Sunday, 16 January 2011 19:52

Carcinogen Risk Assessment

Rate this item
(2 votes)

Whereas the principles and methods of risk assessment for non-carcinogenic chemicals are similar in different parts of the world, it is striking that approaches for risk assessment of carcinogenic chemicals vary greatly. There are not only marked differences between countries, but even within a country different approaches are applied or advocated by various regulatory agencies, committees and scientists in the field of risk assessment. Risk assessment for non-carcinogens is rather consistent and pretty well established partly because of the long history and better understanding of the nature of toxic effects in comparison with carcinogens and a high degree of consensus and confidence by both scientists and the general public on methods used and their outcome.

For non-carcinogenic chemicals, safety factors were introduced to compensate for uncertainties in the toxicology data (which are derived mostly from animal experiments) and in their applicability to large, heterogeneous human populations. In doing so, recommended or required limits on safe human exposures were usually set at a fraction (the safety or uncertainty factor approach) of the exposure levels in animals that could be clearly documented as the no observed adverse effects level (NOAEL) or the lowest observed adverse effects level (LOAEL). It was then assumed that as long as human exposure did not exceed the recommended limits, the hazardous properties of chemical substances would not be manifest. For many types of chemicals, this practice, in somewhat refined form, continues to this day in toxicological risk assessment.

During the late 1960s and early 1970s regulatory bodies, starting in the United States, were confronted with an increasingly important problem for which many scientists considered the safety factor approach to be inappropriate, and even dangerous. This was the problem with chemicals that under certain conditions had been shown to increase the risk of cancers in humans or experimental animals. These substances were operationally referred to as carcinogens. There is still debate and controversy on the definition of a carcinogen, and there is a wide range of opinion about techniques to identify and classify carcinogens and the process of cancer induction by chemicals as well.

The initial discussion started much earlier, when scientists in the 1940s discovered that chemical carcinogens caused damage by a biological mechanism that was of a totally different kind from those that produced other forms of toxicity. These scientists, using principles from the biology of radiation-induced cancers, put forth what is referred to as the “non-threshold” hypothesis, which was considered applicable to both radiation and carcinogenic chemicals. It was hypothesized that any exposure to a carcinogen that reaches its critical biological target, especially the genetic material, and interacts with it, can increase the probability (the risk) of cancer development.

Parallel to the ongoing scientific discussion on thresholds, there was a growing public concern on the adverse role of chemical carcinogens and the urgent need to protect the people from a set of diseases collectively called cancer. Cancer, with its insidious character and long latency period together with data showing that cancer incidences in the general population were increasing, was regarded by the general public and politicians as a matter of concern that warranted optimal protection. Regulators were faced with the problem of situations in which large numbers of people, sometimes nearly the entire population, were or could be exposed to relatively low levels of chemical substances (in consumer products and medicines, at the workplace as well as in air, water, food and soils) that had been identified as carcinogenic in humans or experimental animals under conditions of relatively intense exposures.

Those regulatory officials were confronted with two fundamental questions which, in most cases, could not be fully answered using available scientific methods:

  1.  What risk to human health exists in the range of exposure to chemicals below the relatively intense and narrow exposure range under which a cancer risk could be directly measured?
  2.  What could be said about risks to human health when experimental animals were the only subjects in which risks for the development of cancer had been established?

 

Regulators recognized the need for assumptions, sometimes scientifically based but often also unsupported by experimental evidence. In order to achieve consistency, definitions and specific sets of assumptions were adapted that would be generically applied to all carcinogens.

Carcinogenesis Is a Multistage Process

Several lines of evidence support the conclusion that chemical carcinogenesis is a multistage process driven by genetic damage and epigenetic changes, and this theory is widely accepted in the scientific community all over the world (Barrett 1993). Although the process of chemical carcinogenesis is often separated into three stages—initiation, promotion and progression—the number of relevant genetic changes is not known.

Initiation involves the induction of an irreversibly altered cell and is for genotoxic carcinogens always equated with a mutational event. Mutagenesis as a mechanism of carcinogenesis was already hypothesized by Theodor Boveri in 1914, and many of his assumptions and predictions have subsequently been proven to be true. Because irreversible and self-replicating mutagenic effects can be caused by the smallest amount of a DNA-modifying carcinogen, no threshold is assumed. Promotion is the process by which the initiated cell expands (clonally) by a series of divisions, and forms (pre)neoplastic lesions. There is considerable debate as to whether during this promotion phase initiated cells undergo additional genetic changes.

Finally in the progression stage “immortality” is obtained and full malignant tumours can develop by influencing angiogenesis, escaping the reaction of the host control systems. It is characterized by invasive growth and frequently metastatic spread of the tumour. Progression is accompanied by additional genetic changes due to the instability of proliferating cells and selection.

Therefore, there are three general mechanisms by which a substance can influence the multistep carcinogenic process. A chemical can induce a relevant genetic alteration, promote or facilitate clonal expansion of an initiated cell or stimulate progression to malignancy by somatic and/or genetic changes.

Risk Assessment Process

Risk can be defined as the predicted or actual frequency of occurrence of an adverse effect on humans or the environment, from a given exposure to a hazard. Risk assessment is a method of systematically organizing the scientific information and its attached uncertainties for description and qualification of the health risks associated with hazardous substances, processes, actions or events. It requires evaluation of relevant information and selection of the models to be used in drawing inferences from that information. Further, it requires explicit recognition of uncertainties and appropriate acknowledgement that alternative interpretation of the available data may be scientifically plausible. The current terminology used in risk assessment was proposed in 1984 by the US National Academy of Sciences. Qualitative risk assessment changed into hazard characterization/identification and quantitative risk assessment was divided into the components dose-response, exposure assessment and risk characterization.

In the following section these components will be briefly discussed in view of our current knowledge of the process of (chemical) carcinogenesis. It will become clear that the dominant uncertainty in the risk assessment of carcinogens is the dose-response pattern at low dose levels characteristic for environmental exposure.

Hazard identification

This process identifies which compounds have the potential to cause cancer in humans—in other words it identifies their intrinsic genotoxic properties. Combining information from various sources and on different properties serves as a basis for classification of carcinogenic compounds. In general the following information will be used:

  • epidemiological data (e.g., vinylchloride, arsenic, asbestos)
  • animal carcinogenicity data
  • genotoxic activity/DNA adduct formation
  • mechanisms of action
  • pharmacokinetic activity
  • structure-activity relationships.

 

Classification of chemicals into groups based on the assessment of the adequacy of the evidence of carcinogenesis in animals or in man, if epidemiological data are available, is a key process in hazard identification. The best known schemes for categorizing carcinogenic chemicals are those of IARC (1987), EU (1991) and the EPA (1986). An overview of their criteria for classification (e.g., low-dose extrapolation methods) is given in table 1.

Table 1. Comparison of low-dose extrapolations procedures

  Current US EPA Denmark EEC UK Netherlands Norway
Genotoxic carcinogen Linearized multistage procedure using most appropriate low-dose model MLE from 1- and 2-hit models plus judgement of best outcome No procedure specified No model, scientific expertise and judgement from all available data Linear model using TD50 (Peto method) or “Simple Dutch Method” if no TD50 No procedure specified
Non-genotoxic carcinogen Same as above Biologically-based model of Thorslund or multistage or Mantel-Bryan model, based on tumour origin and dose-response Use NOAEL and safety factors Use NOEL and safety factors to set ADI Use NOEL and safety factors to set ADI  

 

One important issue in classifying carcinogens, with sometimes far-reaching consequences for their regulation, is the distinction between genotoxic and non-genotoxic mechanisms of action. The US Environmental Protection Agency (EPA) default assumption for all substances showing carcinogenic activity in animal experiments is that no threshold exists (or at least none can be demonstrated), so there is some risk with any exposure. This is com- monly referred to as the non-threshold assumption for genotoxic (DNA-damaging) compounds. The EU and many of its members, such as the United Kingdom, the Netherlands and Denmark, make a distinction between carcinogens that are genotoxic and those believed to produce tumours by non-genotoxic mechanisms. For genotoxic carcinogens quantitative dose-response estimation procedures are followed that assume no threshold, although the procedures might differ from those used by the EPA. For non-genotoxic substances it is assumed that a threshold exists, and dose-response procedures are used that assume a threshold. In the latter case, the risk assessment is generally based on a safety factor approach, similar to the approach for non-carcinogens.

It is important to keep in mind that these different schemes were developed to deal with risk assessments in different contexts and settings. The IARC scheme was not produced for regulatory purposes, although it has been used as a basis for developing regulatory guidelines. The EPA scheme was designed to serve as a decision point for entering quantitative risk assessment, whereas the EU scheme is currently used to assign a hazard (classification) symbol and risk phrases to the chemical's label. A more extended discussion on this subject is presented in a recent review (Moolenaar 1994) covering procedures used by eight governmental agencies and two often-cited independent organizations, the Inter- national Agency for Research on Cancer (IARC) and the American Conference of Governmental Industrial Hygienists (ACGIH).

The classification schemes generally do not take into account the extensive negative evidence that may be available. Also, in recent years a greater understanding of the mechanism of action of carcinogens has emerged. Evidence has accumulated that some mechanisms of carcinogenicity are species-specific and are not relevant for man. The following examples will illustrate this important phenomenon. First, it has been recently demonstrated in studies on the carcinogenicity of diesel particles, that rats respond with lung tumours to a heavy loading of the lung with particles. However, lung cancer is not seen in coal miners with very heavy lung burdens of particles. Secondly, there is the assertion of the nonrelevance of renal tumours in the male rat on the basis that the key element in the tumourgenic response is the accumulation in the kidney of α-2 microglobulin, a protein that does not exist in humans (Borghoff, Short and Swenberg 1990). Disturbances of rodent thyroid function and peroxisome proliferation or mitogenesis in the mouse liver have also to be mentioned in this respect.

This knowledge allows a more sophisticated interpretation of the results of a carcinogenicity bioassay. Research towards a better understanding of the mechanisms of action of carcinogenicity is encouraged because it may lead to an altered classification and to the addition of a category in which chemicals are classified as not carcinogenic to humans.

Exposure assessment

Exposure assessment is often thought to be the component of risk assessment with the least inherent uncertainty because of the ability to monitor exposures in some cases and the availability of relatively well-validated exposure models. This is only partially true, however, because most exposure assessments are not conducted in ways that take full advantage of the range of available information. For that reason there is a great deal of room for improving exposure distribution estimates. This holds for both external as well as for internal exposure assessments. Especially for carcinogens, the use of target tissue doses rather than external exposure levels in modelling dose-response relationships would lead to more relevant predictions of risk, although many assumptions on default values are involved. Physiologically based pharmacokinetic (PBPK) models to determine the amount of reactive metabolites that reaches the target tissue are potentially of great value to estimate these tissue doses.

Risk Characterization

Current approaches

The dose level or exposure level that causes an effect in an animal study and the likely dose causing a similar effect in humans is a key consideration in risk characterization. This includes both dose-response assessment from high to low dose and interspecies extrapolation. The extrapolation presents a logical problem, namely that data are being extrapolated many orders of magnitude below the experimental exposure levels by empirical models that do not reflect the underlying mechanisms for carcinogenicity. This violates a basic principle in fitting of empirical models, namely not to extrapolate outside the range of the observable data. Therefore, this empirical extrapolation results in large uncertainties, both from a statistical and from a biological point of view. At present no single mathematical procedure is recognized as the most appropriate one for low-dose extrapolation in carcinogenesis. The mathematical models that have been used to describe the relation between the administered external dose, the time and the tumour incidence are based on either tolerance-distribution or mechanistic assumptions, and sometimes based on both. A summary of the most frequently cited models (Kramer et al. 1995) is listed in table 2.

Table 2. Frequently cited models in carcinogen risk characterization

Tolerance distribution models Mechanistic models  
  Hit-models Biologically based models
Logit One-hit Moolgavkar (MVK)1
Probit Multihit Cohen and Ellwein
Mantel-Bryan Weibull (Pike)1  
Weibull Multistage (Armitage-Doll)1  
Gamma Multihit Linearized Multistage,  

1 Time-to-tumour models.

These dose-response models are usually applied to tumour-incidence data corresponding to only a limited number of experimental doses. This is due to the standard design of the applied bioassay. Instead of determining the complete dose-response curve, a carcinogenicity study is in general limited to three (or two) relatively high doses, using the maximum tolerated dose (MTD) as highest dose. These high doses are used to overcome the inherent low statistical sensitivity (10 to 15% over background) of such bioassays, which is due to the fact that (for practical and other reasons) a relatively small number of animals is used. Because data for the low-dose region are not available (i.e., cannot be determined experimentally), extrapolation outside the range of observation is required. For almost all data sets, most of the above-listed models fit equally well in the observed dose range, due to the limited number of doses and animals. However, in the low-dose region these models diverge several orders of magnitude, thereby introducing large uncertainties to the risk estimated for these low exposure levels.

Because the actual form of the dose-response curve in the low-dose range cannot be generated experimentally, mechanistic insight into the process of carcinogenicity is crucial to be able to discriminate on this aspect between the various models. Comprehensive reviews discussing the various aspects of the different mathematical extrapolation models are presented in Kramer et al. (1995) and Park and Hawkins (1993).

Other approaches

Besides the current practice of mathematical modelling several alternative approaches have been proposed recently.

Biologically motivated models

Currently, the biologically based models such as the Moolgavkar-Venzon-Knudson (MVK) models are very promising, but at present these are not sufficiently well advanced for routine use and require much more specific information than currently is obtained in bioassays. Large studies (4,000 rats) such as those carried out on N-nitrosoalkylamines indicate the size of the study which is required for the collection of such data, although it is still not possible to extrapolate to low doses. Until these models are further developed they can be used only on a case-by-case basis.

Assessment factor approach

The use of mathematical models for extrapolation below the experimental dose range is in effect equivalent to a safety factor approach with a large and ill-defined uncertainty factor. The simplest alternative would be to apply an assessment factor to the apparent “no effect level”, or the “lowest level tested”. The level used for this assessment factor should be determined on a case-by-case basis considering the nature of the chemical and the population being exposed.

Benchmark dose (BMD)

The basis of this approach is a mathematical model fitted to the experimental data within the observable range to estimate or interpolate a dose corresponding to a defined level of effect, such as one, five or ten per cent increase in tumour incidence (ED01, ED05, ED10). As a ten per cent increase is about the smallest change that statistically can be determined in a standard bioassay, the ED10 is appropriate for cancer data. Using a BMD that is within the observable range of the experiment avoids the problems associated with dose extrapolation. Estimates of the BMD or its lower confidence limit reflect the doses at which changes in tumour incidence occurred, but are quite insensitive to the mathematical model used. A benchmark dose can be used in risk assessment as a measure of tumour potency and combined with appropriate assessment factors to set acceptable levels for human exposure.

Threshold of regulation

Krewski et al. (1990) have reviewed the concept of a “threshold of regulation” for chemical carcinogens. Based on data obtained from the carcinogen potency database (CPDB) for 585 experiments, the dose corresponding to 10-6 risk was roughly log-normally distributed around a median of 70 to 90 ng/kg/d. Exposure to dose levels greater than this range would be considered unacceptable. The dose was estimated by linear extrapolation from the TD50 (the dose inducing toxicity is 50% of the animals tested) and was within a factor of five to ten of the figure obtained from the linearized multistage model. Unfortunately, the TD50 values will be related to the MTD, which again casts doubt on the validity of the measurement. However the TD50 will often be within or very close to the experimental data range.

Such an approach as using a threshold of regulation would require much more consideration of biological, analytical and mathematical issues and a much wider database before it could be considered. Further investigation into the potencies of various carcinogens may throw further light onto this area.

Objectives and Future of CarcinogenRisk Assessment

Looking back to the original expectations on the regulation of (environmental) carcinogens, namely to achieve a major reduction in cancer, it appears that the results at present are disappointing. Over the years it became apparent that the number of cancer cases estimated to be produced by regulatable carcinogens was disconcertingly small. Considering the high expectations that launched the regulatory efforts in the 1970s, a major anticipated reduction in the cancer death rate has not been achieved in terms of the estimated effects of environmental carcinogens, not even with ultraconservative quantitative assessment procedures. The main characteristic of the EPA procedures is that low-dose extrapolations are made in the same way for each chemical regardless of the mechanism of tumour formation in experimental studies. It should be noted, however, that this approach stands in sharp contrast to approaches taken by other governmental agencies. As indicated above, the EU and several European governments—Denmark, France, Germany, Italy, the Netherlands, Sweden, Switzerland, UK—distinguish between genotoxic and non-genotoxic carcinogens, and approach risk estimation differently for the two categories. In general, non-genotoxic carcinogens are treated as threshold toxicants. No effect levels are determined, and uncertainty factors are used to provide an ample margin of safety. To determine whether or not a chemical should be regarded as non-genotoxic is a matter of scientific debate and requires clear expert judgement.

The fundamental issue is: What is the cause of cancer in humans and what is the role of environmental carcinogens in that causation? The hereditary aspects of cancer in humans are much more important than previously anticipated. The key to signifi- cant advancement in the risk assessment of carcinogens is a better understanding of the causes and mechanisms of cancer. The field of cancer research is entering a very exciting area. Molecular research may radically alter the way we view the impact of environmental carcinogens and the approaches to control and prevent cancer, both for the general public and the workplace. Risk assessment of carcinogens needs to be based on concepts of the mechanisms of action that are, in fact, just emerging. One of the important aspects is the mechanism of heritable cancer and the interaction of carcinogens with this process. This knowledge will have to be incorporated into the systematic and consistent methodology that already exists for the risk assessment of carcinogens.

 

Back

Read 8909 times Last modified on Tuesday, 26 July 2022 19:40

" DISCLAIMER: The ILO does not take responsibility for content presented on this web portal that is presented in any language other than English, which is the language used for the initial production and peer-review of original content. Certain statistics have not been updated since the production of the 4th edition of the Encyclopaedia (1998)."

Contents

Toxicology References

Andersen, KE and HI Maibach. 1985. Contact allergy predictive tests on guinea pigs. Chap. 14 in Current Problems in Dermatology. Basel: Karger.

Ashby, J and RW Tennant. 1991. Definitive relationships among chemical structure, carcinogenicity and mutagenicity for 301 chemicals tested by the US NTP. Mutat Res 257:229-306.

Barlow, S and F Sullivan. 1982. Reproductive Hazards of Industrial Chemicals. London: Academic Press.

Barrett, JC. 1993a. Mechanisms of action of known human carcinogens. In Mechanisms of Carcinogenesis in Risk Identification, edited by H Vainio, PN Magee, DB McGregor, and AJ McMichael. Lyon: International Agency for Research on Cancer (IARC).

—. 1993b. Mechanisms of multistep carcinogenesis and carcinogen risk assessment. Environ Health Persp 100:9-20.

Bernstein, ME. 1984. Agents affecting the male reproductive system: Effects of structure on activity. Drug Metab Rev 15:941-996.

Beutler, E. 1992. The molecular biology of G6PD variants and other red cell defects. Annu Rev Med 43:47-59.

Bloom, AD. 1981. Guidelines for Reproductive Studies in Exposed Human Populations. White Plains, New York: March of Dimes Foundation.

Borghoff, S, B Short and J Swenberg. 1990. Biochemical mechanisms and pathobiology of a-2-globulin nephropathy. Annu Rev Pharmacol Toxicol 30:349.

Burchell, B, DW Nebert, DR Nelson, KW Bock, T Iyanagi, PLM Jansen, D Lancet, GJ Mulder, JR Chowdhury, G Siest, TR Tephly, and PI Mackenzie. 1991. The UPD-glucuronosyltransferase gene superfamily: Suggested nomenclature based on evolutionary divergence. DNA Cell Biol 10:487-494.

Burleson, G, A Munson, and J Dean. 1995. Modern Methods in Immunotoxicology. New York: Wiley.

Capecchi, M. 1994. Targeted gene replacement. Sci Am 270:52-59.

Carney, EW. 1994. An integrated perspective on the developmental toxicity of ethylene glycol. Rep Toxicol 8:99-113.

Dean, JH, MI Luster, AE Munson, and I Kimber. 1994. Immunotoxicology and Immunopharmacology. New York: Raven Press.

Descotes, J. 1986. Immunotoxicology of Drugs and Chemicals. Amsterdam: Elsevier.

Devary, Y, C Rosette, JA DiDonato, and M Karin. 1993. NFkB activation by ultraviolet light not dependent on a nuclear signal. Science 261:1442-1445.

Dixon, RL. 1985. Reproductive Toxicology. New York: Raven Press.

Duffus, JH. 1993. Glossary for chemists of terms used in toxicology. Pure Appl Chem 65:2003-2122.

Elsenhans, B, K Schuemann, and W Forth. 1991. Toxic metals: Interactions with essential metals. In Nutrition, Toxicity and Cancer, edited by IR Rowland. Boca-Raton: CRC Press.

Environmental Protection Agency (EPA). 1992. Guidelines for exposure assessment. Federal Reg 57:22888-22938.

—. 1993. Principles of neurotoxicity risk assessment. Federal Reg 58:41556-41598.

—. 1994. Guidelines for Reproductive Toxicity Assessment. Washington, DC: US EPA: Office of Research and Development.

Fergusson, JE. 1990. The Heavy Elements. Chap. 15 in Chemistry, Environmental Impact and Health Effects. Oxford: Pergamon.

Gehring, PJ, PG Watanabe, and GE Blau. 1976. Pharmacokinetic studies in evaluation of the toxicological and environmental hazard of chemicals. New Concepts Saf Eval 1(Part 1, Chapter 8):195-270.

Goldstein, JA and SMF de Morais. 1994. Biochemistry and molecular biology of the human CYP2C subfamily. Pharmacogenetics 4:285-299.

Gonzalez, FJ. 1992. Human cytochromes P450: Problems and prospects. Trends Pharmacol Sci 13:346-352.

Gonzalez, FJ, CL Crespi, and HV Gelboin. 1991. cDNA-expressed human cytochrome P450: A new age in molecular toxicology and human risk assessment. Mutat Res 247:113-127.

Gonzalez, FJ and DW Nebert. 1990. Evolution of the P450 gene superfamily: Animal-plant “warfare,” molecular drive, and human genetic differences in drug oxidation. Trends Genet 6:182-186.

Grant, DM. 1993. Molecular genetics of the N-acetyltransferases. Pharmacogenetics 3:45-50.

Gray, LE, J Ostby, R Sigmon, J Ferrel, R Linder, R Cooper, J Goldman, and J Laskey. 1988. The development of a protocol to assess reproductive effects of toxicants in the rat. Rep Toxicol 2:281-287.

Guengerich, FP. 1989. Polymorphism of cytochrome P450 in humans. Trends Pharmacol Sci 10:107-109.

—. 1993. Cytochrome P450 enzymes. Am Sci 81:440-447.

Hansch, C and A Leo. 1979. Substituent Constants for Correlation Analysis in Chemistry and Biology. New York: Wiley.

Hansch, C and L Zhang. 1993. Quantitative structure-activity relationships of cytochrome P450. Drug Metab Rev 25:1-48.

Hayes AW. 1988. Principles and Methods of Toxicology. 2nd ed. New York: Raven Press.

Heindell, JJ and RE Chapin. 1993. Methods in Toxicology: Male and Female Reproductive Toxicology. Vol. 1 and 2. San Diego, Calif.: Academic Press.

International Agency for Research on Cancer (IARC). 1992. Solar and ultraviolet radiation. Lyon: IARC.

—. 1993. Occupational Exposures of Hairdressers and Barbers and Personal Use of Hair Colourants: Some Hair Dyes, Cosmetic Colourants, Industrial Dyestuffs and Aromatic Amines. Lyon: IARC.

—. 1994a. Preamble. Lyon: IARC.

—. 1994b. Some Industrial Chemicals. Lyon: IARC.

International Commission on Radiological Protection (ICRP). 1965. Principles of Environmental Monitoring Related to the Handling of Radioactive Materials. Report of Committee IV of The International Commission On Radiological Protection. Oxford: Pergamon.

International Program on Chemical Safety (IPCS). 1991. Principles and Methods for the Assessment of Nephrotoxicity Associated With Exposure to Chemicals, EHC 119. Geneva: WHO.

—. 1996. Principles and Methods for Assessing Direct Immunotoxicity Associated With Exposure to Chemicals, EHC 180. Geneva: WHO.

Johanson, G and PH Naslund. 1988. Spreadsheet programming - a new approach in physiologically based modeling of solvent toxicokinetics. Toxicol Letters 41:115-127.

Johnson, BL. 1978. Prevention of Neurotoxic Illness in Working Populations. New York: Wiley.

Jones, JC, JM Ward, U Mohr, and RD Hunt. 1990. Hemopoietic System, ILSI Monograph, Berlin: Springer Verlag.

Kalow, W. 1962. Pharmocogenetics: Heredity and the Response to Drugs. Philadelphia: WB Saunders.

—. 1992. Pharmocogenetics of Drug Metabolism. New York: Pergamon.

Kammüller, ME, N Bloksma, and W Seinen. 1989. Autoimmunity and Toxicology. Immune Dysregulation Induced By Drugs and Chemicals. Amsterdam: Elsevier Sciences.

Kawajiri, K, J Watanabe, and SI Hayashi. 1994. Genetic polymorphism of P450 and human cancer. In Cytochrome P450: Biochemistry, Biophysics and Molecular Biology, edited by MC Lechner. Paris: John Libbey Eurotext.

Kehrer, JP. 1993. Free radicals as mediators of tissue injury and disease. Crit Rev Toxicol 23:21-48.

Kellerman, G, CR Shaw, and M Luyten-Kellerman. 1973. Aryl hydrocarbon hydroxylase inducibility and bronochogenic carcinoma. New Engl J Med 289:934-937.

Khera, KS. 1991. Chemically induced alterations maternal homeostasis and histology of conceptus: Their etiologic significance in rat fetal anomalies. Teratology 44:259-297.

Kimmel, CA, GL Kimmel, and V Frankos. 1986. Interagency Regulatory Liaison Group workshop on reproductive toxicity risk assessment. Environ Health Persp 66:193-221.

Klaassen, CD, MO Amdur and J Doull (eds.). 1991. Casarett and Doull´s Toxicology. New York: Pergamon Press.

Kramer, HJ, EJHM Jansen, MJ Zeilmaker, HJ van Kranen and ED Kroese. 1995. Quantitative methods in toxicology for human dose-response assessment. RIVM-report nr. 659101004.

Kress, S, C Sutter, PT Strickland, H Mukhtar, J Schweizer, and M Schwarz. 1992. Carcinogen-specific mutational pattern in the p53 gene in ultraviolet B radiation-induced squamous cell carcinomas of mouse skin. Cancer Res 52:6400-6403.

Krewski, D, D Gaylor, M Szyazkowicz. 1991. A model-free approach to low-dose extrapolation. Env H Pers 90:270-285.

Lawton, MP, T Cresteil, AA Elfarra, E Hodgson, J Ozols, RM Philpot, AE Rettie, DE Williams, JR Cashman, CT Dolphin, RN Hines, T Kimura, IR Phillips, LL Poulsen, EA Shephare, and DM Ziegler. 1994. A nomenclature for the mammalian flavin-containing monooxygenase gene family based on amino acid sequence identities. Arch Biochem Biophys 308:254-257.

Lewalter, J and U Korallus. 1985. Blood protein conjugates and acetylation of aromatic amines. New findings on biological monitoring. Int Arch Occup Environ Health 56:179-196.

Majno, G and I Joris. 1995. Apoptosis, oncosis, and necrosis: An overview of cell death. Am J Pathol 146:3-15.

Mattison, DR and PJ Thomford. 1989. The mechanism of action of reproductive toxicants. Toxicol Pathol 17:364-376.

Meyer, UA. 1994. Polymorphisms of cytochrome P450 CYP2D6 as a risk factor in carcinogenesis. In Cytochrome P450: Biochemistry, Biophysics and Molecular Biology, edited by MC Lechner. Paris: John Libbey Eurotext.

Moller, H, H Vainio and E Heseltine. 1994. Quantitative estimation and prediction of risk at the International Agency for Research on Cancer. Cancer Res 54:3625-3627.

Moolenaar, RJ. 1994. Default assumptions in carcinogen risk assessment used by regulatory agencies. Regul Toxicol Pharmacol 20:135-141.

Moser, VC. 1990. Screening approaches to neurotoxicity: A functional observational battery. J Am Coll Toxicol 1:85-93.

National Research Council (NRC). 1983. Risk Assessment in the Federal Government: Managing the Process. Washington, DC: NAS Press.

—. 1989. Biological Markers in Reproductive Toxicity. Washington, DC: NAS Press.

—. 1992. Biologic Markers in Immunotoxicology. Subcommittee on Toxicology. Washington, DC: NAS Press.

Nebert, DW. 1988. Genes encoding drug-metabolizing enzymes: Possible role in human disease. In Phenotypic Variation in Populations, edited by AD Woodhead, MA Bender, and RC Leonard. New York: Plenum Publishing.

—. 1994. Drug-metabolizing enzymes in ligand-modulated transcription. Biochem Pharmacol 47:25-37.

Nebert, DW and WW Weber. 1990. Pharmacogenetics. In Principles of Drug Action. The Basis of Pharmacology, edited by WB Pratt and PW Taylor. New York: Churchill-Livingstone.

Nebert, DW and DR Nelson. 1991. P450 gene nomenclature based on evolution. In Methods of Enzymology. Cytochrome P450, edited by MR Waterman and EF Johnson. Orlando, Fla: Academic Press.

Nebert, DW and RA McKinnon. 1994. Cytochrome P450: Evolution and functional diversity. Prog Liv Dis 12:63-97.

Nebert, DW, M Adesnik, MJ Coon, RW Estabrook, FJ Gonzalez, FP Guengerich, IC Gunsalus, EF Johnson, B Kemper, W Levin, IR Phillips, R Sato, and MR Waterman. 1987. The P450 gene superfamily: Recommended nomenclature. DNA Cell Biol 6:1-11.

Nebert, DW, DR Nelson, MJ Coon, RW Estabrook, R Feyereisen, Y Fujii-Kuriyama, FJ Gonzalez, FP Guengerich, IC Gunsalas, EF Johnson, JC Loper, R Sato, MR Waterman, and DJ Waxman. 1991. The P450 superfamily: Update on new sequences, gene mapping, and recommended nomenclature. DNA Cell Biol 10:1-14.

Nebert, DW, DD Petersen, and A Puga. 1991. Human AH locus polymorphism and cancer: Inducibility of CYP1A1 and other genes by combustion products and dioxin. Pharmacogenetics 1:68-78.

Nebert, DW, A Puga, and V Vasiliou. 1993. Role of the Ah receptor and the dioxin-inducible [Ah] gene battery in toxicity, cancer, and signal transduction. Ann NY Acad Sci 685:624-640.

Nelson, DR, T Kamataki, DJ Waxman, FP Guengerich, RW Estabrook, R Feyereisen, FJ Gonzalez, MJ Coon, IC Gunsalus, O Gotoh, DW Nebert, and K Okuda. 1993. The P450 superfamily: Update on new sequences, gene mapping, accession numbers, early trivial names of enzymes, and nomenclature. DNA Cell Biol 12:1-51.

Nicholson, DW, A All, NA Thornberry, JP Vaillancourt, CK Ding, M Gallant, Y Gareau, PR Griffin, M Labelle, YA Lazebnik, NA Munday, SM Raju, ME Smulson, TT Yamin, VL Yu, and DK Miller. 1995. Identification and inhibition of the ICE/CED-3 protease necessary for mammalian apoptosis. Nature 376:37-43.

Nolan, RJ, WT Stott, and PG Watanabe. 1995. Toxicologic data in chemical safety evaluation. Chap. 2 in Patty’s Industrial Hygiene and Toxicology, edited by LJ Cralley, LV Cralley, and JS Bus. New York: John Wiley & Sons.

Nordberg, GF. 1976. Effect and Dose-Response Relationships of Toxic Metals. Amsterdam: Elsevier.

Office of Technology Assessment (OTA). 1985. Reproductive Hazards in the Workplace. Document No. OTA-BA-266. Washington, DC: Government Printing Office.

—. 1990. Neurotoxicity: Identifying and Controlling Poisons of the Nervous System. Document No. OTA-BA-436. Washington, DC: Government Printing Office.

Organization for Economic Cooperation and Development (OECD). 1993. US EPA/EC Joint Project On the Evaluation of (Quantitative) Structure Activity Relationships. Paris: OECD.

Park, CN and NC Hawkins. 1993. Technology review; an overview of cancer risk assessment. Toxicol Methods 3:63-86.

Pease, W, J Vandenberg, and WK Hooper. 1991. Comparing alternative approaches to establishing regulatory levels for reproductive toxicants: DBCP as a case study. Environ Health Persp 91:141-155.

Prpi<F"WP MultinationalA Roman"P6.5>ƒ<F255P255>-Maji<F"WP MultinationalA Roman"P6.5%0>ƒ<F255P255>, D, S Telišman, and S Kezi<F"WP MultinationalA Roman"P6.5%0>ƒ<F255P255>. 1984. In vitro study on lead and alcohol interaction and the inhibition of erythrocyte delta-aminolevulinic acid dehydratase in man. Scand J Work Environ Health 10:235-238.

Reitz, RH, RJ Nolan, and AM Schumann. 1987. Development of multispecies, multiroute pharmacokinetic models for methylene chloride and 1,1,1-trichloroethane. In Pharmacokinetics and Risk Assessment, Drinking Water and Health. Washington, DC: National Academy Press.

Roitt, I, J Brostoff, and D Male. 1989. Immunology. London: Gower Medical Publishing.

Sato, A. 1991. The effect of environmental factors on the pharmacokinetic behaviour of organic solvent vapours. Ann Occup Hyg 35:525-541.

Silbergeld, EK. 1990. Developing formal risk assessment methods for neurotoxicants: An evaluation of the state of the art. In Advances in Neurobehavioral Toxicology, edited by BL Johnson, WK Anger, A Durao, and C Xintaras. Chelsea, Mich.: Lewis.

Spencer, PS and HH Schaumberg. 1980. Experimental and Clinical Neurotoxicology. Baltimore: Williams & Wilkins.

Sweeney, AM, MR Meyer, JH Aarons, JL Mills, and RE LePorte. 1988. Evaluation of methods for the prospective identification of early fetal losses in environmental epidemiology studies. Am J Epidemiol 127:843-850.

Taylor, BA, HJ Heiniger, and H Meier. 1973. Genetic analysis of resistance to cadmium-induced testicular damage in mice. Proc Soc Exp Biol Med 143:629-633.

Telišman, S. 1995. Interactions of essential and/or toxic metals and metalloids regarding interindividual differences in susceptibility to various toxicants and chronic diseases in man. Arh rig rada toksikol 46:459-476.

Telišman, S, A Pinent, and D Prpi<F"WP MultinationalA Roman"P6.5J255%0>ƒ<F255P255J0>-Maji<F"WP MultinationalA Roman"P6.5J255%0>ƒ<F255P255J0>. 1993. Lead interference in zinc metabolism and the lead and zinc interaction in humans as a possible explanation of apparent individual susceptibility to lead. In Heavy Metals in the Environment, edited by RJ Allan and JO Nriagu. Edinburgh: CEP Consultants.

Telišman, S, D Prpi<F"WP MultinationalA Roman"P6.5%0>ƒ<F255P255>-Maji<F"WP MultinationalA Roman"P6.5%0>ƒ<F255P255>, and S Kezi<F"WP MultinationalA Roman"P6.5%0>ƒ<F255P255>. 1984. In vivo study on lead and alcohol interaction and the inhibition of erythrocyte delta-aminolevulinic acid dehydratase in man. Scand J Work Environ Health 10:239-244.

Tilson, HA and PA Cabe. 1978. Strategies for the assessment of neurobehavioral consequences of environmental factors. Environ Health Persp 26:287-299.

Trump, BF and AU Arstila. 1971. Cell injury and cell death. In Principles of Pathobiology, edited by MF LaVia and RB Hill Jr. New York: Oxford Univ. Press.

Trump, BF and IK Berezesky. 1992. The role of cytosolic Ca2<F"Symbol"P8>+<F255P255> in cell injury, necrosis and apoptosis. Curr Opin Cell Biol 4:227-232.

—. 1995. Calcium-mediated cell injury and cell death. FASEB J 9:219-228.

Trump, BF, IK Berezesky, and A Osornio-Vargas. 1981. Cell death and the disease process. The role of cell calcium. In Cell Death in Biology and Pathology, edited by ID Bowen and RA Lockshin. London: Chapman & Hall.

Vos, JG, M Younes and E Smith. 1995. Allergic Hyper-sensitivities Induced by Chemicals: Recommendations for Prevention Published on Behalf of the World Health Organization Regional Office for Europe. Boca Raton, FL: CRC Press.

Weber, WW. 1987. The Acetylator Genes and Drug Response. New York: Oxford Univ. Press.

World Health Organization (WHO). 1980. Recommended Health-Based Limits in Occupational Exposure to Heavy Metals. Technical Report Series, No. 647. Geneva: WHO.

—. 1986. Principles and Methods for the Assessment of Neurotoxicity Associated With Exposure to Chemicals. Environmental Health Criteria, No.60. Geneva: WHO.

—. 1987. Air Quality Guidelines for Europe. European Series, No. 23. Copenhagen: WHO Regional Publications.

—. 1989. Glossary of Terms On Chemical Safety for Use in IPCS Publications. Geneva: WHO.

—. 1993. The Derivation of Guidance Values for Health-Based Exposure Limits. Environmental Health Criteria, unedited draft. Geneva: WHO.

Wyllie, AH, JFR Kerr, and AR Currie. 1980. Cell death: The significance of apoptosis. Int Rev Cytol 68:251-306.

@REFS LABEL = Other relevant readings

Albert, RE. 1994. Carcinogen risk assessment in the US Environmental Protection Agency. Crit. Rev. Toxicol 24:75-85.

Alberts, B, D Bray, J Lewis, M Raff, K Roberts, and JD Watson. 1988. Molecular Biology of the Cell. New York: Garland Publishing.

Ariens, EJ. 1964. Molecular Pharmacology. Vol.1. New York: Academic Press.

Ariens, EJ, E Mutschler, and AM Simonis. 1978. Allgemeine Toxicologie [General Toxicology]. Stuttgart: Georg Thieme Verlag.

Ashby, J and RW Tennant. 1994. Prediction of rodent carcinogenicity for 44 chemicals: Results. Mutagenesis 9:7-15.

Ashford, NA, CJ Spadafor, DB Hattis, and CC Caldart. 1990. Monitoring the Worker for Exposure and Disease. Baltimore: Johns Hopkins Univ. Press.

Balabuha, NS and GE Fradkin. 1958. Nakoplenie radioaktivnih elementov v organizme I ih vivedenie [Accumulation of Radioactive Elements in the Organism and their Excretion]. Moskva: Medgiz.

Balls, M, J Bridges, and J Southee. 1991. Animals and Alternatives in Toxicology Present Status and Future Prospects. Nottingham, UK: The Fund for Replacement of Animals in Medical Experiments.

Berlin, A, J Dean, MH Draper, EMB Smith, and F Spreafico. 1987. Immunotoxicology. Dordrecht: Martinus Nijhoff.

Boyhous, A. 1974. Breathing. New York: Grune & Stratton.

Brandau, R and BH Lippold. 1982. Dermal and Transdermal Absorption. Stuttgart: Wissenschaftliche Verlagsgesellschaft.

Brusick, DJ. 1994. Methods for Genetic Risk Assessment. Boca Raton: Lewis Publishers.

Burrell, R. 1993. Human immune toxicity. Mol Aspects Med 14:1-81.

Castell, JV and MJ Gómez-Lechón. 1992. In Vitro Alternatives to Animal Pharmaco-Toxicology. Madrid, Spain: Farmaindustria.

Chapman, G. 1967. Body Fluids and their Functions. London: Edward Arnold.

Committee on Biological Markers of the National Research Council. 1987. Biological markers in environmental health research. Environ Health Persp 74:3-9.

Cralley, LJ, LV Cralley and JS Bus (eds.). 1978. Patty’s Industrial Hygiene and Toxicology. New York: Witey.

Dayan, AD, RF Hertel, E Heseltine, G Kazantis, EM Smith, and MT Van der Venne. 1990. Immunotoxicity of Metals and Immunotoxicology. New York: Plenum Press.

Djuric, D. 1987. Molecular-cellular Aspects of Occupational Exposure to Toxic Chemicals. In Part 1 Toxicokinetics. Geneva: WHO.

Duffus, JH. 1980. Environmental Toxicology. London: Edward Arnold.

ECOTOC. 1986. Structure-Activity Relationship in Toxicology and Ecotoxicology, Monograph No. 8. Brussels: ECOTOC.

Forth, W, D Henschler, and W Rummel. 1983. Pharmakologie und Toxikologie. Mannheim: Biblio- graphische Institut.

Frazier, JM. 1990. Scientific criteria for Validation of in VitroToxicity Tests. OECD Environmental Monograph, no. 36. Paris: OECD.

—. 1992. In Vitro Toxicity—Applications to Safety Evaluation. New York: Marcel Dekker.

Gad, SC. 1994. In Vitro Toxicology. New York: Raven Press.

Gadaskina, ID. 1970. Zhiroraya tkan I yadi [Fatty Tissues and Toxicants]. In Aktualnie Vaprosi promishlenoi toksikolgii [Actual Problems in Occupational Toxicology], edited by NV Lazarev. Leningrad: Ministry of Health RSFSR.

Gaylor, DW. 1983. The use of safety factors for controlling risk. J Toxicol Environ Health 11:329-336.

Gibson, GG, R Hubbard, and DV Parke. 1983. Immunotoxicology. London: Academic Press.

Goldberg, AM. 1983-1995. Alternatives in Toxicology. Vol. 1-12. New York: Mary Ann Liebert.

Grandjean, P. 1992. Individual susceptibility to toxicity. Toxicol Letters 64/65:43-51.

Hanke, J and JK Piotrowski. 1984. Biochemyczne podstawy toksikologii [Biochemical Basis of Toxicology]. Warsaw: PZWL.

Hatch, T and P Gross. 1954. Pulmonary Deposition and Retention of Inhaled Aerosols. New York: Academic Press.

Health Council of the Netherlands: Committee on the Evaluation of the Carcinogenicity of Chemical Substances. 1994. Risk assessment of carcinogenic chemicals in The Netherlands. Regul Toxicol Pharmacol 19:14-30.

Holland, WC, RL Klein, and AH Briggs. 1967. Molekulaere Pharmakologie.

Huff, JE. 1993. Chemicals and cancer in humans: First evidence in experimental animals. Environ Health Persp 100:201-210.

Klaassen, CD and DL Eaton. 1991. Principles of toxicology. Chap. 2 in Casarett and Doull’s Toxicology, edited by CD Klaassen, MO Amdur and J Doull. New York: Pergamon Press.

Kossover, EM. 1962. Molecular Biochemistry. New York: McGraw-Hill.

Kundiev, YI. 1975.Vssavanie pesticidov cherez kozsu I profilaktika otravlenii [Absorption of Pesticides Through Skin and Prevention of Intoxication]. Kiev: Zdorovia.

Kustov, VV, LA Tiunov, and JA Vasiljev. 1975. Komvinovanie deistvie promishlenih yadov [Combined Effects of Industrial Toxicants]. Moskva: Medicina.

Lauwerys, R. 1982. Toxicologie industrielle et intoxications professionelles. Paris: Masson.

Li, AP and RH Heflich. 1991. Genetic Toxicology. Boca Raton: CRC Press.

Loewey, AG and P Siekewitz. 1969. Cell Structure and Functions. New York: Holt, Reinhart and Winston.

Loomis, TA. 1976. Essentials of Toxicology. Philadelphia: Lea & Febiger.

Mendelsohn, ML and RJ Albertini. 1990. Mutation and the Environment, Parts A-E. New York: Wiley Liss.

Mettzler, DE. 1977. Biochemistry. New York: Academic Press.

Miller, K, JL Turk, and S Nicklin. 1992. Principles and Practice of Immunotoxicology. Oxford: Blackwells Scientific.

Ministry of International Trade and Industry. 1981. Handbook of Existing Chemical Substances. Tokyo: Chemical Daily Press.

—. 1987. Application for Approval of Chemicals by Chemical Substances Control Law. (In Japanese and in English). Tokyo: Kagaku Kogyo Nippo Press.

Montagna, W. 1956. The Structure and Function of Skin. New York: Academic Press.

Moolenaar, RJ. 1994. Carcinogen risk assessment: international comparison. Regul Toxicol Pharmacol 20:302-336.

National Research Council. 1989. Biological Markers in Reproductive Toxicity. Washington, DC: NAS Press.

Neuman, WG and M Neuman. 1958. The Chemical Dynamic of Bone Minerals. Chicago: The Univ. of Chicago Press.

Newcombe, DS, NR Rose, and JC Bloom. 1992. Clinical Immunotoxicology. New York: Raven Press.

Pacheco, H. 1973. La pharmacologie moleculaire. Paris: Presse Universitaire.

Piotrowski, JK. 1971. The Application of Metabolic and Excretory Kinetics to Problems of Industrial Toxicology. Washington, DC: US Department of Health, Education and Welfare.

—. 1983. Biochemical interactions of heavy metals: Methalothionein. In Health Effects of Combined Exposure to Chemicals. Copenhagen: WHO Regional Office for Europe.

Proceedings of the Arnold O. Beckman/IFCC Conference of Environmental Toxicology Biomarkers of Chemical Exposure. 1994. Clin Chem 40(7B).

Russell, WMS and RL Burch. 1959. The Principles of Humane Experimental Technique. London: Methuen & Co. Reprinted by Universities Federation for Animal Welfare,1993.

Rycroft, RJG, T Menné, PJ Frosch, and C Benezra. 1992. Textbook of Contact Dermatitis. Berlin: Springer-Verlag.

Schubert, J. 1951. Estimating radioelements in exposed individuals. Nucleonics 8:13-28.

Shelby, MD and E Zeiger. 1990. Activity of human carcinogens in the Salmonella and rodent bone-marrow cytogenetics tests. Mutat Res 234:257-261.

Stone, R. 1995. A molecular approach to cancer risk. Science 268:356-357.

Teisinger, J. 1984. Expositiontest in der Industrietoxikologie [Exposure Tests in Industrial Toxicology]. Berlin: VEB Verlag Volk und Gesundheit.

US Congress. 1990. Genetic Monitoring and Screening in the Workplace, OTA-BA-455. Washington, DC: US Government Printing Office.

VEB. 1981. Kleine Enzyklopaedie: Leben [Life]. Leipzig: VEB Bibliographische Institut.

Weil, E. 1975. Elements de toxicologie industrielle [Elements of Industrial Toxicology]. Paris: Masson et Cie.

World Health Organization (WHO). 1975. Methods Used in USSR for Establishing Safe Levels of Toxic Substances. Geneva: WHO.

1978. Principles and Methods for Evaluating the Toxicity of Chemicals, Part 1. Environmental Health Criteria, no.6. Geneva: WHO.

—. 1981. Combined Exposure to Chemicals, Interim Document no.11. Copenhagen: WHO Regional Office for Europe.

—. 1986. Principles of Toxicokinetic Studies. Environmental Health Criteria, no. 57. Geneva: WHO.

Yoftrey, JM and FC Courtice. 1956. Limphatics, Lymph and Lymphoid Tissue. Cambridge: Harvard Univ. Press.

Zakutinskiy, DI. 1959. Voprosi toksikologii radioaktivnih veshchestv [Problems of Toxicology of Radioactive Materials]. Moscow: Medgiz.

Zurlo, J, D Rudacille, and AM Goldberg. 1993. Animals and Alternatives in Testing: History, Science and Ethics. New York: Mary Ann Liebert.