.<br />

Values are means±standard deviation <strong>of</strong> 10 control and 5 treated animals (a, b) or 8 animals (c) or 5 animals per group (d).<br />

Asterisks indicate results significantly different (two-sided Dunnett’s test) from control: * p


330<br />

ANTIOXIDANTS AND LIGHT STABILISERS: TOXIC EFFECT<br />

Table 23.8 Morphological changes <strong>of</strong> dam and foetal hepatocyte organelles after treatment with Compound F from day 6 through<br />

days 14, 17 and 20 <strong>of</strong> gestation


iochemical investigations which revealed a moderate four-fold induction<br />

<strong>of</strong> peroxisomal β-oxidation in dams and an up to 15-fold induction <strong>of</strong> this<br />

activity at day 21 <strong>of</strong> gestation in foetal livers, respectively, with the final<br />

foetal activity exceeding dam activity by 40 per cent. Strong increases in<br />

lauric acid 11- and 12-hydroxylation, which are known to be associated<br />

with isoenzymes <strong>of</strong> the cytochrome P450 CYP4A gene family and the<br />

phenomenon <strong>of</strong> peroxisome proliferation in rodents, <strong>of</strong> up to 2.6- and 10.<br />

5-fold in dams and 11.5- and 23.2-fold in foetuses, respectively, at day 21<br />

<strong>of</strong> gestation were recorded as well, indicating again a slightly higher final<br />

activity in foetal than in dam liver. By contrast, catalase, known as a<br />

detoxifying enzyme for hydrogen peroxide generated particularly in the<br />

course <strong>of</strong> increased peroxisomal activity, was shown to be induced up to 5.<br />

6-fold in dam and 8.2-fold in foetal liver at day 21 <strong>of</strong> gestation, leaving<br />

foetal liver, however, with only about 30 per cent <strong>of</strong> the final activity seen<br />

in dams (Table 23.9). This discrepancy between the strong induction <strong>of</strong><br />

hydrogen peroxide generating peroxisomal β-oxidation and the<br />

considerably less potent induction <strong>of</strong> the hydrogen peroxide destroying<br />

catalase activity appears to be reflected in the 1.7-fold increase in the lipid<br />

peroxidation product malondialdehyde in foetal livers as well as in the<br />

decrease <strong>of</strong> total and reduced hepatic glutathione levels at day 21 <strong>of</strong><br />

gestation (Tables 23.9 and 23.10). Surprisingly, foetal defense systems<br />

against oxidative stress such as selenium-dependent and<br />

seleniumindependent glutathione peroxidase were found poorly developed<br />

throughout the investigated periods <strong>of</strong> gestation and barely inducible by<br />

the test article leaving the pups with little protection against any kind <strong>of</strong><br />

oxidative insult (Table 23.10). Consequently, Compound F was clearly<br />

identified as a peroxisome proliferator in pregnant rat as well as foetal liver<br />

with high potential for the initiation <strong>of</strong> oxidative damage in foetal tissues.<br />

Implications for human safety assessment<br />

H.THOMAS ET AL. 331<br />

The understanding <strong>of</strong> the mechanisms by which benzotriazole-based UV<br />

light stabilisers exert their hepatotrophic effects in rodents is <strong>of</strong> crucial<br />

importance for the assessment <strong>of</strong> safety aspects in humans. The presented<br />

rat studies have shown that the liver effects exerted by Compound C and<br />

its ester derivatives <strong>Compounds</strong> D, E and F are clearly related to the<br />

induction <strong>of</strong> peroxisome proliferation. The identical nature <strong>of</strong> the observed<br />

effects regardless <strong>of</strong> the alcohol component in the investigated esters<br />

suggests that the toxic potential resides solely with the 3-[3-(2Hbenzotriazole-2-yl)-5-tert-butyl-4-hydroxy-phenyl]<br />

propionic acid whereby<br />

the individual potency appears to be mainly determined by the different<br />

bioavailablity <strong>of</strong> the respective ester compound and the extent and velocity<br />

<strong>of</strong> its hydrolysis in vivo. Also, it appears, that in vitro hydrolysis studies do


332 ANTIOXIDANTS AND LIGHT STABILISERS: TOXIC EFFECT<br />

not actually reflect the hydrolytic capacity <strong>of</strong> the in vivo system for a given<br />

ester.<br />

Liver enlargement and the induction <strong>of</strong> diagnostic enzyme activities is a<br />

characteristic response <strong>of</strong> rodents to treatment with peroxisome<br />

proliferators and results from a combination <strong>of</strong> both hypertrophy and<br />

hyperplasia. According to current opinion, peroxisome proliferation and<br />

liver growth are closely associated with the formation <strong>of</strong> hepatocellular<br />

tumours in rats and mice (Hawkins et al., 1987; Lock et al., 1989; Bentley<br />

et al., 1993). However, a number <strong>of</strong> feeding studies have demonstrated<br />

that there may actually be two types <strong>of</strong> threshold with respect to dose<br />

relationships: at very low doses, administration <strong>of</strong> peroxisome proliferators<br />

will not result in any liver response at all (Bentley et al., 1993). With<br />

increasing doses the first threshold will be exceeded with subsequent<br />

stimulation <strong>of</strong> peroxisome proliferation and DNA synthesis.<br />

As a result <strong>of</strong> several studies it is obvious that a limited extent <strong>of</strong> liver<br />

growth does not automatically lead to tumour formation as has been<br />

demonstrated, for example, with fen<strong>of</strong>ibrate and diethylhexylphthalate in<br />

carcinogenicity assays (Mitchell et al., 1985; Price et al., 1986; Keith et al.,<br />

1991). Thus, a second threshold has to be exceeded at which the<br />

magnitude <strong>of</strong> effects is sufficient to cause tumour development in rodents.<br />

In addition, extended administration <strong>of</strong> the peroxisome proliferator<br />

appears a necessary prerequisite to exceed this tumourigenic threshold.<br />

Also, a large number <strong>of</strong> in vitro and in vivo studies have provided ample<br />

evidence for a marked species difference in susceptibility to the effects <strong>of</strong><br />

peroxisome proliferators. Rats and mice are extremely sensitive while<br />

hamsters show a markedly smaller response and non-human primates and<br />

humans appear to be insensitive or non-responsive (Lake et al., 1989;<br />

Bentley et al., 1993; Graham et al., 1994). The latter finding is supported<br />

by epidemiological evidence from long-term treatment <strong>of</strong> patients with<br />

hypolipidaemic agents (Bentley et al., 1993). Therefore the available<br />

evidence strongly supports the conclusion that the effects <strong>of</strong> benzotriazolebased<br />

light stabilisers in rodents are <strong>of</strong> no relevance to human safety<br />

assessment.<br />

The action <strong>of</strong> Compound F as a strong peroxisome proliferator in foetal<br />

livers starting as early as day 15 <strong>of</strong> gestation suggests that treatment related<br />

initiation <strong>of</strong> high level oxidative stress under conditions <strong>of</strong> poorly<br />

developed foetal protection and detoxification systems. This view is<br />

supported by substantially elevated hepatic malondialdehyde levels and<br />

essentially depleted glycogen stores in hepatocytes <strong>of</strong> foetuses from treated<br />

dams on day 21 <strong>of</strong> gestation. The latter indicates an extensive glucose<br />

consumption, presumably via the pentose phosphate pathway, to supply<br />

the hydrogen peroxide and lipid peroxide detoxifying glutathione<br />

peroxidase system with the necessary reduction equivalents (NADPH).<br />

Under conditions <strong>of</strong> limited degradation <strong>of</strong> hydrogen peroxide, this


Table 23.9 The effect <strong>of</strong> Compound F on dam and foetal absolute liver weight and selected biochemical liver parameters related to<br />

and indicative for peroxisome proliferation after treatment from day 6 through days 14, 17 and 20 <strong>of</strong> gestation<br />

H.THOMAS ET AL. 333<br />

Notes:<br />

bld: below the limit <strong>of</strong> detection.<br />

Cyanide-insensitive peroxisomal fatty acid -oxidation and catalase activities were determined with [lhydrogen<br />

peroxide as substrate, respectively.<br />

C]-palmitoyl-CoA and<br />

Values are means±standard deviation from 6 dams per group and 6 pools <strong>of</strong> 2–9 foetuses per dam depending on the litter size.<br />

Asterisks indicate results significantly different (two-sided Dunnett’s test) from control: * p


334<br />

ANTIOXIDANTS AND LIGHT STABILISERS: TOXIC EFFECT<br />

Table 23.10 The effect <strong>of</strong> Compound F on selected biochemical dam and foetal liver parameters related to defence mechanisms<br />

against oxidative stress after treatment from day 6 through days 14, 17 and 20 <strong>of</strong> gestation<br />

Notes:<br />

Selenium-dependent and selenium-independent glutathione peroxidase activities were determined with cumene hydroperoxide and<br />

hydrogen per-oxide as substrate, respectively.<br />

Values are means±standard deviation from 6 dams per group and 6 pools <strong>of</strong> 2–9 foetuses per dam depending on the litter size.<br />

Asterisks indicate results significantly different (two-sided Dunnett’s test) from control: * p


compound is known to pass the liver and to be systemically distributed<br />

throughout the foetal body. Thus the endothelial cell injuries observed in<br />

the course <strong>of</strong> the Segment II study are regarded secondary to the effect <strong>of</strong><br />

peroxisome proliferation and to have arisen as a consequence <strong>of</strong> oxidative<br />

damage which has been demonstrated to occur in various endothelial cell<br />

systems, for example as a result <strong>of</strong> iron-mediated oxygen free radical attack<br />

(Brieland et al., 1992; Barchowsky et al., 1994; Krautschick et al., 1995).<br />

Therefore, the observed foetotoxicity <strong>of</strong> Compound F in rats is regarded as<br />

occurring solely as a consequence <strong>of</strong> and secondary to peroxisome<br />

proliferation, and there is no evidence to assume that this effect as a result<br />

<strong>of</strong> exposure to benzotriazole based light stabilisers may occur in species<br />

that are non-responsive to the action <strong>of</strong> peroxisome proliferators as stated<br />

above, including humans.<br />

References<br />

H.THOMAS ET AL. 335<br />

ABRAHAM, R., BENITZ, K.F., PATIL, G. and LYON, R., 1986, Rapid induction<br />

<strong>of</strong> forestomach tumors in partially hepatectomized Wistar rats given butylated<br />

hydroxyanisole, Experimental and Molecular Pathology, 44, 14–20.<br />

ANON., 1986, Butylated hydroxyanisole (BHA), IARC Monographs on the<br />

Evaluation <strong>of</strong> the Carcinogenic Risk <strong>of</strong> Chemicals to Man, Vol. 40, pp. 123–<br />

59.<br />

BARCHOWSKY, A., WILLIAMS, M.E., BINZ, C.C. and CHEPENIK, K.P., 1994,<br />

Oxidant-sensitive protein phosphorylation in endothelial cells, Free Radical<br />

Biology and Medicine, 16, 771–7.<br />

BARTER, R.A. and KLAASSEN, C.D., 1992, Rat liver microsomal UDPglucuronosyltransferase<br />

activity toward thyroxin: characterisation, induction<br />

and form specificity, <strong>Toxicology</strong> and Applied Pharmacology, 115, 261–7.<br />

BENSON, A.M., CHA, Y.-H., BUEDING, E., HEINE, H.S. and TALALAY, P.,<br />

1979, Elevation <strong>of</strong> extrahepatic glutathione S-transferase and epoxide<br />

hydratase activities by 2(3)-tert-butyl-4-hydroxyanisole, Cancer Research, 39,<br />

2971–7.<br />

BENTLEY, P., CALDER, I., ELCOMBE, C., GRASSO, P., STRINGER, D. and<br />

WIEGAND, H.-J., 1993, Hepatic peroxisome proliferation in rodents and its<br />

significance for humans, Food and Chemical <strong>Toxicology</strong>, 31, 857–907.<br />

BRIELAND, J.K., CLARKE, S.J., KARMIOL, S., PHAN, S.H. and FANTONE, J.<br />

C., 1992, Transferrin: a potential source <strong>of</strong> iron for oxygen free radicalmediated<br />

endothelial cell injury, Archives <strong>of</strong> Biochemistry and Biophysics,<br />

294, 265–70.<br />

CHOE, S.Y., KIM, H.M. and YANG, K.H., 1984, Effects <strong>of</strong> butylated<br />

hydroxytoluene (BHT) on biliary excretion <strong>of</strong> xenobiotics and bile flow in<br />

rats, Drug and Chemical <strong>Toxicology</strong>, 7, 149–65.<br />

CONNING, D.M. and PHILLIPS, J.C., 1986, Comparative metabolism <strong>of</strong> BHA,<br />

BHT and other phenolic antioxidants and its toxicological relevance, Food and<br />

Chemical <strong>Toxicology</strong>, 24, 1145–8.


336 ANTIOXIDANTS AND LIGHT STABILISERS: TOXIC EFFECT<br />

CURRAN, P.G. and DEGROOT, L.J., 1991, The effect <strong>of</strong> hepatic enzyme-inducing<br />

drugs on thyroid hormones and the thyroid gland, Endocrine Reviews, 12,<br />

135–50.<br />

GOPINATH, C., 1991, Causes and interpretation <strong>of</strong> thyroid tumours in rodent<br />

bioassays, in Current Issues in Drug Development, Eynsham, Oxford: HRC<br />

Information Press Inc.<br />

GRAHAM, M.J., WILSON, S.A., WINHAM, M.A., SPENCER, A.J., REES, J.A.,<br />

OLD, S.L. and BONNER, F.W., 1994, Lack <strong>of</strong> peroxisome proliferation in<br />

marmoset liver following treatment with cipr<strong>of</strong>ibrate for 3 years, Fundamental<br />

and Applied <strong>Toxicology</strong>, 22, 58–64.<br />

GRASSO, P. et al., 1991, Role <strong>of</strong> persistent, non-genotoxic tissue damage in rodent<br />

cancer and relevance to humans, Annual Review <strong>of</strong> Pharmacology and<br />

<strong>Toxicology</strong>, 31, 253–87.<br />

GREGUS, Z. and KLASSEN, C.D., 1988, Effect <strong>of</strong> butylated hydroxyanisole on<br />

hepatic glucuronidation and biliary excretion <strong>of</strong> drugs in mice, Journal <strong>of</strong><br />

Pharmacy and Pharmacology, 40, 237–42.<br />

HAWKINS, J.M., JONES, W.E., BONNER, F.W. and GIBSON, G.G., 1987, The<br />

effect <strong>of</strong> peroxisome proliferators on microsomal, peroxisomal, and<br />

mitochondrial enzyme activities in the liver and kidney, Drug Metabolism<br />

Reviews, 18, 441–515.<br />

HILL, R.N., ERDREICH, L.S., PAYNTER, O.E., ROBERTS, P.A., ROSENTHAL,<br />

S.L. and WILKINSON, C.F., 1989, Thyroid follicular cell carcinogenesis,<br />

Fundamental and Applied <strong>Toxicology</strong>, 12, 629–97.<br />

ITO, N., FUKUSHIMA, S. and TSUDA, H., 1986a, Carcinogenicity and<br />

modification <strong>of</strong> the carcinogenic response by BHA, BHT, and other<br />

antioxidants, CRC Critical Reviews in <strong>Toxicology</strong>, 15, 109–50.<br />

ITO, N., FUKUSHIMA, S., TAMANO, S., HIROSE, M. and HAGIWARA, A.,<br />

1986b, Dose response in butylated hydroxyanisole induction <strong>of</strong> forestomach<br />

carcinogenesis in F344 rats, Journal <strong>of</strong> the National Cancer Institute, 77, 1261–<br />

5.<br />

JOHNSON, S., McKILLOP, D., MILLER, J. and SMITH, I.K., 1993, The effects <strong>of</strong><br />

rat thyroid function <strong>of</strong> an hepatic microsomal enzyme inducer, Human &<br />

Experimental <strong>Toxicology</strong>, 12, 153–8.<br />

KEITH, Y., CANNING, P.M., FOSTER, J., LHUGUENOT, J.C. and ELCOMBE,<br />

C. R., 1991, Peroxisome proliferation due to di-(2-ethylhexyl) adipate, 2-ethyl<br />

hexanol and 2-ethyl hexanoic acid, Archives <strong>of</strong> <strong>Toxicology</strong>, 66, 321–6.<br />

KRAUTSCHICK, I., KRUGMANN, J. and NEUENFELD, M., 1995, The effect <strong>of</strong><br />

per-oxides on the vascular endothelium <strong>of</strong> isolated pig aorta in vitro,<br />

Experimental and Toxicologic Pathology, 47, 51–61.<br />

LAKE, B.G., EVANS, J.G., GRAY, T.J.B., KÖRÖSI, S.A. and NORTH, C.J., 1989,<br />

Comparative studies on nafenopin-induced hepatic peroxisome proliferation in<br />

the rat, Syrian hamster, guinea pig and marmoset, <strong>Toxicology</strong> and Applied<br />

Pharmacology, 99, 148–60.<br />

LIANG, H., MORIN, O. and BURGER, A.G., 1993, Effect <strong>of</strong> the antioxidant TK<br />

12627 (Irganox) on monodeiodination and on the levels <strong>of</strong> messenger<br />

ribonucleic acid <strong>of</strong> 5′-deiodinase type I and spot 14, Acta Endocrinologica,<br />

128, 451–8.


H.THOMAS ET AL. 337<br />

LOCK, E.A., MITCHELL, A.M. and ELCOMBE, C.R., 1989, Biochemical<br />

mechanisms <strong>of</strong> induction <strong>of</strong> hepatic peroxisome proliferation, Annual Review<br />

<strong>of</strong> Pharmacology and <strong>Toxicology</strong>, 29, 145–63.<br />

McCLAIN, R.M., 1989, The significance <strong>of</strong> hepatic microsomal enzyme induction<br />

and altered thyroid function in rats: implications for thyroid gland neoplasia.<br />

Toxicologic Pathology, 17, 294–306.<br />

MITCHELL, A.M., LHUGUENOT, J.C., BRIDGES, J.W. and ELCOMBE, C.R.,<br />

1985, Identification <strong>of</strong> the proximate peroxisome proliferator(s) derived from<br />

di(2-ethylhexyl) phthalate, <strong>Toxicology</strong> and Applied Pharmacology, 80, 23–32.<br />

MUAKKASSAH-KELLY, S.F., KRINKE, A.-L., MALINOWSKI, W., STÄUBLI,<br />

W., BENTLEY, P., WAECHTER, F., JUGE-AUBRY, C. and BURGER, A.G.,<br />

1991, The effect <strong>of</strong> short term feeding <strong>of</strong> the antioxidant triethyleneglycol-bis-3<br />

(3-tert-butyl-4-hydroxy-5-methyl)propionate on serum thyrotropin and thyroid<br />

hormones in the male rat, <strong>Toxicology</strong> and Applied Pharmacology, 107, 129–<br />

40.<br />

PAYNTER, O.E., BURIN, G.J., JAEGER, R.B. and GREGORIO, C.A., 1988,<br />

Goitrogens and thyroid follicular cell neoplasia: evidence for a threshold<br />

process. Regulatory <strong>Toxicology</strong> and Pharmacology, 8, 102–19.<br />

PERCHELLET, J.-P. and PERCHELLET, E.M., 1989, Antioxidants and<br />

multistage carcinogenesis in mouse skin, Free Radical Biology and Medicine,<br />

1, 377–408.<br />

PRICE, S.C., HINTON, R.H., MITCHELL, F.E., HALL, D.E., GRASSO, P.,<br />

BLANAERD, G.F. and BRIDGES, J.W., 1986, Time and dose study on the<br />

response <strong>of</strong> rats to the hypolipidaemic drug fen<strong>of</strong>ibrate, <strong>Toxicology</strong>, 41, 169–<br />

91.<br />

PROCHASKA, H.J. and TALALAY, P., 1988, Regulatory mechanisms <strong>of</strong><br />

mon<strong>of</strong>unctional and bifunctional anticarcinogenic enzyme inducers in murine<br />

liver, Cancer Research, 48, 4776–82.<br />

RODRIGUES, A.D., FERNANDEZ, D., NOSARZEWSKI, M.A., PIERCE, W.M.<br />

JR. and PROUGH, R.A., 1991, Inhibition <strong>of</strong> hepatic microsomal cytochrome<br />

P-450 dependent monooxygenation activity by the antioxidant 3-tert-butyl-4hydroxyanisole,<br />

Chemical Research in <strong>Toxicology</strong>, 4, 281–9.<br />

SØNDERGAARD, D. and OLSEN, P., 1982, The effect <strong>of</strong> butylated<br />

hydroxytoluene (BHT) on the rat thyroid, <strong>Toxicology</strong> Letters, 10, 239–44.<br />

THOMAS, G.A. and WILLIAMS, E.D., 1991, Evidence for and possiblemechanisms<br />

<strong>of</strong> non-genotoxic carcinogenesis in the rodent thyroid, Mutation Research, 248,<br />

357–70.<br />

VERHAGEN, H., FURNÉE, C., SCHUTTE, B., HERMANS, R.J.J., BOSMAN, F.T.,<br />

BLIJHAM, G.H., TEN HOOR, F., HENDERSON, P.T. and KLEINJANS,<br />

J.C. S., 1989, Butylated hydroxyanisole-induced alterations in cell kinetic<br />

parameters in rat forestomach in relation to its oxidative cytochrome P-450mediated<br />

metabolism, Carcinogenesis, 10, 1947–51.<br />

VERHAGEN, H., SCHILDERMAN, P.A.E.L. and KLEINJANS, J.C.S., 1991,<br />

Butylated hydroxyanisole in perspective, Chemico-Biological Interactions, 80,<br />

109– 34.<br />

VISSER, T.J., KAPTEIN, E., VAN TOOR H., VAN RAAIJ, J.A.G.M., VAN DEN<br />

BERG, K.J., TJONG TJIN JOE, C., VAN ENGELEN, J.G.M. and BROUWER<br />

A., 1993, Glucuronidation <strong>of</strong> thyroid hormone in rat liver: effects <strong>of</strong> in vivo


338 ANTIOXIDANTS AND LIGHT STABILISERS: TOXIC EFFECT<br />

treatment with microsomal enzyme inducers and in vitro assay conditions,<br />

Endocrinology, 133, 2177–86.<br />

WESTERMARK, K., KARLSSON, F.A. and WESTERMARK, B., 1985,<br />

Thyrotropin modulates EGF receptor function in porcine thyroid follicle cells,<br />

Molecular and Cellular Endocrinology, 40, 17–23.<br />

WILLIAMS, G.M., McQUEEN, C.A. and TONG, C., 1990a, Toxicity studies <strong>of</strong><br />

butylated hydroxyanisole and butylated hydroxytoluene. I. Genetic and<br />

cellular effects, Food and Chemical <strong>Toxicology</strong>, 28, 793–8.<br />

WILLIAMS, G.M., WANG, C.X. and IATROPOULOS, M.J., 1990b, Toxicity<br />

studies <strong>of</strong> butylated hydroxyanisole and butylated hydroxytoluene, II. Chronic<br />

feeding studies. Food and Chemical <strong>Toxicology</strong>, 28, 799–806.<br />

ZBINDEN, G., 1987, Assessment <strong>of</strong> hyperplastic and neoplastic lesions <strong>of</strong> the<br />

thyroid gland, Trends in Pharmacological Sciences, 8, 11–14.


24<br />

<strong>Toxicology</strong> <strong>of</strong> Surfactants: Molecular,<br />

Mechanistic and Regulatory Aspects<br />

WALTER STERZEL<br />

Henkel KGaA, Düsseldorf<br />

Introduction<br />

The vast distribution <strong>of</strong> surfactants in various products in everyday use<br />

requires that the unwanted effects as well as the desired properties are<br />

known in order to recognize possible risks and prevent any damage to the<br />

health <strong>of</strong> humans. In order to understand the effects <strong>of</strong> surfactants on the<br />

organism, their most important biochemical effects, which depend on the<br />

interaction <strong>of</strong> surface active agents with basic biological structures like<br />

membranes, proteins and enzymes, are discussed. Following this discussion<br />

the local effects <strong>of</strong> surfactants are described. Local in this sense are all the<br />

effects encountered directly at the point <strong>of</strong> contact with the outer surfaces<br />

<strong>of</strong> the body, such as skin and mucous membrane irritation as well as<br />

allergies arising from skin contact. After this section the toxicokinetic<br />

properties <strong>of</strong> surfactants, providing information about type and extent <strong>of</strong><br />

absorption by organisms, metabolic pathways and their elimination are<br />

discussed. The section on systemic effects deals, in contrast to local effects,<br />

with reactions arising after the substance has entered the organism by<br />

swallowing, skin penetration or inhalation.<br />

Due to their technical and economic importance, surfactants have been<br />

used extensively for decades. This resulted in an abundance <strong>of</strong> scientific<br />

publications concerning their effects on organisms. As a complete review<br />

would exceed the scope <strong>of</strong> this contribution, the focus will centre on a<br />

description <strong>of</strong> exemplary data which are important for the evaluation <strong>of</strong><br />

the safety <strong>of</strong> surfactants.<br />

Biochemical properties <strong>of</strong> surfactants<br />

Surfactants come into immediate contact with the body during cleaning <strong>of</strong><br />

the skin and act on the skin cells directly. When surfactants are swallowed<br />

unin tentionally, tissue damage is also possible. The question <strong>of</strong> the effects<br />

on the cells and cell components like membranes, proteins and enzymes is<br />

therefore also important from a toxicological point <strong>of</strong> view.


340 TOXICOLOGY OF SURFACTANTS<br />

Interactions with membranes<br />

Due to their ability to absorb at interfaces, surfactants can interact with<br />

biological membranes. This interaction depends on the concentration <strong>of</strong> the<br />

surfactant and can be described in the following sequence (Helenius and<br />

Simons, 1975). In the first instance the monomeric surfactant molecule<br />

adsorbs onto the membrane. For a low surfactant/membrane ratio this<br />

changes the permeability <strong>of</strong> the membrane and leads to cell lysis at higher<br />

concentrations. At even higher surfactant concentrations, the lamellar<br />

structure <strong>of</strong> the membrane is lost and it is solubilized. A further increase in<br />

surfactant concentration results in the separation <strong>of</strong> the phospholipids from<br />

the protein. This allows surfactant molecules to adsorb on previously<br />

hidden regions <strong>of</strong> the protein molecule. For the solubilization <strong>of</strong> integral<br />

membrane proteins the formation <strong>of</strong> micelle/protein complexes seems to be<br />

a prerequisite. A significant solubilization <strong>of</strong> these proteins is possible only<br />

if the critical micelle concentration c M is exceeded. This is indicated by the<br />

fact that the microsomal membrane bound enzyme arylsulphatase-C could<br />

only be extracted from the membrane with retention <strong>of</strong> the biological<br />

activity after micelles were formed (Chang et al., 1985).<br />

As a consequence <strong>of</strong> these interactions, surfactants are able to influence<br />

the metabolism <strong>of</strong> membrane components (DeLeo, 1989). This has been<br />

demonstrated by studies on the pathophysiology <strong>of</strong> surfactant-mediated<br />

skin irritation. In vitro cultured corneocytes showed an increased release <strong>of</strong><br />

cholin metabolites after incubation with anionic surfactants. This effect<br />

was less pronounced after treatment with nonionic surfactants. In<br />

conclusion, these investigations demonstrated that the release <strong>of</strong><br />

metabolites is correlated with the irritation potential <strong>of</strong> surfactants.<br />

Interactions with proteins<br />

Depending on the structure <strong>of</strong> the surfactant the interactions with proteins<br />

are based on polar or hydrophobic interactions. The binding <strong>of</strong> surfactants<br />

to protein molecules is a function <strong>of</strong> the concentration <strong>of</strong> free surfactant in<br />

equilibrium with the protein. The binding is affected by the pH,<br />

temperature and ionic strength <strong>of</strong> the solution. These factors can lead to<br />

conformational changes <strong>of</strong> proteins and thereby increase or decrease the<br />

number <strong>of</strong> available binding sites. Natural bovine albumin, for example,<br />

has 10 binding sites for decyl glucoside at 10°C and 13 at 25°C<br />

(Wasylewski and Kozik, 1979). According to a theory developed by Jones<br />

(1975), surfactants adsorb onto proteins in multiple equilibria. Only a few<br />

surfactant molecules (


W.STERZEL 341<br />

pyruvate oxidase can form this type <strong>of</strong> binding (Schwuger and Bartnik,<br />

1980). Binding <strong>of</strong> more surfactant molecules leads to conformational<br />

changes in the protein. It is obvious that conformational changes allow<br />

binding <strong>of</strong> further surfactant molecules on hydrophobic regions which were<br />

previously not exposed.<br />

According to their different chemical structure (e.g. anionic, cationic,<br />

amphoteric or nonionic) surfactants differ significantly in their ability to<br />

carry out cooperative binding and therefore they differ in their biological<br />

activity. Anionic surfactants form adsorption complexes with proteins due<br />

to polar and hydrophobic interactions. Polar interactions between the<br />

negatively charged hydrophilic group <strong>of</strong> the surfactant and the positively<br />

charged groups <strong>of</strong> the protein molecule are the precondition for the<br />

formation <strong>of</strong> hydrophobic associations between surfactant molecule and<br />

protein molecule (Garcia-Dominguez, 1977; Schwuger and Bartnik, 1980).<br />

In the case <strong>of</strong> dodecylsulphate and tetradecylsulphate the binding results in<br />

denaturation <strong>of</strong> the proteins (Makino et al., 1973). Cationic surfactants can<br />

interact by polar and hydrophobic binding as well. Polar interactions result<br />

in electrostatic bonds between the negatively charged groups <strong>of</strong> the protein<br />

molecule and the positively charged surfactant molecule. For example, the<br />

enzyme, glucose oxidase, is deactivated by hexadecyl trimethyl ammonium<br />

bromide through formation <strong>of</strong> an ion pair between the cationic surfactant<br />

and the anionic amino acid side chain <strong>of</strong> the enzyme molecule (Tsuge,<br />

1984). Nonionic or amphoteric surfactants and proteins show either no<br />

interaction at all or interactions that are extremely weak and normally<br />

close to the limits <strong>of</strong> sensitivity <strong>of</strong> the analytical methods used. For this<br />

reason, nonionic surfactants will not dissolve sparingly soluble proteins,<br />

denature proteins, or contribute to a swelling <strong>of</strong> the epidermis. Figure 24.1<br />

shows the solubility <strong>of</strong> the protein zein, which is almost insoluble in water,<br />

and is more or less solubilized by sodium dodecyl sulphate and alkyl<br />

ethyleneglycol ether sulphates, while the nonionic ethoxylated nonylphenol<br />

is ineffective (Schwuger and Bartnik, 1980). A further reason for the poor<br />

interactions between nonionic surfactants and proteins could be that the<br />

concentration necessary for cooperative binding with the protein is not<br />

attained with nonionic surfactants due to their low critical micelle<br />

concentration c M (Makino et al., 1973).<br />

An important consequence <strong>of</strong> the interactions between anionic<br />

surfactants and proteins is the swelling <strong>of</strong> the stratum corneum <strong>of</strong> the skin.<br />

Hydrophobic interactions between surfactant chains and the protein result<br />

in pendant ionic head groups and subsequently in swelling because <strong>of</strong><br />

electrostatic repulsion between them. As the substrate matrix expands and<br />

the tertiary structure is disrupted, hydration occurs which leads to swelling<br />

(Blake-Haskins, 1986).


342 TOXICOLOGY OF SURFACTANTS<br />

Figure 24.1 Zein solubility c z <strong>of</strong> saturated solutions as a function <strong>of</strong> surfactant<br />

concentration. Zein concentration, 50 g lit −1 , mixing period, 2 h, temperature, 40°C<br />

( , sodium dodecyl sulphate; , alkyl ether sulphate (2EO); , nonyl phenol<br />

ethoxylate (9EO)).<br />

Interactions with enzymes<br />

Surfactants which are capable <strong>of</strong> massive cooperative binding, such as<br />

many anionic and cationic surfactants, induce conformational changes in<br />

the protein molecule which in general lead to loss <strong>of</strong> biological activity.<br />

The following mechanisms <strong>of</strong> enzyme inactivation by surfactants have to<br />

be considered (Ne’eman et al., 1971):<br />

1. Disruption <strong>of</strong> the quaternary structure <strong>of</strong> the enzyme when the enzyme<br />

protein consists <strong>of</strong> several subunits.


2. Induction <strong>of</strong> conformational changes in the tertiary or secondary<br />

structure <strong>of</strong> the enzyme protein.<br />

3. In the case <strong>of</strong> membrane-bound enzymes, separation <strong>of</strong> the enzyme<br />

protein from essential membrane lipids.<br />

4. Binding at active sites <strong>of</strong> the enzyme.<br />

While the effect <strong>of</strong> cationic surfactants on membranes is comparable to<br />

that <strong>of</strong> anionic surfactants, many proteins are obviously more resistant<br />

towards the denaturing activity <strong>of</strong> .cationic surfactants (Nozaki et al.,<br />

1974). Binding <strong>of</strong> tetradecyl trimethyl ammonium chloride onto bovine<br />

serum albumin and other proteins is comparable to that <strong>of</strong> sodium dodecyl<br />

sulphate. However, the cooperative binding with subsequent denaturation<br />

requires a ten-fold higher concentration <strong>of</strong> cationic surfactant. The<br />

saturation <strong>of</strong> the surfactant/protein complex is prevented by the competing<br />

formation <strong>of</strong> surfactant micelles. Contrary to the irreversibly denaturing<br />

effect <strong>of</strong> sodium dodecyl sulphate, the effect <strong>of</strong> some cationic surfactants on<br />

proteins is reversible (Nakaya et al., 1971).<br />

Local effects<br />

Skin compatibility<br />

W.STERZEL 343<br />

The damaging effects <strong>of</strong> surfactants on skin manifest themselves in dryness,<br />

roughness and scaling. In addition, symptoms <strong>of</strong> inflammation (reddening,<br />

swelling) can develop, which can result, in severe cases, in complete<br />

destruction <strong>of</strong> the tissue. All these symptoms are a result <strong>of</strong> the described<br />

biochemical properties <strong>of</strong> surfactants. The skin is defatted by the more or<br />

less pronounced property <strong>of</strong> the surfactants to emulsify lipids and thus<br />

partially or completely removing the surface film <strong>of</strong> lipids. This leads to a<br />

disturbance <strong>of</strong> the barrier function <strong>of</strong> the skin resulting in increased<br />

permeability for chemical substances and a loss <strong>of</strong> water. Anionic<br />

surfactants can cause swelling <strong>of</strong> the skin. As a result, they facilitate the<br />

transport <strong>of</strong> substances to lower layers where inflammation reactions can be<br />

induced (Scholz, 1967). The reaction <strong>of</strong> surfactants with proteins dissolves<br />

proteins out <strong>of</strong> the skin and leads to their denaturation. These changes in<br />

the matrix material have an effect on the resistance <strong>of</strong> the skin (Götte,<br />

1967) and, along with degreasing and drying, are an additional cause <strong>of</strong> an<br />

increase in skin roughness (Imokawa, 1975).<br />

The majority <strong>of</strong> the knowledge about skin compatibility <strong>of</strong> surfactants<br />

originates from studies with experimental animals, preferably rabbits.<br />

Furthermore, it is possible to evaluate new substances directly on human<br />

skin after careful exclusion <strong>of</strong> unreasonable risks. A critical overview <strong>of</strong><br />

different test methods is given by Kästner (1980). In this context, the


344 TOXICOLOGY OF SURFACTANTS<br />

problem <strong>of</strong> labelling chemical products with respect to their toxicological<br />

properties has to be addressed. Different national or international<br />

regulations, e.g. the EG directive 67/548 within the European Community,<br />

dictate that these products are labelled ‘irritating’ or ‘corrosive’ whenever<br />

exactly defined effects are observed in appropriate tests. With consumer<br />

protection in mind, exceedingly stringent test procedures have been<br />

established. These conditions frequently result in an unfavourable<br />

classification especially for surfactants. When interpreting data from such<br />

studies, it is important to consider that unrealistic conditions <strong>of</strong> exposure<br />

were involved.<br />

Since anionic surfactants are the class with the greatest economic<br />

importance, they are the best studied. No general statement is possible with<br />

regard to a classification <strong>of</strong> the various groups <strong>of</strong> anionic surfactants in<br />

order <strong>of</strong> their skin compatibility, since within each class <strong>of</strong> substances<br />

significant differences exist in their effect on skin depending on the<br />

respective structure. Opdyke et al. (1965), for example, found a decrease in<br />

the skin irritation potential <strong>of</strong> different alkyl ether sulphates with<br />

increasing level <strong>of</strong> ethoxylation. The effect <strong>of</strong> the alkyl chain length <strong>of</strong><br />

anionic surfactants was examined in different test models for soaps, alkyl<br />

sulphates, alkyl sulphonates, alkylbenzene sulphonates as well as alphaolefin<br />

sulphonates (Kästner, 1980). As shown in Table 24.1, it could be<br />

established in all cases that compounds with a saturated side chain <strong>of</strong> 10–<br />

12 C atoms exert the largest effect, or rather, have the highest potential for<br />

damage. When the results <strong>of</strong> skin compatibility tests for the most<br />

important classes <strong>of</strong> anionic surfactants are summarized, it becomes<br />

evident that the undiluted products have to be regarded as strongly<br />

irritating substances. Even at concentrations <strong>of</strong> 10 per cent moderate to<br />

strong effects have to be expected. However, at concentrations less than 1<br />

per cent, which is the range corresponding to typical use levels in<br />

detergents, only minimal irritation is observed.<br />

Nonionic surfactants have a good skin compatibility at normal use<br />

levels. Although studies with alcohol ethoxylates were reported in which a<br />

strong irritant effect was observed (Grupp et al., 1960), these studies used<br />

concentrations far above the usual exposure levels <strong>of</strong> consumers.<br />

Independent <strong>of</strong> their structure, cationic surfactants cause severe skin<br />

damage in high concentrations, while typical application levels are<br />

generally tolerated well.<br />

Mucous membrane compatibility<br />

When talking about mucous membrane compatibility one has to consider<br />

not only the mucous membranes <strong>of</strong> the eye. In addition, the mucous<br />

membranes in the mouth, upper and lower gastrointestinal tract as well as<br />

the urogenital tract have to be considered. In general, the effects <strong>of</strong>


Table 24.1 Structure/activity relationships <strong>of</strong> anionic surfactants<br />

W.STERZEL 345<br />

a Test model: A=epicutaneous, mouse; B=intracutaneous, mouse; C=epicutaneous,<br />

man; D=roughness <strong>of</strong> skin; E=swelling <strong>of</strong> collagen in vitro; F=denaturation <strong>of</strong><br />

protein in vitro.<br />

b Number <strong>of</strong> carbon atoms in the alkyl chain.<br />

surfactants on mucous membranes are based on the same biochemical<br />

mechanisms that are described in the chapter on skin compatibility. Special<br />

characteristics in the fine structure <strong>of</strong> mucous membranes, like the absence<br />

<strong>of</strong> keratin, result in a significantly higher sensitivity <strong>of</strong> these tissues towards<br />

chemical substances. Irritating materials affecting the eye cause reddening<br />

through increased blood flow in the conjunctivae with enlargement <strong>of</strong> the<br />

blood vessels. This can finally lead to the destruction <strong>of</strong> the cell walls<br />

accompanied by bleeding. Depending on the severity <strong>of</strong> the effects, a more<br />

or less pronounced swelling or reflex-induced closure <strong>of</strong> the eyelid will<br />

occur, followed by tearing and secretion. If the degree <strong>of</strong> irritation is low,<br />

epithelium damage develops on the cornea which can be visualized only<br />

with special techniques (staining, slit lamp microscope) and which is<br />

generally reversible. In severe cases the effects result in irreversible clouding<br />

<strong>of</strong> the cornea and therefore lead to an impairment <strong>of</strong> the eyesight.<br />

The classical method for the evaluation <strong>of</strong> mucous membrane<br />

compatibility <strong>of</strong> chemicals is the so-called Draize test on the rabbit eye<br />

(Draize et al., 1944). A structure/activity relationship with respect to the<br />

length <strong>of</strong> the respective alkyl chains <strong>of</strong> anionic surfactants can, as for the<br />

skin compatibility, also be observed for the mucous membrane<br />

compatibility (Kästner, 1980). According to this, the maximum irritation<br />

occurs at chain lengths <strong>of</strong> C 10−14. for n-alkyl sulphates as well as for nalkyl<br />

sulphonates. Although the irritation potential <strong>of</strong> the different<br />

surfactant classes extends over a large range, it can be concluded that the<br />

mucous membrane compatibility decreases in the following order:


346 TOXICOLOGY OF SURFACTANTS<br />

nonionic>anionic>cationic surfactants (Draize and Kelley, 1952; Hazleton,<br />

1952; Grant, 1962).<br />

Sensitization<br />

Aside from acute irritation, chemical substances can cause allergies after<br />

contact with the skin or a mucous membrane. The development <strong>of</strong> an<br />

allergy is dependent on certain preconditions. An essential factor is the<br />

individual disposition which is predominantly genetically determined. An<br />

additional important point is the extent <strong>of</strong> damage to the tissue at the point<br />

<strong>of</strong> contact <strong>of</strong> the chemical substance (inflammation), which promotes<br />

sensitization. In addition, the sensitization potential <strong>of</strong> a substance is <strong>of</strong><br />

decisive importance. For products with low molecular weights, this<br />

potential is dependent on their chemical properties. Small molecules are by<br />

themselves not able to trigger a reaction <strong>of</strong> the immune system. They<br />

become immunologically active only after binding to endogeneous<br />

proteins. Since the majority <strong>of</strong> the surfactants can only form weak and<br />

reversible bindings via hydrophobic and electrostatic interactions, this<br />

prerequisite is not fulfilled.<br />

Once the organism is sensitized towards a certain chemical, renewed<br />

contact with trace amounts <strong>of</strong> this material can provoke allergic reactions,<br />

which especially affect the skin and respiratory tract. Typical symptoms are<br />

itching, eczema, exanthema, rhinitis and bronchial asthma.<br />

Anionic surfactants and surfactant containing products were tested for<br />

sensitizing properties by numerous laboratories (Götte, 1967; Kästner,<br />

1980; Siwak et al., 1982) without detecting any significant increase in risk.<br />

The same holds true for nonionic surfactants (Siwak et al., 1982). Some<br />

cationic surfactants, which are able to form stable complexes by the<br />

formation <strong>of</strong> ion pairs with anionic groups <strong>of</strong> proteins, proved to be<br />

allergenic (Schallreuter and Wood, 1986).<br />

Toxicokinetics<br />

Percutaneous absorption<br />

The most important exposure <strong>of</strong> humans occurs through the skin with the<br />

use <strong>of</strong> cosmetics and toiletries. The skin comes in contact with surfactants<br />

also during dishwashing or when washing hands. Since these products are<br />

used over a long period <strong>of</strong> time, possible long-term effects must be<br />

evaluated. Measurement <strong>of</strong> percutaneous absorption <strong>of</strong> surfactants is<br />

important because it provides data for the toxicologist concerning the<br />

amount <strong>of</strong> surfactants which could enter the body through the skin in the<br />

most unfavourable case. Together with other toxicological information,


this allows a realistic evaluation <strong>of</strong> the risk when these compounds are<br />

used.<br />

Due to their economic importance, most studies have been carried out<br />

with anionic surfactants. Fewer studies exist for the other classes <strong>of</strong><br />

surfactants. In vitro measurements <strong>of</strong> the percutaneous absorption <strong>of</strong><br />

sodium dodecyl sulphate indicated a low absorption value for rat skin as<br />

well as for human skin (Blank and Gould, 1961; Embery and Dugard,<br />

1969; Howes, 1975). The low cutaneous absorption <strong>of</strong> sodium dodecyl<br />

sulphate could also be confirmed in experiments with rats (Greb, 1980).<br />

After application <strong>of</strong> a 0.7 per cent aqueous solution <strong>of</strong> sodium dodecyl<br />

sulphate (contact time 15 min), a cutaneous absorption <strong>of</strong> 0.26 µg cm −2<br />

within 24 h was measured (Howes,1975).<br />

In summarizing the results <strong>of</strong> the available studies, one can conclude that<br />

only small amounts <strong>of</strong> surfactants are resorbed through the intact skin.<br />

Since human skin in general is less permeable to chemicals (Rice, 1977;<br />

Wester and Maibach, 1982), the amounts <strong>of</strong> surfactants absorbed<br />

cutaneously in everyday use are probably even smaller. If the epidermis is<br />

removed completely or partially, e.g. damaged skin, the degree <strong>of</strong><br />

absorption can increase substantially (Scala et al., 1968). In vitro studies<br />

demonstrated that cationic surfactants are absorbed by the skin to a much<br />

lesser extent than anionic surfactants (Scala et al., 1968; Geisler, 1976;<br />

Faucher et al., 1979).<br />

The degree <strong>of</strong> percutaneous absorption is generally larger for nonionic<br />

surfactants than for anionic or cationic surfactants. Studies on the<br />

percutaneous absorption <strong>of</strong> alkyl polyethyleneglycol ethers <strong>of</strong> the structure<br />

C 12-(CH 2-CH 2-O) 3H, C 12-(CH 2-CH 2-O) 6H , C 12-(CH 2-CH 2-O) 10H, and<br />

C 15-(CH 2-CH 2-O) 3H were performed under conditions <strong>of</strong> use (Black and<br />

Howes, 1979). The aqueous solutions <strong>of</strong> the applied surfactants were<br />

washed <strong>of</strong>f after a contact time with the skin <strong>of</strong> 15 min. Under these<br />

conditions, the penetration <strong>of</strong> the alkyl polyethyleneglycol ethers was<br />

greater than the penetration <strong>of</strong> the analogous alcohol sulphates or alcohol<br />

ether sulphates. The penetration increased with increasing length <strong>of</strong> the<br />

carbon chain. Percutaneous absorption decreases for an ethylene oxide<br />

content <strong>of</strong> 6 moles or more in the ethoxylate moiety.<br />

Intestinal absorption, metabolism and excretion<br />

W.STERZEL 347<br />

The ingestion <strong>of</strong> surfactants is possible e.g. through the use <strong>of</strong><br />

surfactantcontaining toothpaste, through residues from dishwashing<br />

detergents and through traces <strong>of</strong> surfactants in potable water. Anionic<br />

surfactants are resorbed well in the intestine (Michael, 1968; Black and<br />

Howes, 1980; Bartnik and Künstler, 1987). After absorption, a part <strong>of</strong> this<br />

is excreted together with bile in the faeces and is subject to a enterohepatic<br />

cycle. The majority <strong>of</strong> the absorbed surfactant is metabolized in the liver


348 TOXICOLOGY OF SURFACTANTS<br />

and the respective metabolites are eliminated in the urine. The metabolic<br />

degradation <strong>of</strong> the linear alkyl chain is performed by -oxidation followed<br />

by β-oxidation. The ether-linkage in the ethoxylate portion <strong>of</strong> sulphated<br />

alcohol ethoxylates seems to be resistant to metabolism.<br />

Linear alkylbenzene sulphonates and branched alkylbenzene sulphonates<br />

are metabolized to short chain sulphophenyl carboxylic acids. N-alkyl<br />

sulphates are metabolized by -oxidation <strong>of</strong> the hydrophobic end followed<br />

by β-oxidation. Butyric acid-4-sulphate and acetic acid-2-sulphate are the<br />

end products, which are then further converted in small amounts nonenzymatically<br />

to sulphate and -butyrolactone (Ottery et al., 1970). Studies<br />

by Taylor et al. (1978) demonstrated that alkyl sulphonates are degraded<br />

via the same pathway as alkyl sulphates.<br />

Cationic surfactants can be assumed to be resorbed in the intestine only<br />

to a small extent. This was confirmed in a study with trimethyl cetyl<br />

ammonium bromide (Isomaa, 1975; Isomaa et al., 1976). Due to the low<br />

level <strong>of</strong> resorbed surfactant, an unquestionable identification <strong>of</strong> the<br />

metabolites was not possible. Parts <strong>of</strong> absorbed cationic surfactants were,<br />

as found for anionic surfactants, excreted together with bile in the faeces<br />

and to a lesser degree with the urine.<br />

Nonionic surfactants are resorbed to a large degree in the intestine<br />

(Drotman, 1980). A significant part <strong>of</strong> the material is eliminated with the<br />

bile. Cleavage <strong>of</strong> the ether linkage is obviously possible. Homologous<br />

ethyleneglycol ethers are probably generated as metabolites along with the<br />

corresponding carboxylic acids which are formed through oxidation <strong>of</strong> the<br />

terminal hydroxymethyl group (Drotman, 1980). The sorbitan fatty esters,<br />

which are <strong>of</strong>ten used as emulsifiers, and the ethoxylated fatty acid esters<br />

are hydrolysed in the gastrointestinal tract after oral administration<br />

through cleavage <strong>of</strong> the ester bond. While the resulting fatty acid is treated<br />

metabolically like a natural fatty acid, the polyol component <strong>of</strong> the<br />

sorbitan fatty acid is absorbed in the intestine, but is not further oxidized<br />

and is eliminated predominantly with the urine (Elworthy and Treon, 1967).<br />

Systemic effects<br />

Talking about systemic effects means, in contrast to local effects, the<br />

description <strong>of</strong> reactions arising after the substance has entered the organism<br />

after swallowing, skin penetration or inhalation. For surfactants,<br />

resorption through the skin has to be considered in particular. As described<br />

in the previous section, it is relatively small. But for products that<br />

frequently come into close contact with the skin, either unintentionally or<br />

due to their intended use, the resorption <strong>of</strong> very small amounts over a long<br />

period <strong>of</strong> time cannot be prevented.


Acute toxicity<br />

In general, the acute oral toxicity <strong>of</strong> surfactants is low. The LD 50 values<br />

typically range between several hundred and several thousand mg kg −1 <strong>of</strong><br />

bodyweight. This is <strong>of</strong> the same order <strong>of</strong> magnitude as for table salt<br />

(Swisher, 1968). The most important effects are damage to the mucous<br />

membranes <strong>of</strong> the gastrointestinal tract. High doses induce vomiting and<br />

diarrhea (Weaver and Griffith, 1969). Surfactants exhibit significantly<br />

higher toxicity when the gastrointestinal tract is by-passed through<br />

intravenous injections. Even at very low concentrations, the interaction<br />

with the membrane <strong>of</strong> erythrocytes leads to their destruction. Inhalation <strong>of</strong><br />

surfactant-containing dusts or aerosols in higher concentrations leads to<br />

disturbances <strong>of</strong> the lung function (Coate et al., 1978). This effect can be<br />

attributed to interactions with the surface active film that lines the vesicles<br />

<strong>of</strong> the lung (Kissler et al., 1981). As with local compatibility, there are also<br />

pronounced structure/activity relationships for acute toxicity. Gale (1953)<br />

has investigated the acute toxicity <strong>of</strong> sodium alkyl sulphates from C 8 to C 18<br />

and found the strongest effect for C 12 sulphate.<br />

The anaesthetic properties <strong>of</strong> certain alcohol ethoxylates which can be<br />

observed after intravenous application as well as after application to the<br />

skin or the mucous membranes are remarkable. Ethoxylates <strong>of</strong> unbranched<br />

primary alcohols with 9 ethylene oxide units were found to exhibit local<br />

anaesthetic properties starting with an alkyl chain <strong>of</strong> C 8. The activity<br />

increases with increasing chain length (Zipf and Dittmann, 1964).<br />

Chronic toxicity<br />

W.STERZEL 349<br />

In order to exclude any adverse effects arising from the repeated exposure<br />

against small amounts <strong>of</strong> surfactants over a prolonged period <strong>of</strong> time,<br />

representatives <strong>of</strong> all important classes <strong>of</strong> surfactants were examined for<br />

chronic toxic effects. In these tests, dosages <strong>of</strong> several thousands ppm were<br />

administered over a period <strong>of</strong> up to 2 years. No observable effects were<br />

detected with linear alkylbenzene sulphonates in 2 year studies with rats<br />

using concentrations up to 0.5 per cent (feed) or 0.1 per cent (drinking<br />

water) (Buehler et al., 1971). A sodium alkyl sulphate with an average<br />

chain length <strong>of</strong> C12 was tolerated by rats up to 1 per cent in the feed for 1<br />

year without any remarkable side effects (Fitzhugh and Nelson, 1948). C14 −16α-olefin sulphonates were applied over 2 years in a feeding study in<br />

dosages up to 0.5 per cent without causing any remarkable effect (Hunter<br />

and Benson, 1976). Analogous studies were reported for alcohol<br />

ethoxylates and alkylphenol sulphates, which revealed no toxic symptoms<br />

at doses up to 0.1 per cent and 1.4 per cent, respectively (Larson et al.,<br />

1963; Siwak et al., 1982). Studies on cationic surfactants reported a noobservable-effect-level<br />

<strong>of</strong> 0.25 per cent (Coulston et al., 1961). In all these


350 TOXICOLOGY OF SURFACTANTS<br />

long-term studies, the dosages that were tolerated without damage were in<br />

the range <strong>of</strong> several thousand ppm, indicating large margins <strong>of</strong> safety. This<br />

was confirmed by Hunter and Benson (1976), who calculated for a<br />

relevant example that the respective dosage lies at least by a factor <strong>of</strong> 1000<br />

over the estimated maximum daily exposure level <strong>of</strong> humans. Besides these<br />

data from animal experiments, a series <strong>of</strong> studies exists in which volunteers<br />

ingested considerable amounts <strong>of</strong> anionic or nonionic surfactants over<br />

several weeks, without any noticeable severe adverse effects (Swisher,<br />

1968).<br />

Mutagenicity<br />

Mutagenicity is the induction <strong>of</strong> irreversible changes in genetic material. If<br />

normal cells (somatic cells) are the target, malformation results in<br />

the developing organism. In case <strong>of</strong> the mature organism, it can lead to<br />

tumour formation. If germ cells are the target, the danger exists that the<br />

genetic defect will be passed on to the <strong>of</strong>fspring. All classes <strong>of</strong> surfactants<br />

have been evaluated in numerous test systems. The collected data allow the<br />

conclusion that surfactants pose no considerable risk <strong>of</strong> genetic damage<br />

(Yam et al., 1984; Fowler, 1988; Oba and Takei, 1992).<br />

Carcinogenicity<br />

Due to the widespread use and contact with surfactants the question <strong>of</strong><br />

irreversible damage has to be raised in addition to the problem <strong>of</strong> other<br />

chronic effects. The following compounds were evaluated for<br />

carcinogenicity after administration in the drinking water or feed:<br />

alkylbenzene sulphonate (Buehler et al., 1971), alkyl sulphates (Fitzhugh<br />

and Nelson, 1948), α-olefin<br />

sulphonates (Hunter and Benson, 1976), secalkane<br />

sulphonate (Quack and Rend, 1976), alcohol ether sulphates<br />

(Tusing et al., 1962; Siwak et al., 1982), alcohol ethoxylates (Siwak et al.,<br />

1982) and alkylphenol ethoxylates (Larson et al., 1963; Smyth and<br />

Calandra, 1969). None <strong>of</strong> these experiments provided any indication <strong>of</strong><br />

increased risk <strong>of</strong> cancer after oral ingestion <strong>of</strong> surfactants. The question <strong>of</strong><br />

possible carcinogenic effects <strong>of</strong> surfactants on the skin has also been<br />

studied extensively. Summaries exist by Oba and Takei (1992) and Siwak et<br />

al. (1982).<br />

Embryotoxicity<br />

The effects <strong>of</strong> substances on the organism during pregnancy can lead to<br />

delayed development or death <strong>of</strong> the embryo or malformation. Studies with<br />

the following surfactants revealed no indications <strong>of</strong> embryotoxic activity:<br />

alcohol ethoxylates (Nomura et al., 1980), α-olefin<br />

sulphonates (Palmer et


al., 1975), alcohol ether sulphates and linear alkylbenzene sulphonates<br />

(Nolen et al., 1975). Concerns which started with the publication in 1969<br />

(Mikami et al., 1969) that surfactants had caused malformations in animal<br />

studies could not be reproduced (Oba and Takei, 1980). The findings <strong>of</strong><br />

Mikami et al. (1969) were interpreted to be a result <strong>of</strong> methodical<br />

inadequacies and misinterpretations (Charlesworth, 1976).<br />

Summary<br />

Due to their physico-chemical properties, surfactants are capable <strong>of</strong><br />

reacting with biological membranes, proteins and enzymes. Most <strong>of</strong> their<br />

toxicological properties can be traced back to these interactions. During<br />

the application <strong>of</strong> surfactant-containing products, the most important<br />

aspect <strong>of</strong> consumer safety is local compatibility. No indications <strong>of</strong><br />

systemic, chronic or irreversible damage could be found. Estimates <strong>of</strong> the<br />

amounts <strong>of</strong> orally ingested surfactants typically encountered were reviewed<br />

by several authors. Based on these estimates, a total daily intake <strong>of</strong><br />

surfactants in the range <strong>of</strong> 0.3–3 mg per person was calculated by Swisher<br />

(1968). Due to the low rate <strong>of</strong> percutaneous absorption exposure through<br />

the skin can be neglected. If the above mentioned highest conceivable daily<br />

intake is compared with the dosage that was tolerated without adverse<br />

effects in studies concerning systemic effects, it becomes quite clear that<br />

these amounts can be regarded as harmless. In conclusion, it can be stated<br />

that the use <strong>of</strong> surfactants does not pose a health risk for humans.<br />

References<br />

W.STERZEL 351<br />

BARTNIK, F.G. and KÜNSTLER, K., 1987, Biological effects, toxicology and<br />

human safety, in Falbe, J. (Ed.). Surfactants in Consumer Products, Theory,<br />

Technology and Application, p. 475, Heidelberg: Springer Verlag.<br />

BLACK, J.G. and HOWES, D., 1979, J. Soc. Cosmet. Chem., 30, 157.<br />

BLACK, J.G. and HOWES, D., 1980, Absorption, metabolism, and excretion <strong>of</strong><br />

anionic surfactants, in Gloxhuber, C. (Ed.) Anionic Surfactants, Biochemistry,<br />

<strong>Toxicology</strong>, Dermatology; Surfactant Sci. Ser., Vol 10, p. 51, New York,<br />

Basel: Marcel Dekker.<br />

BLAKE-HASKINS, J.C., 1986, J. Soc. Cosmet. Chem., 37, 199.<br />

BLANK, H.J. and GOULD, E., 1961, J. Invest. Dermatol, 37, 311.<br />

BUEHLER, E.V., NEWMAN E.A. and KING, W.R., 1971, Toxicol. Appl.<br />

Pharmacol, 18, 83.<br />

CHANG, P.L., AMEEN, M., LAFFERTY, K.I., VAREY, P.A., DAVIDSON, A.R.<br />

and DAVIDSON, R.G., 1985, Anal. Biochem., 144, 362.<br />

CHARLESWORTH, F.A., 1976, Food Cosmet. Toxicol, 14, 152.<br />

COATE, W.B., BUSEY, W.M., SCJOENFISCH, W.H., BROWN, N.M. and<br />

NEWMAN, E.A., 1978, Toxicol Appl. Pharmacol, 45, 477.


352 TOXICOLOGY OF SURFACTANTS<br />

COULSTON, F., DROBECK, H.P., MIELENS, Z.E. and GARVIN, P.J., 1961,<br />

Toxicol. Appl. Pharmacol., 3, 584.<br />

DELEO, V.A., 1989, J. Toxicol-Cut. Ocular Toxicol., 8, 227.<br />

DRAIZE, J.H., WOODWARD, G. and CALVERY, H., 1944, J. Pharmacol. Exp.<br />

Ther., 82, 377.<br />

DRAIZE, J.H. and KELLEY, E.A., 1952, Drug Cosmet. Ind., 71, 36.<br />

DROTMAN, R.B., 1980, Toxicol. Appl. Pharmacol, 52, 38.<br />

ELWORTHY, P.H. and TREON, J.F., 1967, Physiologic activity <strong>of</strong> nonionic<br />

surfactants, in Schick, M.J. (Ed.) Nonionic Surfactants, p. 923, New York,<br />

Basel: Marcel Dekker.<br />

EMBERY, G. and DUGARD, P.H., 1969, Br. J. Derm., 81 Supplement 4, 63.<br />

FAUCHER, J.A., GODDARD, E.D. and KULKARNI, R.D., 1979, J. Am. Oil<br />

Chem. Soc., 56, 776.<br />

FITZHUGH, O.G. and NELSON, A.A., 1948, Am. Pharm. Assoc., 37, 29.<br />

FOWLER, C., 1988, Toxic. in Vitro, 2, 65.<br />

GALE, L.E. and SCOTT, P.M., 1953, J. Am. Pharm. Assoc., Sci. Ed., 42, 283.<br />

GARCIA DOMINGUEZ, J., 1977, J. Cosmet. Chem., 28, 165.<br />

GEISLER, R.W., 1976, Toxicol. Appl. Pharmacol, 37, 98.<br />

GÖTTE, E., 1967, Tenside, 4, 209.<br />

GRANT, W.W., 1962, <strong>Toxicology</strong> <strong>of</strong> the Eye, p. 511, Springfield 111: Charles C.<br />

Thomas.<br />

GREB, W. and WINGEN, F., 1980, Seifen, Fette, Öle, Wachse, 106, 327.<br />

GRUPP, T.C., DICK, L.C. and OSER, M., 1960, Toxicol. Appl. Pharmacol., 2,<br />

133.<br />

HAZLETON, L.W., 1952, Proc. Sci. Sect. Toilet Goods Ass., 17, 5.<br />

HELENIUS, A. and SIMONS, K., 1975, Biochim. Biophys. Acta, 414, 29.<br />

HOWES, D., 1975, J. Soc. Cosmet. Chem., 26, 47.<br />

HUNTER, B. and BENSON, H.G., 1976, <strong>Toxicology</strong>, 5, 359.<br />

IMOKAWA, G., SUMURA, K. and KATSUMI, M., 1975, J. Am. Oil Chem. Soc.,<br />

52, 484.<br />

ISOMAA, B., REUTER, J. and DJUPSUND, B.M., 1976, Arch. Toxicol, 35, 91.<br />

ISOMAA, B., 1975, Food Cosmet. Toxicol, 13, 231.<br />

JONES, M.N., 1975, Biochem. J., 151, 109.<br />

KÄSTNER, W., 1980, Local tolerance (animal tests): mucous membranes and skin,<br />

in Gloxhuber, C. (Ed.) Anionic Surfactants, Biochemistry, <strong>Toxicology</strong>,<br />

Dermatology; Surfactant Sci. Ser., Vol. 10, pp. 127, New York, Basel: Marcel<br />

Dekker.<br />

KISSLER, W., MORGENROTH, K. and WELLER, W., 1981, Prog. Resp. Res., 15,<br />

121.<br />

LARSON, P.S., BORZELLECA, J.F., BOWMAN, E.R., CRAWFORD, E.M.,<br />

SMITH, R.B.Jr., and HENNIGAR, G.R., 1963, Toxicol. Appl Pharmacol., 5,<br />

782.<br />

MAKINO, S., REYNOLDS, J.A. and TANFORD, C., 1973, J. Biol. Chem., 248,<br />

4926.<br />

MICHAEL, W.R., 1968, Toxicol. Appl Pharmacol., 12, 473.<br />

MIKAMI, Y., NAGAI, H., SAKAI, Y., FUKUSHIMA, S. and NISHINO, T., 1969,<br />

Cong. Anom. (Jap.), 9, 230.


W.STERZEL 353<br />

NAKAYA, K., YAMADA, K., ONOZAWA, M. and NAKAMURA, Y., l97l,<br />

Biochem. Biophys. Acta, 251, 7.<br />

NE’EMAN, Z., KAHANE, J. and RAZIN, S., 1971, Biochem. Biophys. Acta, 249,<br />

169, 176.<br />

NOLEN, G.A., KLUSMAN, L.W., PATRICK, L.F. and GEIL, R.G., 1975,<br />

<strong>Toxicology</strong>, 4, 231.<br />

NOMURA, T., KIMURA, S., HATA, S., KANZAKI, T. and TANAKA, H., 1980,<br />

Life Sci., 26, 49.<br />

NOZAKI, Y., REYNOLDS, J.A. and TANFORD, C., 1974, J.Biol. Chem., 249,<br />

4452.<br />

OBA, K. and TAKEI, R., 1992, Carcinogenic, mutagenic/genetic toxicity, and<br />

teratogenic properties, in Gloxhuber, C. and Künstler, K. (Eds) Anionic<br />

Surfactants, Biochemistry, <strong>Toxicology</strong>, Dermatology; Surfactant Sci. Ser., Vol.<br />

43, p. 331, New York, Basel: Marcel Dekker.<br />

OPDYKE, D.L. and BURNETT, M.C., 1965, Proc. Sci. Sect. Toilet. Goods Assoc.,<br />

44, 3.<br />

OTTERY, J., OLAVESEN, A.A. and DODGSON, K.S., 1970, Life Sci., 9, 1335.<br />

PALMER, A.K., READSHAW, M.A. and NEUFF, A.M., 1975, <strong>Toxicology</strong>, 3,<br />

107.<br />

QUACK, J.M. and REND, A.K., 1976, Fette-Seifen-Anstrichmittel, 78, 200.<br />

RICE, D.P., 1977, Appl. Pharmacol., 39, 377.<br />

SCALA, J., McOSTER, D.E. and RELLER, H.H., 1968, J. Invest. Dermatol., 50,<br />

371.<br />

SCHALLREUTER, K. and WOOD, J.M., 1986, Biochem. Biophys. Res.<br />

Commun., 135, 221.<br />

SCHOLZ, J., 1967, Arch. Exp. Pathol. Pharmacol, 232, 241.<br />

SCHWUGER, M.J. and BARTNIK, F.G., 1980, Interactions <strong>of</strong> anionic surfactants<br />

with proteins, enzymes and membranes, in Gloxhuber, C. (Ed.) Anionic<br />

Surfactants, Biochemistry, <strong>Toxicology</strong>, Dermatology; Surfactant Sci. Ser., Vol.<br />

10, p. 1, New York, Basel: Marcel Dekker.<br />

SIWAK, A., GOYER, M., PERWAK, J. and THAYER, P., 1982, in Mittal, K.L.,<br />

Fendler, E.J. (Eds), Solution Behavior <strong>of</strong> Surfactants, Vol. I, p. 161, New<br />

York, London: Plenum Publishing Corp.<br />

SMYTH, J.F. and CALANDRA, J.C., 1969, Toxicol. Appl. Pharmacol., 14, 315.<br />

SWISHER, R.D., 1968, Arch. Environ. Hth, 17, 232.<br />

TAYLOR, A.J., POWELL, G.M., HOWES, D., BLACK, J.G. and OLAVESEN, A.<br />

H., 1978, Biochem. J., 174, 405.<br />

TSUGE, H., 1984, Agric. Biol. Chem., 48, 19.<br />

TUSING, T.W., PAYNTER, O.E., OPDYKE, D.L. and SNYDER, F.H., 1962,<br />

Toxicol. Appl. Pharmacol., 4, 402.<br />

WASYLESWSKI, Z. and KOZIK, A., 1979, Eur. J. Biochem., 95, 121.<br />

WEAVER, J.E. and GRIFFITH, J.F., 1969, Toxicol. Appl Pharmacol., 8, 214.<br />

WESTER, R.C. and MAIBACH, H.I., 1982, Occup. Ind. Dermatol, p. 201,<br />

Chicago: Year Book, Med.<br />

YAM, J., BOOMAN, K.A., BRODDLE, W., GEIGER, L., HEINZE, J.E., LIN, Y.<br />

J., MCCARTHY, K., REISS, S., SAWIN, V., SEDLAK, R.I., SLESINSKI, R.S.<br />

and WRIGHT, G.A., 1984, Food Chem. Toxicol., 22, 761.<br />

ZIPF, H.E. and DITTMANN, E.C., 1964, Arch. Exp. Pathol. Pharmacol, 247, 544.


354


PART SEVEN<br />

Controversial mechanistic and regulatory<br />

issues in the safety assessment <strong>of</strong><br />

industrial chemicals


25<br />

Low Dose <strong>of</strong> a Genotoxic Carcinogen does not<br />

‘Cause’ Cancer; it Accelerates Spontaneous<br />

Carcinogenesis<br />

WERNER K.LUTZ<br />

University <strong>of</strong> Würzburg, Würzburg<br />

Definitions <strong>of</strong> cancer risk<br />

The risk <strong>of</strong> cancer is normally expressed as a fraction <strong>of</strong> a population<br />

diagnosed with cancer within a specified period <strong>of</strong> time. In an animal<br />

bioassay for carcinogenicity, this period usually is 2 years; in cancer<br />

epidemiology, a life span <strong>of</strong> 65 years (0–64) is <strong>of</strong>ten used. The two periods<br />

can be considered equivalent with respect to the process <strong>of</strong> carcinogenesis:<br />

its rate in different species is inversely correlated with the natural life span<br />

and basal metabolism and the background cancer incidence in 2-year old<br />

rats or mice is very similar to the one seen in 65-year-old humans<br />

(Anisimov, 1989; Raabe, 1989; Tennant, 1993).<br />

For an individual, a cancer risk can only be 0 or 1, depending on<br />

whether the situation is analysed before or after the diagnosis <strong>of</strong> the<br />

tumour. The population-based expression <strong>of</strong> a cancer risk therefore is not<br />

easily visualized and does not take into account interindividual differences<br />

in susceptibility.<br />

In the following discussion, the dose-response relationship in chemical<br />

carcinogenesis is analysed in terms <strong>of</strong> an effect <strong>of</strong> a carcinogen on the<br />

individual tumour latency time (Kodell et al., 1980; Littlefield et al., 1980;<br />

Day, 1983; Gaylor, 1992). Together with the idea <strong>of</strong> background DNA<br />

damage responsible for what is considered ‘spontaneous’ tumour formation<br />

and including individual variability for the rate <strong>of</strong> this process, it will be<br />

shown that low doses <strong>of</strong> genotoxic carcinogens might accelerate the<br />

spontaneous process <strong>of</strong> carcinogenesis but are not expected to induce<br />

cancer ‘out <strong>of</strong> the blue’.<br />

Linear dose response for a DNA-reactive carcinogen<br />

The effect <strong>of</strong> a carcinogen at low dose is normally extrapolated from data<br />

obtained in 2-year bioassays. At the end <strong>of</strong> the 2-year treatment period, the<br />

surviving animals are killed and analysed for the presence <strong>of</strong> tumours. The


fraction <strong>of</strong> tumour-bearing animals is then plotted against the dose, and<br />

lowdose effects are estimated from some model curve fitted to the data<br />

points.<br />

The shape <strong>of</strong> the dose-response curve at the low-dose end is strongly<br />

debated, especially for nongenotoxic carcinogens. For DNA-reactive<br />

carcinogens, it is widely accepted that there is no dose without effect, and a<br />

linear extrapolation is used (Lutz, 1990b). This is based on the idea that 1<br />

molecule <strong>of</strong> a DNA-reactive carcinogen could form a dangerous DNA<br />

adduct in a critical gene and activate an oncogene or inactivate a tumour<br />

suppressor gene by mutation, if the adduct is not repaired before DNA<br />

replication.<br />

Table 25.1 shows the consequences <strong>of</strong> linear interpolation between the<br />

tumour incidence in the controls and in a dosed group <strong>of</strong> a bioassay for<br />

carcinogenicity. An organ-specific tumour incidence <strong>of</strong> 4 per cent in the<br />

control group (2/50) and 14 per cent (7/50) after treatment for 2 years at<br />

10 mg kg −1 per day is assumed. With linear interpolation, a treatmentrelated<br />

increment in tumour incidence <strong>of</strong> 1 per cent would be calculated<br />

per mg kg −1 per day so that at 1 mg kg −1 per day, a 5 per cent total tumour<br />

incidence would be expected.<br />

In humans, increments in cancer risk in the per cent range would not be<br />

acceptable. A risk <strong>of</strong> 1 in 1 million lives might be considered negligible and<br />

the respective exposure could be regarded as ‘virtually safe.’ With the<br />

example given in Table 25.1 and upon linear interpolation, this ‘virtually<br />

safe’ dose would be calculated as 0.0001 mg kg −1 per day.<br />

What does it mean: ‘1 additional tumour in 1000 000<br />

lives’?<br />

The fact that a cancer risk is only 10 −6 cannot be a consolation for the<br />

affected individual. For this person, the cancer risk was 1. In the public<br />

opinion, therefore, an increase by one tumour case per one million lives is<br />

<strong>of</strong>ten interpreted to mean that one additional individual has got cancer<br />

who could otherwise have lived a much longer tumour-free life. For<br />

reasons explained below, this fear appears unfounded.<br />

Endogenous DNA damage; individual susceptibility<br />

W.K.LUTZ 357<br />

Carcinogenesis is a multi-stage process based on the accumulation <strong>of</strong> a<br />

number <strong>of</strong> critical DNA-related changes, due to, for example, DNAcarcinogen<br />

adducts. Evidence <strong>of</strong> background DNA damage from<br />

endogenous and unavoidable substances is accumulating (Ames, 1989;<br />

Loeb, 1989; Lutz, 1990a). It is due to, for instance, electrophiles such as<br />

S-adenosylmethionine, epoxides, quinones, or aldehydes, or to reactive<br />

oxygen species. In addition, DNA is not a chemically stable molecule, it


358 CARCINOGENESIS AT LOW DOSE<br />

Table 25.1 Linear low-dose interpolation <strong>of</strong> a cancer risk based on a hypothetical 2year<br />

bioassay for carcinogenicity<br />

Notes:<br />

Data in boldface; extrapolations in italics.<br />

depurinates and deaminates spontaneously. Finally, DNA replication is not<br />

100 per cent correct so that mutations cannot be avoided completely. The<br />

resulting background DNA damage is responsible for spontaneous<br />

mutations and for what is called spontaneous tumour formation. The<br />

process <strong>of</strong> carcinogenesis therefore has a non-zero rate even if exposure to<br />

exogenous DNA-reactive carcinogens could be avoided.<br />

The level <strong>of</strong> the background mutation rate is expected to show<br />

interindividual variability. It depends both upon genetic and life-style<br />

factors which govern, for instance, enzyme activities responsible for<br />

carcinogen metabolism or DNA repair (Harris, 1989). The rate <strong>of</strong><br />

spontaneous carcinogenesis is further governed by the inherited and<br />

acquired presence <strong>of</strong> activated oncogenes or absence <strong>of</strong> tumour suppressor<br />

genes (Scrable et al., 1990). Therefore, each individual in a heterogeneous<br />

population is expected to have its own endogenous cancer risk expressed as<br />

an individual time-to-tumour or tumourfree lifetime.<br />

Exogenous DNA damage; acceleration <strong>of</strong> spontaneous<br />

carcinogenesis<br />

Exposure to an additional, exogenous DNA-reactive molecule adds to the<br />

background DNA damage, increases the probability <strong>of</strong> a mutation and<br />

accelerates the multi-stage process <strong>of</strong> carcinogenesis. At low doses <strong>of</strong> the<br />

exogenous carcinogen, the rate <strong>of</strong> the process is expected to be dominated<br />

by the background damage so that the exogenous factor cannot constitute<br />

a cancer risk independent <strong>of</strong> the spontaneous process. The acceleration<br />

must be dose dependent and might be related to the background rate<br />

operating in each individual.<br />

It is true, therefore, that even a few molecules <strong>of</strong> a DNA-reactive<br />

carcinogen can have an effect. However, this effect cannot be a tumour


‘out <strong>of</strong> the blue’, in an individual that would otherwise have a low cancer<br />

risk. It ‘only’ reduces the individual’s tumour-free lifetime.<br />

No cancer ‘out <strong>of</strong> the blue’<br />

W.K.LUTZ 359<br />

Figure 25.1 Schematic representation <strong>of</strong> the time course <strong>of</strong> tumour appearance in a<br />

group <strong>of</strong> individuals with large differences in susceptibility. Solid line: background<br />

process <strong>of</strong> spontaneous carcinogenesis; arrows: acceleration <strong>of</strong> the spontaneous<br />

process by exposure to an additional carcinogen.<br />

This interpretation does not contradict the understanding that a low dose <strong>of</strong><br />

a carcinogen could increase the tumour incidence from 40000 to 40001 per<br />

1000000 lives, in the example shown in Table 25.1. The connection<br />

between the two approaches is shown in Figure 25.1. The solid line shows<br />

the appearance <strong>of</strong> a spontaneous tumour in individuals <strong>of</strong> a group <strong>of</strong> 20<br />

people. At the age <strong>of</strong> 65 years, 4 individuals have a tumour diagnosed. This<br />

is equivalent to a cumulative tumour incidence <strong>of</strong> 20 per cent. Exposure <strong>of</strong><br />

this group to an additional exogenous carcinogen would result in some<br />

reduction in the tumour-free lifespan in all individuals. In the example<br />

shown in Figure 25.1, this shift would move one additional individual to<br />

an age <strong>of</strong> diagnosis


360 CARCINOGENESIS AT LOW DOSE<br />

years. This is equivalent to an increase from 40000 to 40001 as a<br />

cumulative incidence 0–64, but it has a completely different meaning. It can<br />

now be excluded that the additional individual would have lived tumour<br />

free for 80 years in the absence <strong>of</strong> the exogenous carcinogen.<br />

Final remarks<br />

With the ideas presented, fear <strong>of</strong> cancer from low dose or from rare<br />

exposures can possibly be reduced. This does not mean that small cancer<br />

risks should be tolerated. Carcinogens in the environment, for instance,<br />

affect all <strong>of</strong> us; the tumour-free life span is reduced in the entire<br />

population.<br />

The model should be valid for tissues with a high spontaneous tumour<br />

incidence and an exponentially steep age dependence indicative <strong>of</strong> a<br />

multistage requirement <strong>of</strong> 5–6 steps. For cells that can be transformed in 2<br />

or 3 steps or that are specifically sensitive in certain phases <strong>of</strong> the<br />

development (in utero or during childhood), the model has to be<br />

reconsidered. This might be necessary for tumours with incidence peaks at<br />

a young age (leukaemia, tumours <strong>of</strong> the lymphatic tissues, brain, testis).<br />

Nevertheless, the latter tumour types are rare in comparison with cancer <strong>of</strong><br />

the old age so that the concept should hold for the majority <strong>of</strong> the tumours<br />

in humans.<br />

References<br />

AMES, B.N., 1989, Endogenous DNA damage as related to cancer and aging,<br />

Mutat. Res., 214, 41–6.<br />

ANISIMOV, V.N., 1989, Dependence <strong>of</strong> susceptibility to carcinogenesis on species<br />

life span, Arch. Geschwulstforsch., 59, 205–13.<br />

DAY, N.E., 1983, Time as a determinant <strong>of</strong> risk in cancer epidemiology: the role <strong>of</strong><br />

multi-stage models, Cancer Surv., 2, 577–93.<br />

GAYLOR, D.W., 1992, Relationship between the shape <strong>of</strong> dose-response curves<br />

and background tumour rates, Regul. Toxicol. Pharmacol., 16, 2–9.<br />

HARRIS, C.C., 1989, Interindividual variation among humans in carcinogen<br />

metabolism, DNA adduct formation and DNA repair, Carcinogenesis, 10,<br />

1563–6.<br />

KODELL, R.L., FARMER, J.H., LITTLEFIELD, N.A., FRITH, C.H., 1980, Analysis<br />

<strong>of</strong> life-shortening effects in female Balb/c mice fed 2-acetylamin<strong>of</strong>luorene, J.<br />

Environ. Pathol. Toxicol, 3 69–88.<br />

LITTLEFIELD, N.A., FARMER, J.H. and GAYLOR, D.W., 1980, Effects <strong>of</strong> dose<br />

and time in a long-term, low-dose carcinogenic study, J. Environ. Pathol.<br />

Toxicol., 3, 17–34.<br />

LOEB, L.A., 1989, Endogenous carcinogenesis: molecular oncology into the<br />

twentyfirst century—Presidential address, Cancer Res., 49, 5489–96.


W.K.LUTZ 361<br />

LUTZ, W.K., 1990a, Endogenous genotoxic agents and processes as a basis <strong>of</strong><br />

spontaneous carcinogenesis, Mutat. Res., 238, 287–95.<br />

LUTZ, W.K., 1990b, Dose response relationship and low dose extrapolation in<br />

chemical carcinogenesis, Carcinogenesis, 11, 1243–7.<br />

RAABE, O.G., 1989, Scaling <strong>of</strong> fatal cancer risks from laboratory animals to man,<br />

Hlth Phys., 57 suppl. 1, 419–32.<br />

SCRABLE, H.J., SAPIENZA, C. and CAVENEE, W.K., 1990, Genetic and<br />

epigenetic losses <strong>of</strong> heterozygosity in cancer predisposition and progression,<br />

Adv. Cancer Res., 54, 25–62.<br />

TENNANT, R.W., 1993, Stratification <strong>of</strong> rodent carcinogenicity bioassay results to<br />

reflect relative human hazard, Mutat. Res., 286, 111–18.


26<br />

Controversial Mechanistic and Regulatory Issues<br />

in Safety Assessment <strong>of</strong> <strong>Industrial</strong> Chemicals —an<br />

Industry Point <strong>of</strong> View<br />

HEINZ-PETER GELBKE<br />

BASF AG, Ludwigshafen<br />

Introduction<br />

In the appropriate classification and risk assessment <strong>of</strong> industrial chemicals<br />

three main players are involved: the scientific community, regulatory<br />

authorities and the chemical industry. The rules <strong>of</strong> the game are given by<br />

the definitions <strong>of</strong> the classification criteria and by guidelines for the risk<br />

assessment process. These definitions and guidelines are sometimes very<br />

flexible and open to different interpretations but in other cases precisely<br />

defined. Mostly these rules have been set up by regulatory bodies, e.g. the<br />

EU or national authorities, but sometimes also by scientific committees like<br />

the MAK commission in Germany or by IARC.<br />

Looking now at these three main players, the scientific community will<br />

provide the data as the starting point for each individual chemical. Possible<br />

controversies centre around the question, how data gaps may be bridged by<br />

scientifically valid assumptions. This indeed may <strong>of</strong>ten be necessary, if a<br />

complete toxicological and mechanistic data base is not available.<br />

Nevertheless, a scientifically based consensus can <strong>of</strong>ten be achieved for this<br />

bridging process.<br />

On the other hand, the other two players—industry and regulatory<br />

authorities—<strong>of</strong>ten do not reach mutually agreed decisions, although both <strong>of</strong><br />

them finally strive for the same target: protection <strong>of</strong> human health and the<br />

environment in an industrialized community. Controversial issues may<br />

sometimes stem from different approaches for bridging the scientific data<br />

gaps, but mostly they arise from political, economic or social aspects,<br />

which <strong>of</strong>ten are not outspoken. Just to give some examples: anticipated<br />

reaction <strong>of</strong> the society, possible emotions <strong>of</strong> the consumer, possible<br />

influences on the next election, availability <strong>of</strong> technical alternatives,<br />

different perceptions <strong>of</strong> the risk-benefit balance, overall economic situation<br />

<strong>of</strong> the community, different evaluations in other countries, impact on<br />

worldwide competitiveness, etc.<br />

In the following an industry point <strong>of</strong> view will be presented for some<br />

specific problems <strong>of</strong> today in the area <strong>of</strong> classification which is a more


qualitative approach and for risk assessment which in addition has to take<br />

into account quantitative aspects. This will be discussed separately for<br />

toxicological effects with thresholds (‘classical’ organ toxicity, reproductive<br />

and developmental toxicity) and without thresholds (mutagenicity,<br />

carcinogenicity). Apart from this subdivision there is one general concern<br />

<strong>of</strong> industry, that is to appropriately take into account exposure.<br />

Exposure<br />

Every classification and especially risk assessment decision should not only<br />

be based qualitatively on the toxicological pr<strong>of</strong>ile, but it should also take<br />

into account quantitatively the toxicological dose-response relationship as<br />

compared to human exposure.<br />

As a first rough approximation the different exposure pr<strong>of</strong>iles may be<br />

grouped into four main categories:<br />

1.<br />

Exposure during chemical production<br />

Many high production volume chemicals are used mainly or even<br />

exclusively as intermediates within the chemical industry. Although large<br />

amounts <strong>of</strong> these materials may be produced or processed within only a few<br />

facilities, exposure is <strong>of</strong>ten quite low and can be controlled or reduced by<br />

technical means. In addition there are many specific features enabling an<br />

efficient exposure control, such as: a trained workforce, site-specific and<br />

personal protection devices, stringent surveillance <strong>of</strong> workforce and work<br />

procedures, medical programmes tailored to the specificities <strong>of</strong> the work<br />

place, well defined exposures at the specific work sites, the exposed<br />

population is well known and limited, specified exposure duration,<br />

relatively homogeneous age and better health status <strong>of</strong> the workforce as<br />

compared to the general population, etc.<br />

2.<br />

Exposure <strong>of</strong> the downstream user during industrial/<br />

manufacturing applications<br />

H.-P.GELBKE 363<br />

In principle, the exposure scenarios may be quite similar to those <strong>of</strong><br />

chemical production, since many <strong>of</strong> the features described above also relate<br />

to or may be implemented at smaller workshops <strong>of</strong> the downstream user.<br />

Unfortunately, in reality <strong>of</strong>ten quite high exposures prevail in small<br />

workshops, possibly due to limited expenditures into exposure reduction<br />

measures or to a workforce not specifically trained for handling <strong>of</strong><br />

dangerous chemicals.


364 CONTROVERSIAL ISSUES IN SAFETY ASSESSMENT<br />

3.<br />

Exposure <strong>of</strong> the consumer<br />

When looking at the vast array <strong>of</strong> chemicals in use today, most <strong>of</strong> them<br />

will have industrial applications and only relatively few will directly be<br />

used by consumers. Especially highly reactive chemicals with their inherent<br />

potential for health hazards will generally not enter into consumer<br />

application fields simply due to their limited stability. But if a chemical gets<br />

to the consumer, efficient control measures can hardly be implemented, be<br />

it for the exposure per se, the exposed population, appropriate handling or<br />

prevention <strong>of</strong> misuse.<br />

4.<br />

Exposure <strong>of</strong> the general population via the environment<br />

Apart from highly reactive substances, all chemicals will to some extent<br />

enter the environment depending on their application fields and their<br />

processing to other end products. The environmental concentrations are<br />

determined by the amount released, by the distribution media, local<br />

situations and the efficacy <strong>of</strong> the different degradation processes. In<br />

general, the exposure <strong>of</strong> the population via the environment will be very<br />

low as compared to the workplace and health hazards are not to be<br />

expected. Of course highly persistent and highly toxic substances can be an<br />

exemption to this rule and should be monitored carefully.<br />

These exposure scenarios can be exemplified by a textile dyestuff:<br />

manufacturing within the chemical industry makes use <strong>of</strong> various starting<br />

materials and intermediates, which will not end up—apart from minute<br />

impurities—in the final product, and downstream exposure to these<br />

materials will be negligible. The manufacture <strong>of</strong> the dyestuff, its further<br />

handling, processing and formulation within the chemical industry can<br />

easily be controlled. But when this material is used downstream in textiledyeing<br />

workshops an efficient exposure control and appropriate handling<br />

may not always be guaranteed and higher exposures are conceivable.<br />

During the dyeing process, parts <strong>of</strong> the material enter into the<br />

environment, for example into aqueous media. This might lead to an<br />

exposure <strong>of</strong> the general population, albeit at very low concentrations.<br />

Consumer exposure can occur via migration <strong>of</strong> the dyestuff from the<br />

textile, sweat being the carrier medium or for small children the saliva, but<br />

exposure will most <strong>of</strong>ten be so low that a health hazard is not to be<br />

expected.<br />

It is one <strong>of</strong> the main concerns <strong>of</strong> industry within the processes <strong>of</strong><br />

classification and risk assessment that sufficient consideration is <strong>of</strong>ten not<br />

given to the different exposure pr<strong>of</strong>iles <strong>of</strong> each chemical. This results in<br />

simplified black and white decisions, which are not very helpful for an


appropriate and cost effective health protection within our industrialized<br />

world.<br />

Threshold effects<br />

Classification<br />

H.-P.GELBKE 365<br />

The classification system <strong>of</strong> the EU resides in the designation <strong>of</strong><br />

appropriate R-phrases. In most cases they are rather meant for hazard<br />

identification (e.g. irritation, sensitization, carcinogenicity) and not so<br />

much for risk characterization. Sometimes, risk aspects are also involved<br />

when specific dose levels are decisive for a specific R-phrase (e.g. acute<br />

toxicity). Thereby not only the effect per se is taken into account but also<br />

the strength <strong>of</strong> the effect.<br />

This latter principle also applies to the R 48-phrase (‘danger <strong>of</strong> serious<br />

damage to health by prolonged exposure'), if severe (irreversible) toxic<br />

effects are observed at dose levels <strong>of</strong> ≤50 mg kg −1 body weight per day in a<br />

90-day test after oral administration. For other routes and durations <strong>of</strong><br />

exposure similar provisions exist. The strategy to use dose limits is<br />

certainly appropriate for threshold effects.<br />

Although for reproductive and developmental effects thresholds are also<br />

accepted in most cases, no such dose limits are given apart from 1000 mg<br />

kg −1 body weight per day. This dose limit is not equivalent to that for the R<br />

48-phrase, because it only stems from the limit dose levels <strong>of</strong> the test<br />

guideline and is higher by more than one magnitude. Thus, for<br />

reproductive/ developmental toxicity classification, exposure and risk<br />

considerations are not influential in contrast to the R 48. Such a simplified<br />

‘yes or no’ approach for a threshold effect is not justifiable neither from a<br />

scientific point <strong>of</strong> view nor for an appropriate health protection. This is<br />

even more so, if the proposal for the ‘Restrictions on Marketing and Use<br />

Directive’ (13th Amendment to Directive 76/769/EEC) is implemented<br />

calling for a general prohibition <strong>of</strong> category 1 and 2 reproductive/<br />

developmental toxicants in consumer products in concentrations <strong>of</strong> ≥0.5%<br />

—if no specific concentration limit has been accepted according to the<br />

preparation guideline (Commission Directive 93/18/EEC; Council Directive<br />

88/379/EEC).<br />

The inconsistency <strong>of</strong> the approaches for ‘classical’ organ toxicity and<br />

developmental/reproductive toxicity can easily be demonstrated by the<br />

following theoretical example:<br />

For neurotoxicity a LOEL <strong>of</strong> 40 mg kg −1 day −1 in a 90-day oral test will<br />

result in a R 48-phrase, but a LOEL <strong>of</strong> 80 mg kg −1 day −1 would not<br />

lead to classification. On the other hand, slight foetal weight


366 CONTROVERSIAL ISSUES IN SAFETY ASSESSMENT<br />

reduction or impairment <strong>of</strong> fertility observed at 800 mg kg −1 day −1 in<br />

anappropriate developmental or reproductive test without parental<br />

toxicity would lead to classification into the respective category 3 or<br />

possibly category 2, even if a NOEL was found at 400 mg kg −1 day −1 .<br />

This certainly is not appropriate when considering the reversibility<br />

and severity <strong>of</strong> the effects and the differences in the NOELs and<br />

LOELs <strong>of</strong> one order <strong>of</strong> magnitude.<br />

Risk assessment<br />

The general strategy in risk assessment <strong>of</strong> chemicals with threshold effects<br />

is to use ‘assessment’ factors (‘uncertainty’ or ‘safety’ factors) (AF) for<br />

setting appropriate exposure limits. This concept was originally introduced<br />

by the WHO to establish ADI values (acceptable daily intake) for pesticide<br />

residues in food. Here generally a ‘safety’ factor <strong>of</strong> 100 is applied to the<br />

NOEL <strong>of</strong> a chronic experiment; higher or lower factors might be used for<br />

specific effects or experimental conditions.<br />

This basic approach can be generalized from consumer exposure to<br />

pesticide residues in food to other chemicals and exposure scenarios. The<br />

main problem then will be the selection <strong>of</strong> an appropriate AF. First <strong>of</strong> all,<br />

there are some general considerations to be taken into account:<br />

– Should AFs for the workforce and the general population differ because<br />

<strong>of</strong> the age characteristics and the general health status <strong>of</strong> workers?<br />

– Should different AFs be selected for chemicals and pesticides, taking into<br />

account that pesticides are specifically tailored for biological activity?<br />

– What are suitable AFs for route to route extrapolations if the<br />

experimental exposure does not correspond to that <strong>of</strong> humans? Thereby<br />

metabolic firstpass effects in the liver and different efficacies <strong>of</strong> the<br />

adsorption barriers <strong>of</strong> skin, lung and the intestines have to be taken into<br />

account.<br />

– What is the appropriate dose parameter, mg kg −1 body weight, mg m −2<br />

surface area or concentration?<br />

– What are suitable AFs for developmental effects which may occur in<br />

principle after a single exposure and may lead to irreversible lifetime<br />

impairment?<br />

– Are specific AFs necessary for toxic effects on the reproductive organs as<br />

compared to toxic effects on other organ systems?<br />

Apart from these general considerations there are also specific criteria<br />

decisive for the selection <strong>of</strong> AFs depending on each single chemical, its<br />

total data base and the experimental details. Very <strong>of</strong>ten the final AF is<br />

obtained by additional default factors which are to account for


H.-P.GELBKE 367<br />

experimental insufficiencies or to bridge data gaps. Just to give some<br />

examples for such specific considerations:<br />

– reversible versus irreversible effects,<br />

– duration <strong>of</strong> the study,<br />

– NOAEL versus NOEL,<br />

– LOAEL versus NOAEL,<br />

– local versus systemic effects,<br />

– species-specific effects,<br />

– species differences in anatomy or physiology,<br />

– similar versus different results observed in experiments with various<br />

species,<br />

– biokinetics and metabolism (e.g. metabolic pathways are species specific<br />

or occur only at high doses),<br />

– structure-activity considerations.<br />

This listing certainly not being complete clearly demonstrates that<br />

appropriate AFs cannot be arrived at by a simple cook-book procedure,<br />

but a flexible case-by-case approach is required for each individual<br />

chemical and data set. This is extremely important in order to avoid overconservative<br />

AFs; and in the long run over-conservative risk assessments<br />

are just as prohibitive for an appropriate health protection in an<br />

industrialized world as an underestimation <strong>of</strong> risk may lead to a more<br />

immediate danger to health. Over-conservative risk evaluations will result<br />

in a wrong allocation <strong>of</strong> resources, an unjustified prohibition <strong>of</strong> valuable<br />

chemicals, a wrong or unnecessary selection <strong>of</strong> alternative materials. etc.<br />

What might be an indication for an over-conservative AF? In principle,<br />

AFs for threshold effects should then be questioned to be over-conservative<br />

if they lead to acceptable human exposures which would also be<br />

appropriate for non-threshold effects (e.g. carcinogenicity) This can be<br />

exemplified by the following consideration:<br />

For a carcinogenicity experiment a ‘LOAEL’ in classical terms would<br />

be equivalent roughly to a dose just leading to a statistically increased<br />

tumour incidence <strong>of</strong> about 5 per cent. A ‘virtual NOAEL’ in the same<br />

classical sense without a statistically significant increase could then be<br />

at a dose with an actual tumour incidence <strong>of</strong> 1 per cent, which will<br />

not show up as a substance related effect under usual experimental<br />

conditions. At such a dose level the extra tumour risk would be 1/100.<br />

Applying an AF <strong>of</strong> 1000 to this ‘virtual NOAEL' would result in an<br />

exposure level with a risk <strong>of</strong> 1/10 5 , and an AF <strong>of</strong> 10000 in one with a<br />

risk <strong>of</strong> 1/10 6 using a simple linear extrapolation without further<br />

default considerations. Exposure levels with a risk <strong>of</strong> 1/10 5 or 1/10 6 are<br />

under discussion as ‘virtually safe doses’ for the workforce or the


368 CONTROVERSIAL ISSUES IN SAFETY ASSESSMENT<br />

general population. Thus, AFs <strong>of</strong> >1000 should always be questioned<br />

as possibly over-conservative for threshold effects, since exposure<br />

levels thereby obtained could also be acceptable for non-threshold<br />

effects like carcinogenicity. The assumptions underlying such high AFs<br />

should be re-examined critically.<br />

In the light <strong>of</strong> these considerations AFs for a developmental toxicity <strong>of</strong><br />

1000– 5000, as they are under discussion by some groups today, should<br />

also be questioned as possibly being over-conservative. It should not only be<br />

taken into account that developmental effects most <strong>of</strong>ten will have<br />

thresholds but also that their severities span a wide range from slight foetal<br />

weight impairment up to disabling malformations.<br />

In addition an unreflected selection <strong>of</strong> default factors in ‘classical’ organ<br />

toxicity can easily lead to over-conservative AFs: starting with a factor <strong>of</strong><br />

10 each for inter- and intra-species variability yields the AF <strong>of</strong> 100 used for<br />

ADI-calculations. In addition the following default factors are sometimes<br />

proposed:<br />

– Extrapolation from subacute/subchronic exposure to chronic exposure:<br />

a default factor <strong>of</strong> 10.<br />

– Extrapolation to the NOEL, if only a LOEL was obtained: a default<br />

factor <strong>of</strong> 2–5.<br />

– Taking into account an inappropriate experimental design: a default<br />

factor <strong>of</strong> 2–5–10.<br />

Thereby, for a multiple dose study with a marginal effect at the lowest dose<br />

level and an experimental design not fully in accordance with today’s<br />

standards, these default factors would result in a final assessment factor <strong>of</strong><br />

4000– 50000. It is highly questionable whether such an AF is really<br />

appropriate for threshold effects in comparison to the example given above<br />

for carcinogenicity.<br />

Non-threshold effects<br />

Other principles and approaches for classification and risk assessment have<br />

to be applied for non-threshold effects since safe exposure levels cannot be<br />

defined at which an adverse health effect will definitely not occur. Thus,<br />

for these compounds carcinogenic and mutagenic effects cannot be<br />

excluded even at very low dose levels albeit with extremely low<br />

probability.


Classification<br />

H.-P.GELBKE 369<br />

The present classification systems <strong>of</strong> scientific organizations (e.g. IARC,<br />

German MAK-commission) or regulatory agencies (e.g. EPA, EU<br />

commission) reside in quite a simplistic ‘strength <strong>of</strong> evidence’ approach:<br />

how valid are the experimental or epidemiological data?<br />

In future we should strive for a ‘weight <strong>of</strong> evidence’ approach which<br />

appears much more appropriate for a realistic human health protection,<br />

since it takes into consideration both risks and benefits <strong>of</strong> man-made and<br />

natural chemicals. Such a classification system basically asks the question:<br />

what do the experimental data really mean to humans at specific exposure<br />

levels? Thereby both qualitative and quantitative aspects are considered.<br />

Qualitatively whether and to what extent the mechanisms leading to an<br />

adverse effect in animals will also act in humans, and quantitatively to put<br />

the experimental dose-response relationship into context with human<br />

exposure. Of course, worst case exposure scenarios have to be taken into<br />

account. If under these considerations the experimental carcinogenic effect<br />

would not be relevant for humans a classification would not be<br />

appropriate.<br />

This latter quantitative aspect has led to discussions in several groups, as<br />

to whether a separate category for ‘weak carcinogens’ should be<br />

established, since more and more compounds turn out to be experimental<br />

carcinogens with a very weak or questionable effect. Such a category could<br />

be used for example for compounds which:<br />

– would only have insignificant effects even under worst case exposure<br />

scenarios,<br />

– did not give a carcinogenic response in appropriate animal experiments<br />

but are metabolized to carcinogenic intermediates or exert genotoxic<br />

effects in vivo,<br />

– show metabolic toxification to genotoxic metabolites only at high doses<br />

where the ‘normal’ metabolic detoxification pathway is overwhelmed.<br />

It is doubtful whether such a new category would really mean a step<br />

forward and be helpful. First <strong>of</strong> all why should compounds be classified if<br />

the carcinogenic effect is not to be expected in humans even under worst<br />

case exposure scenarios? And secondly how can the message <strong>of</strong> ‘weak<br />

carcinogenicity’ be brought over to the public without raising an emotional<br />

over-reaction to these compounds.<br />

Apart from these considerations on the general approach (‘strength’ or<br />

‘weight <strong>of</strong> evidence’) there is one specific major problem: presently, only<br />

criteria for classification are well defined, but those for non-classification<br />

are either not or only very vaguely described. It is also an important<br />

challenge to set up practicable and clear-cut non-classification criteria


370 CONTROVERSIAL ISSUES IN SAFETY ASSESSMENT<br />

which can be used by industry to tailor experiments in order to refute the<br />

classification <strong>of</strong> a questionable animal carcinogen. In the long run there<br />

will be no benefit if about 50 per cent <strong>of</strong> all chemicals are classified as<br />

carcinogens, neither for the public nor for industry nor for an adequate<br />

protection <strong>of</strong> human health.<br />

Risk assessment<br />

Most scientific committees and regulatory agencies refrain from<br />

scientifically based risk assessments for carcinogens but rather propagate<br />

an exposure as low as possible. And if a risk assessment is really carried<br />

out, it usually just applies a simplistic mathematical extrapolation using the<br />

linearized multistage model and highly conservative default assumptions to<br />

bridge data gaps. These mathematical procedures arrive at a scientifically<br />

unjustified numerical precision <strong>of</strong> the risk estimate. One <strong>of</strong> the problems is<br />

to explain to the public the real meaning and the uncertainties <strong>of</strong> such a<br />

risk assessment. A possible alternative could be to substitute the<br />

mathematical extrapolation by an appropriate assessment factor which <strong>of</strong><br />

course has to take into account the severity and irreversibility <strong>of</strong> the<br />

carcinogenic effect. The simplistic mathematical modelling might be used<br />

only for selection <strong>of</strong> priority chemicals for further in-depth investigations.<br />

On the other hand, a mathematical risk assessment can be an<br />

appropriate procedure for chemicals with a broad experimental data base,<br />

when the most relevant default assumptions are substituted by real data.<br />

This would be <strong>of</strong> primary importance for:<br />

– the selection <strong>of</strong> the mathematical model: the simplistic linearized<br />

multistage model presently in use could be substituted by biologically<br />

driven models, like that proposed by Sielken (1989) or the MVK-model<br />

(Moolgavkar and Knudson, 1981; Moolgavkar et al., 1988).<br />

– dose scaling from animals to humans: presently the experimental dose in<br />

mg kg −1 body weight is <strong>of</strong>ten extrapolated to humans by transforming<br />

the dose to mg m −2 body surface. This is used both for compounds<br />

which are metabolically toxified and detoxified, the scientific basis for<br />

such an undifferentiated procedure is at best highly doubtful. In the<br />

future this default assumption could be substituted by physiologically<br />

based pharmacokinetic (PBPK) modelling.<br />

– estimation <strong>of</strong> the target dose: presently the external dose to which the<br />

animals are exposed is considered to be proportional to the dose<br />

reaching the target tissue or the target chemical entities—generally the<br />

DNA. Again in future this could be substituted by adequate PBPK<br />

modelling.


For the time being, there are only few compounds with an experimental<br />

data base broad enough to substitute these default assumptions in every<br />

respect. But there are important industrial chemicals—and their number<br />

will increase eventually—for which at least some data are available for a<br />

justified substitution <strong>of</strong> part <strong>of</strong> the default assumptions. And this should be<br />

done as far as possible when a mathematical risk assessment is carried out.<br />

Conclusions<br />

Problems <strong>of</strong> classification and risk assessment have been discussed<br />

separately for toxicological effects with thresholds (‘classical’ organ<br />

toxicity, reproductive and developmental toxicity) and without thresholds<br />

(mutagenicity, carcinogenicity). With regard to the general procedures <strong>of</strong><br />

today, for industry there are two main points <strong>of</strong> concern:<br />

1. Not only for risk assessment but also for classification, exposure<br />

considerations should be taken into account. In principle, there are<br />

four different exposure scenarios: (a) for chemical production or within<br />

chemical industry, (b) for industrial application by downstream users,<br />

(c) for the consumer and (d) for the general population via the<br />

environment. For a given chemical the exposure may vary widely for<br />

the different scenarios, and there are many chemicals which are only<br />

used within chemical industry, which will never reach the consumer or<br />

which will enter into the environment only in minute quantities.<br />

Exposure estimates, including worst case scenarios, should be included<br />

in the processes <strong>of</strong> risk assessment and classification in order to avoid<br />

over-conservative results, which are not in the interest <strong>of</strong> adequate<br />

health protection.<br />

2. To get away from risk assessment procedures based on default<br />

assumptions which will <strong>of</strong>ten lead to over-conservative results; a case<br />

by case approach making use <strong>of</strong> all available data is scientifically far<br />

more appropriate.<br />

These problems have been elaborated for the:<br />

H.-P.GELBKE 371<br />

– classification <strong>of</strong> chemicals for reproductive or developmental toxicity<br />

within the regulatory framework <strong>of</strong> the EU,<br />

– selection <strong>of</strong> appropriate ‘assessment factors’ for chemicals with<br />

threshold effects,<br />

– classification and risk assessment <strong>of</strong> chemicals with non-threshold<br />

effects (especially carcinogens).


372 CONTROVERSIAL ISSUES IN SAFETY ASSESSMENT<br />

References<br />

MOOLGAVKAR, S.H. and KNUDSON, A.G., 1981, Mutation and cancer: a<br />

model for human carcinogenesis, Journal <strong>of</strong> the National Cancer Institute, 66,<br />

1037–52.<br />

MOOLGAVKAR, S.H., DEWANJI, A. and VENZON, D.J., 1988, A stochastic<br />

two-stage model for cancer risk assessment, I. The hazard function and the<br />

probability <strong>of</strong> tumour. Risk Analysis, 8, 383–92.<br />

SIELKEN, R.L.JR, 1989, Useful tools for evaluating and presenting more science in<br />

quantitative cancer risk assessments, Toxic Substances Journal, 9, 353–404.


Absorption <strong>of</strong> organic solvents 3–7<br />

Acceptable daily intake (ADI) 43, 168<br />

Acetylcholinesterase (AChE) 238<br />

N-acetyl-S-(5-ethoxy-l,2,4-thiadiazol-3yl-methyl)<br />

-L-cysteine (ET-MA) 26<br />

N-acetyl-β-glucosaminidase (NAG) 117<br />

Acid anhydrides 152–7<br />

Acrylamide 238<br />

Acrylonitrile 23, 313<br />

Adduct determination 81–7<br />

limitations <strong>of</strong> methods for 84<br />

Adduct formation <strong>of</strong> reactive<br />

compounds 74<br />

Adipate esters 223<br />

Airway hyperreactivity 128, 131<br />

Airway inflammation, animal models<br />

129<br />

Alachlor 208<br />

Alcohol ethoxylates, anaesthetic<br />

properties 349<br />

Alkaline phosphatase (ALP) 117<br />

Alkylating agents 76–9<br />

Alkyldeoxyguanosine adducts 186<br />

Allergic contact dermatitis 137<br />

Allometric equation 44, 46<br />

Allometric scaling 44–55<br />

Allometry 44<br />

Antioxidants 316–39<br />

Aromatic amines 74, 77<br />

Ascorbate 241<br />

Assessment factors (AFs) 366–71, 370<br />

Astrocytes 240<br />

ATP 78, 79, 159<br />

Axonal proteins 238<br />

Azo colorants 301<br />

carcinogenic 301–6<br />

Index<br />

Benzene 39<br />

Benzidene 301<br />

Benzo(a)pyrene (BP) 159, 186<br />

1,4-benzothiazines 64<br />

Benzotriazole-based light stabilisers<br />

322–29<br />

blood kinetics 323–29<br />

blood metabolites 323–29<br />

effects on rat dam and foetal liver<br />

329–4<br />

in vitro hydrolysis 323<br />

liver enzyme induction 327–31<br />

safety assessment 331, 334–7<br />

BGA, collaborative study 199<br />

Bioactivation mechanisms 11, 35–41<br />

Biocides 208<br />

Bioinactivation mechanisms 11<br />

Biological effect monitoring (BEM) 19<br />

Biological effects, determination <strong>of</strong> 189–<br />

6<br />

Biological monitoring (BM) 19–1<br />

definition 2<br />

organic solvents 2–3<br />

Biomarkers<br />

glutathione conjugation products as<br />

20–2<br />

<strong>of</strong> neurotoxicity 238–4<br />

Biotransformation 12<br />

Bisphenol A diglycidylether (BPADGE)<br />

83<br />

Body metabolic potential 48<br />

Body surface area (BSA) 46–8<br />

BP-DNA adducts 187<br />

Bromobenzene 63<br />

Bromo-diglutathionyl hydroquinones<br />

64


374 INDEX<br />

2-bromo-glutathionyl 64<br />

Bromohydroquinone 63–5<br />

o-bromophenol 63<br />

Bronchial challenge tests 149<br />

Bronchial hyperreactivity 151<br />

Bronchoalveolar lavage 117, 123<br />

1,3-butadiene<br />

PBPK/PBTK model 16<br />

physiologically based toxicokinetic<br />

modeling 31–3<br />

2-butoxyacetic acid (BAA) 172–80<br />

2-butoxyethanol, PBPK/PBTK model<br />

166–4, 172–80<br />

Calcium 245–9, 250, 267<br />

Cancer risk<br />

‘absolute’ 191<br />

assessment 188–6<br />

definitions 356<br />

management <strong>of</strong> 181<br />

Carbamazepine 322<br />

Carbendazim 285–8<br />

Carbon disulphide 238<br />

Carcinogen(esis) 179–6, 356<br />

azo colorants 301–6<br />

DNA-reactive 356<br />

dose-response relationship in 356<br />

epidemiological approaches to<br />

detect and identify 183–9<br />

genotoxic 181–8<br />

identification <strong>of</strong> 183–93<br />

peroxisome proliferation 224–30<br />

role <strong>of</strong> CYPs in activation and<br />

detoxication <strong>of</strong> 206<br />

spontaneous process 356, 357–3<br />

surfactants 350<br />

Carcinogenic potency 181<br />

Carcinogenic Substances Regulation<br />

301<br />

Catabolic metabolism 260<br />

Catalase 241<br />

Cell-mediated immune responses in<br />

chemical respiratory allergy 142–6<br />

Cellular nucleophiles 73<br />

Cerebral calcium accumulation 240<br />

Chemical pesticides, immunotoxicity <strong>of</strong><br />

203<br />

Chemical respiratory allergy, cellmediated<br />

immune responses in 142–6<br />

Chlordecone 241<br />

Chlorinated solvents 223<br />

Chlor<strong>of</strong>orm 38–1<br />

Chromates 301<br />

Chrysotile asbestos fibres<br />

pulmonary toxic effects 116–30<br />

size-separation methods 118–3<br />

Classification systems 368–3<br />

Color Index 301<br />

Colorants (dyes and pigments) 301–10<br />

regulatory aspects (FRG) 304–7<br />

Cosmetics 208<br />

Cultured porcine thyrocytes 261<br />

Cyclophosphamide 286–90<br />

Cytochrome P-450 (CYP) 15, 60, 74,<br />

206–15, 318, 327, 332<br />

Cytokine products <strong>of</strong> murine Th 1 and<br />

Th 2 cells 141<br />

Cytokines 140<br />

Dangerous compounds 208<br />

1,2-DCV-Cys 29, 31<br />

DCV-G 29<br />

1,2-DCV-G 29, 31<br />

1,2-DCV-Nac 29<br />

2,2-DCV-Nac 29<br />

Dermal uptake <strong>of</strong> organic solvents 5–7<br />

Detoxication, enzymes involved in 60–4<br />

Developmental neurotoxins 247–2<br />

Diagonal radioactive zones (DRZ) 158<br />

Diarylide pigments 81–3<br />

1,2-dibromoethane 40, 65–8<br />

3,3′-dichlorobenzidine (DCB) 81–3,<br />

301, 303<br />

1,2-dichloroethane 65–8<br />

2,5-dichloro-3-(glutathion-S-y1)<br />

hydroquinone 64<br />

Dichloromethane 39<br />

1,3-dichloropropene (DCP) 21–3<br />

S-(l,2-dichlorovinyl)glutathione. See 1,<br />

2-DCV-G<br />

di-(2-ethylhexyl)adipate (DEHA) 223,<br />

224, 227, 229–4<br />

di-(2-ethylhexyl)phthalate (DEHP) 223,<br />

224, 227–4, 286, 287


5α-dihydrotestosterone 212<br />

Diisocyanate asthma 151<br />

di-(isodecyl)phthalate 223<br />

3,3′-dimethoxybenzine 301<br />

3,3′-dimethylbenzidine 301<br />

Dioctyl phthalate 152<br />

Diphenylhydantoin 322<br />

2,6-di-tert-butyl-4-methyl phenol<br />

(BHT) 316–20<br />

DNA adducts 74, 77–81, 83, 84, 158,<br />

159–9, 183–91, 189, 224<br />

immunoenrichment <strong>of</strong> 185–2<br />

DNA binding 60<br />

DNA damage 183, 192, 208, 356<br />

endogenous 356–1<br />

exogenous 357–3<br />

DNA reactions 73<br />

DNA-reactive carcinogens 356<br />

dose-response curve 356<br />

DNA repair 192<br />

DNA replication 180, 192, 357<br />

DNA synthesis 227, 230<br />

Dopamine 246<br />

Dose determination 188<br />

Dose-response relationship<br />

and exposure pr<strong>of</strong>iles 362–68<br />

DNA-reactive carcinogens 356<br />

in carcinogenesis 356<br />

low-dose range 190<br />

DTH reactions 196<br />

Dyes. See Colorants (dyes and<br />

pigments)<br />

EC annex VII and VIII toxicity tests<br />

280<br />

ECETOC 202<br />

EDB, conjugation in rats and man 67<br />

Electrophilic agents 60, 71<br />

Electrophilic centres 72<br />

Electrophilic compounds, examples <strong>of</strong><br />

72<br />

Electrophilic metabolites 60<br />

Embryotoxicity tests 289–3<br />

Emulsifiers 312<br />

Endocrine dysfunction, xenobioticinduced<br />

254<br />

INDEX 375<br />

Endocrine toxicity, classification <strong>of</strong> 254–<br />

59<br />

Endocrine toxicology, thyroid 254–80<br />

Entire mammalian tests 283<br />

Environmental exposure 363<br />

Environmental monitoring (EM) 19–1<br />

Epoxide hydrolases (EH) 60<br />

Equivalent radiation dose concept 192–<br />

8<br />

Ethanol 241<br />

5-ethoxy-1,2,4-thiadiazole-3-carboxylic<br />

acid (ET-CA) 25–7<br />

Ethylene glycol methyl ether (EGME)<br />

286–91<br />

Etridiazol, disposition <strong>of</strong> 25–7<br />

European Union (EU) 167<br />

Exposure pr<strong>of</strong>iles 362–68<br />

Fatty amines 312<br />

FDA Segment I study for medicines 283<br />

Fecundity tests 284<br />

Fertility<br />

and embryotoxicity 285<br />

toxicity 281<br />

Fibre aerosols 97<br />

Fibre glass 91, 99–4<br />

airborne levels in workplace 109<br />

industrial hygiene studies 105–10<br />

lung fibre levels in workers 110<br />

Fibre recovery from lung tissue 118<br />

Fischer 344 study (Kimber-White) 198–<br />

4<br />

Flame retardant 312–18<br />

Flavin-containing monooxygenase<br />

(FMO) 211–16<br />

Fluoranthene-DNA adducts 187<br />

Foetal abnormalities 291<br />

Food additives 208<br />

Food and Agriculture Organization 208<br />

Fotemustine 46, 47<br />

Free radical formation 240–4, 246<br />

Full scale testing 288–1<br />

Furazolidone 68<br />

Gas chromatography (GC) 74, 76, 81


376 INDEX<br />

Gas chromatography/mass<br />

spectrometry (GC/MS) 74–9, 81, 84,<br />

185<br />

Genotoxic carcinogens 168<br />

Genotoxic hazards 181–8<br />

Genotoxicity 184, 281<br />

in vitro assays 184<br />

Germ cell mutagens 168<br />

Glial fibrillary acidic protein (GFAP)<br />

240–7, 247–2<br />

Gliotypic proteins 238–3<br />

Glutathione (GSH) 15, 20, 24, 184,<br />

241<br />

Glutathione conjugation, reversible 67–<br />

9<br />

Glutathione S-transferase (GST) 15, 20,<br />

24, 60–4, 66, 161, 212, 329<br />

Glycophorin A 160<br />

GM-CSF 143<br />

Growth desensitising mechanism<br />

(GDM) 263, 265<br />

Gunn rat hepatocytes in vitro, studies<br />

on 274<br />

Haemoglobin 76<br />

Haemoglobin adducts 189<br />

Health surveillance (HS) 19<br />

Heavy metals 244–9<br />

Hepatic metabolism, xenobiotics acting<br />

on 267–76<br />

‘Hepatic pharmacokinetic stuff’ 49<br />

Hepatocarcinogenesis, mechanisms <strong>of</strong><br />

226–1<br />

Hepatocytes<br />

co-cultures<br />

phase 1 reactions 210–16<br />

phase 2 reactions 212–17<br />

long-term cultures <strong>of</strong> 207–14<br />

Herbicides 208<br />

Hexamethylene diisocyanate (HDI) 151<br />

n-hexane 3, 7, 238<br />

2,5-hexanedione 3<br />

HGPRT mutation assay 192<br />

HHPA 152<br />

Himic anhydride (HA) 152<br />

Hormone elimination 269<br />

Hormone synthesis 258<br />

Host resistance (HR) studies 200<br />

HPLC 74–83, 84<br />

Human allergic disease 142<br />

Human clearance prediction 49, 53<br />

Human exposure monitoring 188<br />

Human unbound clearances 51–3<br />

Hydroxymethylethenodeoxyadenosine<br />

(HMEdA) 83<br />

1-hydroxypyrene 159<br />

Hypersensitivity reactions, Type I-IV<br />

196<br />

Hypothalamic-pituitary-thyroid-liver<br />

(H-P-T-L) axis 256–60<br />

investigative tests on 267<br />

thyroid toxicity via 265–76<br />

toxicological 266<br />

xenobiotic toxic effects on 258<br />

Hypoxanthin guanine phosphoribosyl<br />

transferase (HPRT) 160<br />

ICICIS collaborative study 197–3<br />

IFN- 140–6<br />

IgA 199, 200<br />

IgE 153<br />

IgE antibody 138<br />

IgE antibody responses, induction and<br />

regulation <strong>of</strong> 140–5<br />

IgG 153, 200<br />

IgG2a antibody 140<br />

IgM 200<br />

Immune system, evaluation <strong>of</strong> toxicity<br />

to 196–10<br />

Immunoenrichment <strong>of</strong> DNA adducts<br />

185–2<br />

Immunological methods 77, 78<br />

Immunotoxic side effects, screening <strong>of</strong><br />

197<br />

Immunotoxicity <strong>of</strong> chemical pesticides<br />

203<br />

Immunotoxicity testing, direct food<br />

additives 202<br />

Immunotoxicology 196<br />

collaborative studies 197, 198<br />

screening tests 196<br />

Insulin-like growth factor 1 (IGF,) 265<br />

Interferon (IFN- ) 140–6<br />

Interleukin 3 (IL-3) 141,143


Interleukin 4 (IL-4) 140, 141, 143<br />

Interleukin 5 (IL-5) 141–6<br />

Interleukin 10 (IL-10) 141<br />

Interleukin 12 (IL-12) 142<br />

International Agency for Research on<br />

Cancer (IARC) 91, 93–7, 163<br />

International Programme on Chemical<br />

Safety (IPCS) 91<br />

Iron 246<br />

Isocyanates 151–6<br />

Isophorone diisocyanate (IPDI) 151<br />

Joint Expert Committee on Food<br />

Additives (JECFA) 208<br />

Kainic acid (KA) 242–7<br />

Labelling 296–9<br />

Lactate dehydrogenase (LDH) 117<br />

Late respiratory systemic syndrome<br />

(LRRS) 152–7<br />

Lead 244–8<br />

Leaving group 65–8<br />

Life cycle exposure 283<br />

Life span correction 47–49<br />

Light microscopic histopathology 123–6<br />

Light stabilisers 316–39<br />

benzotriazole-based 322–9<br />

Linearized multistage cancer model<br />

(LMS) 170<br />

Lipophilic compounds 206<br />

Liver enzyme induction 318–2<br />

benzotriazole-based light stabilisers<br />

327–31<br />

LOAEL 168, 367<br />

Local lymph node assay (LLNA) 196<br />

LOEL 43<br />

Low molecular weight (MW) organic<br />

chemicals 187<br />

Lung burden analysis 103–8, 121–5<br />

Lung digestion/biodurability studies<br />

125<br />

Lung dissection 118<br />

Lung fibre burden 98–1<br />

Lung tissue, fibre recovery from 118<br />

MAK-list 304<br />

INDEX 377<br />

Maleic anhydride (MA) 152<br />

Malformations 291<br />

Malignancy, critical mutations leading<br />

to 190<br />

Manganese 245, 246<br />

Man-made vitreous fibres (MMVFs)<br />

animal inhalation studies 95–106<br />

carcinogenic potential 91–115<br />

cell culture studies 94<br />

comparison <strong>of</strong> human exposures<br />

used in rat chronic inhalation studies<br />

108–13<br />

epidemiological studies 91, 93–7<br />

implantation studies 95<br />

potential biological effects 91<br />

previous inhalation studies 106–9<br />

toxicologic studies 94<br />

Maximum life potential (MLP) 47–54<br />

Maximum tolerated dose (MTD) 170<br />

Meehs Formula 47<br />

Mercaptans 21<br />

Mercapturic acids 21–4, 184–90, 187–3<br />

toxicokinetics <strong>of</strong> 23<br />

urinary excretion 15, 25<br />

Metabolic activation 60–4<br />

Metabolism and toxicity 206–12<br />

Methamphetamine 241<br />

Methylene chloride 65<br />

Methylene diphenyldiisocyanate (MDI)<br />

151<br />

Methylmercury 241, 245<br />

N-methyl-N-nitrosourea (MNU) 263<br />

Michaelis-Menten kinetics 173<br />

Mitogenic stiraulation (ConA.LPS) 199<br />

Model neurotoxins 241–52<br />

Monitoring<br />

environmental (EM) 19–1<br />

human exposure 188<br />

in occupational toxicology 19–1<br />

polycyclic aromatic hydrocarbon<br />

(PAH) exposure 158–6<br />

see also Biological effect monitoring<br />

(BEM);<br />

Biological monitoring (BM)<br />

Monoclonal antibodies (Mabs) 185–2,<br />

318<br />

MPP + 241<br />

MPTP 241


378 INDEX<br />

mRNA analysis 211<br />

Mutagenic potency 190–6<br />

Mutagenicity, surfactants 349–2<br />

N 7 -deoxyguanosine (N 7 -dG) 186<br />

NADPH-cytochrome P450 reductase<br />

15<br />

Naphthalene diisocyanate (NDI) 151<br />

β-naphth<strong>of</strong>lavone 266, 267<br />

2-naphthylamine 40–3, 301<br />

Neuropathy target esterase (NTE) 238<br />

Neurotoxicity, biomarkers <strong>of</strong> 238–4<br />

Neurotoxicity assessment 285<br />

Neurotoxicity testing 237–56<br />

Neurotypic proteins 238–3<br />

Nitroarenes 74, 77<br />

NK activity 199<br />

NK test 200<br />

NOAEL 168, 174, 367<br />

NOEL 43, 366, 367<br />

Nucleophilic centres 73<br />

Occupational asthma 148–9<br />

chemical agents causing 148–5<br />

incidence 148, 151<br />

initial diagnosis 149<br />

Occupational toxicology, monitoring in<br />

19–1<br />

OECD guidelines 421 282–7<br />

OECD guidelines 422 282–91<br />

OECD single generation study 283<br />

17β-oestradiol 212<br />

Organic solvents<br />

absorption <strong>of</strong> 3–7<br />

biological monitoring <strong>of</strong> 2–3<br />

dermal uptake <strong>of</strong> 5–7<br />

pulmonary uptake <strong>of</strong> 3–5<br />

Organophosphate-induced delayed<br />

neuropathy (OPIDN) 238<br />

Organophosphate pesticides 237–2<br />

Parkinson’s disease 241, 245<br />

PBPK/PBTK models<br />

1,3-butadiene 16<br />

2-butoxyethanol 166–4, 172–80<br />

development <strong>of</strong> 31–3<br />

in risk assessment 170–80<br />

PCA-DNA adducts 187<br />

PCNB 267<br />

Pentafluorophenyl isothiocyanate<br />

(PFPITC) 76<br />

Pentafluorophenyl thiohydantoine<br />

(PFPTH) 77<br />

Perchlorate-discharge test 260<br />

Peroxisome proliferation 223–40<br />

carcinogenicity 224–30<br />

in rodent liver 223–8<br />

mechanisms <strong>of</strong> 226–1<br />

risk assessment 229–5<br />

rodent 223–9<br />

species differences in response 227–3<br />

Pesticides 203, 202<br />

human exposure to 208<br />

immunotoxicity <strong>of</strong> 203<br />

organophosphate 237–2<br />

Pharmaceuticals 202<br />

Phencyclidine 48<br />

Phenobarbital 267, 273<br />

Phenobarbitone 322<br />

Phenolic antioxidants 316–25<br />

blood kinetics 317–1<br />

blood metabolites 317–1<br />

effects on serum thyrotropin and<br />

thyroid hormones 318–4<br />

liver enzyme induction 318–2<br />

model compound 317<br />

risk assessment 321–5<br />

Phthalate esters 223, 224<br />

Phthalic anhydride 152<br />

Physiologically-based pharmaco<br />

(toxico-) kinetic models. See PBPK/<br />

PBTK models<br />

Phytopharmaceuticals 208<br />

Pigments. See Colorants (dyes and<br />

pigments)<br />

Plaque assay (PFCA) 199, 200<br />

Plasticisers 223, 229–4<br />

Polychlorinated biphenyls (PCBs) 247–<br />

2, 267<br />

Polycyclic aromatic hydrocarbons<br />

(PAHs) 74–7, 78, 158, 186<br />

biomonitoring exposure 158–6<br />

Polyisocyanates 151–6<br />

Postlabelling 78–79, 81<br />

Post-natal manifestations 284


Post-radiolabelling technology 184<br />

Prenatal effects 284, 285, 289<br />

Production volume triggers 280, 281<br />

Propylthiouracil (PTU) 260<br />

Protein adducts 73, 74, 79, 81, 184–90<br />

Protein-binding 258<br />

Pulmonary cell proliferation 118, 124–7<br />

Pulmonary hyperreactivity to industrial<br />

pollutants 128–9<br />

guinea-pig model 131–6<br />

rat model 134–7<br />

Pulmonary sensitization, mechanisms <strong>of</strong><br />

137–51<br />

Pulmonary toxic effects<br />

chrysotile asbestos fibres 116–30<br />

para-aramid fibrils 116–30<br />

Pulmonary uptake <strong>of</strong> organic solvents<br />

3–5<br />

Pyrethroid insecticides 238<br />

Pyromellitic dianhydride (PMDA) 152<br />

Quinone-thioethers 63<br />

Quinones 63–6<br />

Rad-equivalence values 192<br />

Radioallergosorbent tests (RAST) 138<br />

Radioimmunoassays 77<br />

Reactive airways dysfunction syndrome<br />

(RADS) 128, 148, 151, 153<br />

Reactive chemicals, metabolism 60–71<br />

Reactive compounds<br />

adduct formation <strong>of</strong> 74<br />

determination in unknown mixtures<br />

83–6<br />

determination <strong>of</strong> 71–88<br />

interaction with cellular constituents<br />

71–5<br />

Reactive metabolites 71<br />

Refractory ceramic fibres (RCFs) 91,<br />

99–2<br />

industrial hygiene studies 105<br />

Repeated dose toxicity 281<br />

Reproductive toxicity 279–298<br />

detecting effects on males 293<br />

evaluation for 282<br />

interpretation/extrapolation <strong>of</strong> 296–<br />

9<br />

INDEX 379<br />

labelling 296–9<br />

manifestations <strong>of</strong> 291–4<br />

methods for detecting effects on 282<br />

overlap <strong>of</strong> 281<br />

Respiratory allergic hypersensitivity<br />

137<br />

Restricted test systems 283<br />

Rifampicin 322<br />

Risk assessment 162–9, 166–84<br />

and exposure pr<strong>of</strong>iles 362–68<br />

approaches to 167–6<br />

cancer 188–6<br />

guidelines for 361<br />

in vitro approach 207–13<br />

mathematical procedures 369–3<br />

non-threshold effects 368<br />

peroxisome proliferation 229–5<br />

phenolic antioxidants 321–5<br />

physiologically based<br />

pharmacokinetic (PBPK) models in.<br />

See PBPK/PBTK models<br />

textile chemicals 315<br />

threshold effects 366–71<br />

see also Safety assessment<br />

RNA probes 211<br />

Rock (stone) wool 91, 102–5<br />

industrial hygiene studies 108<br />

R-phrases 364<br />

Safety assessment 361–74<br />

benzotriazole-based light stabilisers<br />

331–7<br />

see also Risk assessment<br />

Salmonella typhimurium 66<br />

SHIELD system 149<br />

Slag wool 91, 102–5<br />

industrial hygiene studies 108<br />

Solid phase assays 77<br />

Sperm analysis 284<br />

Spermatogenesis 284, 286, 288<br />

Structure-activity databases 282<br />

Structure-activity relationships,<br />

surfactants 344<br />

Styrene 38–38<br />

Styrene oxide 38–38<br />

Superoxide dismutase (SOD) 241<br />

Surface markers 200


380 INDEX<br />

Surfactants 338–55<br />

acute toxicity 348–1<br />

biochemical properties 338–5<br />

carcinogenicity 350<br />

chronic toxicity 349<br />

embryotoxicity 350<br />

excretion 347–50<br />

interactions with enzymes 341–5<br />

interactions with membranes 339<br />

interactions with proteins 339–3<br />

intestinal absorption 347–50<br />

local effects 342–48<br />

metabolism 347–50<br />

mucous membrane compatibility<br />

345<br />

mutagenicity 349–2<br />

oral toxicity 348<br />

percutaneous absorptions 346–9<br />

sensitization 345–8<br />

skin compatibility 342–7<br />

structure/activity relationships 344<br />

systemic effects 348<br />

toxicokinetics 346–50<br />

Surveillance <strong>of</strong> Work-related and<br />

Occupational Respiratory Disease<br />

Project (SWORD) 148<br />

Synaptophysin 242<br />

Systemic bioavailability <strong>of</strong> colorants<br />

301<br />

Systemic toxicity assessment 285<br />

Target dose determination 188–4<br />

Temelastine 271 272, 273<br />

2-tert-buty1–4-methoxyphenol (BHA)<br />

316<br />

Testosterone 212<br />

Tetrachloroethane 7<br />

Tetrachloroethene 8, 7<br />

Tetrachlorophthalic anhydride (TCPA)<br />

152, 153<br />

Textile chemicals 308–18<br />

finishing plant 309–13<br />

handling and processing 312–18<br />

irritant properties 311, 312<br />

new developments regarding<br />

toxicology 309<br />

oral toxicity 310<br />

process <strong>of</strong>f-gas 315<br />

risk assessment 315<br />

temperature effects 311<br />

toxicological pr<strong>of</strong>ile 311<br />

toxicology assessment needs 315<br />

Thalidomide 291<br />

T helper (Th) cells 140–5<br />

Thioethers 21–3<br />

Threshold effects<br />

classification 364–9<br />

risk assessment 366–71<br />

Thyroglobulin (TBG) 258, 260<br />

Thyroid<br />

endocrine toxicology 254–80<br />

tumours <strong>of</strong> 256<br />

Thyroid binding pre-albumin (TBPA)<br />

258–2<br />

Thyroid follicles 260<br />

Thyroid follicular capability 262<br />

Thyroid follicular cell hyperplasia and<br />

neoplasia, pathobiology <strong>of</strong> 262–7<br />

Thyroid function<br />

control <strong>of</strong> 258<br />

perturbation <strong>of</strong> 256–60<br />

Thyroid hormones 258, 260, 318–4<br />

Thyroid lesions, pathobiology <strong>of</strong> 258<br />

Thyroid neoplasia 262, 321<br />

Thyroid stimulating hormone (TSH)<br />

258, 260, 262, 263, 265, 320, 321,<br />

322<br />

Thyroid toxicity 256<br />

via H-P-T-L axis 265–76<br />

Thyroid tumorigenesis 264<br />

Thyrotrophin releasing hormone (TRH)<br />

258, 262<br />

Thyrotropin 318–4<br />

Thyroxine 260, 269<br />

accumulation 271, 273<br />

clearance <strong>of</strong> 267–76<br />

T lymphocytes 142<br />

Toluene 241<br />

2,4- and 2,6-toluene diisocyanate (TDI)<br />

151<br />

Toxicity, and metabolism 206–12<br />

Toxicity tests, EC annex VII and VIII<br />

280<br />

Toxicodynamic interactions 20<br />

Toxicokinetic interactions 20


Toxicokinetic parameters 16<br />

Toxicokinetics, principles <strong>of</strong> 15<br />

Transition metals 246<br />

Trichloroacetic acid (TCA) 28, 29<br />

Trichloroethane 7, 8<br />

Trichloroethylene (TRI) 26–31<br />

effects related to 26–31<br />

hepatocarcinogenicity induced by<br />

28<br />

oxidative metabolism 28, 29<br />

2,5,6-trichloro-3-glutathion-S-yl)<br />

hydroquinone 64<br />

Triethyl lead 241<br />

tri-(2-ethylhexyl)trimellitate) 223<br />

Trimellitic anhydride (TMA) 152, 153<br />

Trimethyltin (TMT) 241–6<br />

Tumour incidence 189–5<br />

Two generation studies 291–5<br />

alternatives to 295–8<br />

UDP-glucuronosyltransferase 321, 327,<br />

329<br />

UDP-glucuronyltransferase (UDP-GT)<br />

213, 260, 267, 271, 274<br />

University <strong>of</strong> Pittsburgh study 91, 92<br />

Urinary excretion<br />

mercapturic acids 15, 25<br />

xenobiotics 16–18<br />

US Environmental Protection Agency<br />

91<br />

US-NTP study 200<br />

Veterinary medicinal chemicals 202<br />

Vinyl chloride 36–9<br />

Western blotting 211<br />

World Health Organization (WHO) 91<br />

Xenobiotic-induced endocrine<br />

dysfunction 254<br />

Xenobiotics 11<br />

assessment <strong>of</strong> long-term toxicity<br />

207<br />

biological effects 14<br />

disposition <strong>of</strong> 12–15<br />

overall exposure to 20<br />

urinary excretion <strong>of</strong> 16–18<br />

INDEX 381

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!