Sunday, 16 January 2011 18:45

Biomarkers

Rate this item
(0 votes)

The word biomarker is short for biological marker, a term that refers to a measurable event occurring in a biological system, such as the human body. This event is then interpreted as a reflection, or marker, of a more general state of the organism or of life expectancy. In occupational health, a biomarker is generally used as an indicator of health status or disease risk.

Biomarkers are used for in vitro as well as in vivo studies that may include humans. Usually, three specific types of biological markers are identified. Although a few biomarkers may be difficult to classify, usually they are separated into biomarkers of exposure, biomarkers of effect or biomarkers of susceptibility (see table 1).

Table 1. Examples of biomarkers of exposure or biomarkers of effect  that are used in toxicological studies in occupational health

Sample Measurement Purpose
Exposure biomarkers
Adipose tissue Dioxin Dioxin exposure
Blood Lead Lead exposure
Bone Aluminium Aluminium exposure
Exhaled breath Toluene Toluene exposure
Hair Mercury Methylmercury exposure
Serum Benzene Benzene exposure
Urine Phenol Benzene exposure
Effect biomarkers
Blood Carboxyhaemoglobin Carbon monoxide exposure
Red blood cells Zinc-protoporphyrin Lead exposure
Serum Cholinesterase Organophosphate exposure
Urine Microglobulins Nephrotoxic exposure
White blood cells DNA adducts Mutagen exposure

 

Given an acceptable degree of validity, biomarkers may be employed for several purposes. On an individual basis, a biomarker may be used to support or refute a diagnosis of a particular type of poisoning or other chemically-induced adverse effect. In a healthy subject, a biomarker may also reflect individual hypersusceptibility to specific chemical exposures and may therefore serve as a basis for risk prediction and counselling. In groups of exposed workers, some exposure biomarkers can be applied to assess the extent of compliance with pollution abatement regulations or the effectiveness of preventive efforts in general.

Biomarkers of Exposure

An exposure biomarker may be an exogenous compound (or a metabolite) within the body, an interactive product between the compound (or metabolite) and an endogenous component, or another event related to the exposure. Most commonly, biomarkers of exposures to stable compounds, such as metals, comprise measurements of the metal concentrations in appropriate samples, such as blood, serum or urine. With volatile chemicals, their concentration in exhaled breath (after inhalation of contamination-free air) may be assessed. If the compound is metabolized in the body, one or more metabolites may be chosen as a biomarker of the exposure; metabolites are often determined in urine samples.

Modern methods of analysis may allow separation of isomers or congeners of organic compounds, and determination of the speciation of metal compounds or isotopic ratios of certain elements. Sophisticated analyses allow determination of changes in the structure of DNA or other macromolecules caused by binding with reactive chemicals. Such advanced techniques will no doubt gain considerably in importance for applications in biomarker studies, and lower detection limits and better analytical validity are likely to make these biomarkers even more useful.

Particularly promising developments have occurred with biomarkers of exposure to mutagenic chemicals. These compounds are reactive and may form adducts with macromolecules, such as proteins or DNA. DNA adducts may be detected in white blood cells or tissue biopsies, and specific DNA fragments may be excreted in the urine. For example, exposure to ethylene oxide results in reactions with DNA bases, and, after excision of the damaged base, N-7-(2-hydroxyethyl)guanine will be eliminated in the urine. Some adducts may not refer directly to a particular exposure. For example, 8-hydroxy-2´-deoxyguanosine reflects oxidative damage to DNA, and this reaction may be triggered by several chemical compounds, most of which also induce lipid peroxidation.

Other macromolecules may also be changed by adduct formation or oxidation. Of special interest, such reactive compounds may generate haemoglobin adducts that can be determined as biomarkers of exposure to the compounds. The advantage is that ample amounts of haemoglobin can be obtained from a blood sample, and, given the four-month lifetime of red blood cells, the adducts formed with the amino acids of the protein will indicate the total exposure during this period.

Adducts may be determined by sensitive techniques such as high-performance lipid chromatography, and some immunological methods are also available. In general, the analytical methods are new, expensive and need further development and validation. Better sensitivity can be obtained by using the 32P post labelling assay, which is a nonspecific indication that DNA damage has taken place. All of these techniques are potentially useful for biological monitoring and have been applied in a growing number of studies. However, simpler and more sensitive analytical methods are needed. Given the limited specificity of some methods at low-level exposures, tobacco smoking or other factors may impact significantly on the measurement results, thus causing difficulties in interpretation.

Exposure to mutagenic compounds, or to compounds which are metabolized into mutagens, may also be determined by assessing the mutagenicity of the urine from an exposed individual. The urine sample is incubated with a strain of bacteria in which a specific point mutation is expressed in a way that can be easily measured. If mutagenic chemicals are present in the urine sample, then an increased rate of mutations will occur in the bacteria.

Exposure biomarkers must be evaluated with regard to temporal variation in exposure and the relation to different compartments. Thus, the time frame(s) represented by the biomarker, that is, the extent to which the biomarker measurement reflects past exposure(s) and/or accumulated body burden, must be determined from toxicokinetic data in order to interpret the result. In particular, the degree to which the biomarker indicates retention in specific target organs should be considered. Although blood samples are often used for biomarker studies, peripheral blood is generally not regarded as a compartment as such, although it acts as a transport medium between compartments. The degree to which the concentration in the blood reflects levels in different organs varies widely between different chemicals, and usually also depends upon the length of the exposure as well as time since exposure.

Sometimes this type of evidence is used to classify a biomarker as an indicator of (total) absorbed dose or an indicator of effective dose (i.e., the amount that has reached the target tissue). For example, exposure to a particular solvent may be evaluated from data on the actual concentration of the solvent in the blood at a particular time following the exposure. This measurement will reflect the amount of the solvent that has been absorbed into the body. Some of the absorbed amount will be exhaled due to the vapour pressure of the solvent. While circulating in the blood, the solvent will interact with various components of the body, and it will eventually become subject to breakdown by enzymes. The outcome of the metabolic processes can be assessed by determining specific mercapturic acids produced by conjugation with glutathione. The cumulative excretion of mercapturic acids may better reflect the effective dose than will the blood concentration.

Life events, such as reproduction and senescence, may affect the distribution of a chemical. The distribution of chemicals within the body is significantly affected by pregnancy, and many chemicals may pass the placental barrier, thus causing exposure of the foetus. Lactation may result in excretion of lipid-soluble chemicals, thus leading to a decreased retention in the mother along with an increased uptake by the infant. During weight loss or development of osteoporosis, stored chemicals may be released, which can then result in a renewed and protracted “endogenous” exposure of target organs. Other factors may affect individual absorption, metabolism, retention and distribution of chemical compounds, and some biomarkers of susceptibility are available (see below).

Biomarkers of Effect

A marker of effect may be an endogenous component, or a measure of the functional capacity, or some other indicator of the state or balance of the body or organ system, as affected by the exposure. Such effect markers are generally preclinical indicators of abnormalities.

These biomarkers may be specific or non-specific. The specific biomarkers are useful because they indicate a biological effect of a particular exposure, thus providing evidence that can potentially be used for preventive purposes. The non-specific biomarkers do not point to an individual cause of the effect, but they may reflect the total, integrated effect due to a mixed exposure. Both types of biomarkers may therefore be of considerable use in occupational health.

There is not a clear distinction between exposure biomarkers and effect biomarkers. For example, adduct formation could be said to reflect an effect rather than the exposure. However, effect biomarkers usually indicate changes in the functions of cells, tissues or the total body. Some researchers include gross changes, such as an increase in liver weight of exposed laboratory animals or decreased growth in children, as biomarkers of effect. For the purpose of occupational health, effect biomarkers should be restricted to those that indicate subclinical or reversible biochemical changes, such as inhibition of enzymes. The most frequently used effect biomarker is probably inhibition of cholinesterase caused by certain insecticides, that is, organophosphates and carbamates. In most cases, this effect is entirely reversible, and the enzyme inhibition reflects the total exposure to this particular group of insecticides.

Some exposures do not result in enzyme inhibition but rather in increased activity of an enzyme. This is the case with several enzymes that belong to the P450 family (see “Genetic determinants of toxic response”). They may be induced by exposures to certain solvents and polyaromatic hydrocarbons (PAHs). Since these enzymes are mainly expressed in tissues from which a biopsy may be difficult to obtain, the enzyme activity is determined indirectly in vivo by administering a compound that is metabolized by that particular enzyme, and then the breakdown product is measured in urine or plasma.

Other exposures may induce the synthesis of a protective protein in the body. The best example is probably metallothionein, which binds cadmium and promotes the excretion of this metal; cadmium exposure is one of the factors that result in increased expression of the metallothionein gene. Similar protective proteins may exist but have not yet been explored sufficiently to become accepted as biomarkers. Among the candidates for possible use as biomarkers are the so-called stress proteins, originally referred to as heat shock proteins. These proteins are generated by a range of different organisms in response to a variety of adverse exposures.

Oxidative damage may be assessed by determining the concentration of malondialdehyde in serum or the exhalation of ethane. Similarly, the urinary excretion of proteins with a small molecular weight, such as albumin, may be used as a biomarker of early kidney damage. Several parameters routinely used in clinical practice (for example, serum hormone or enzyme levels) may also be useful as biomarkers. However, many of these parameters may not be sufficiently sensitive to detect early impairment.

Another group of effect parameters relate to genotoxic effects (changes in the structure of chromosomes). Such effects may be detected by microscopy of white blood cells that undergo cell division. Serious damage to the chromosomes—chromosomal aberrations or formation of micronuclei—can be seen in a microscope. Damage may also be revealed by adding a dye to the cells during cell division. Exposure to a genotoxic agent can then be visualized as an increased exchange of the dye between the two chromatids of each chromosome (sister chromatid exchange). Chromosomal aberrations are related to an increased risk of developing cancer, but the significance of an increased rate of sister chromatid exchange is less clear.

More sophisticated assessment of genotoxicity is based on particular point mutations in somatic cells, that is, white blood cells or epithelial cells obtained from the oral mucosa. A mutation at a specific locus may make the cells capable of growing in a culture that contains a chemical that is otherwise toxic (such as 6-thioguanine). Alternatively, a specific gene product can be assessed (e.g., serum or tissue concentrations of oncoproteins encoded by particular oncogenes). Obviously, these mutations reflect the total genotoxic damage incurred and do not necessarily indicate anything about the causative exposure. These methods are not yet ready for practical use in occupational health, but rapid progress in this line of research would suggest that such methods will become available within a few years.

Biomarkers of Susceptibility

A marker of susceptibility, whether inherited or induced, is an indicator that the individual is particularly sensitive to the effect of a xenobiotic or to the effects of a group of such compounds. Most attention has been focused on genetic susceptibility, although other factors may be at least as important. Hypersusceptibility may be due to an inherited trait, the constitution of the individual, or environmental factors.

The ability to metabolize certain chemicals is variable and is genetically determined (see “Genetic determinants of toxic response”). Several relevant enzymes appear to be controlled by a single gene. For example, oxidation of foreign chemicals is mainly carried out be a family of enzymes belonging to the P450 family. Other enzymes make the metabolites more water soluble by conjugation (e.g., N-acetyltransferase and μ-glutathion-S-transferase). The activity of these enzymes is genetically controlled and varies considerably. As mentioned above, the activity can be determined by administering a small dose of a drug and then determining the amount of the metabolite in the urine. Some of the genes have now been characterized, and techniques are available to determine the genotype. Important studies suggest that a risk of developing certain cancer forms is related to the capability of metabolizing foreign compounds. Many questions still remain unanswered, thus at this time limiting the use of these potential susceptibility biomarkers in occupational health.

Other inherited traits, such as alpha1-antitrypsin deficiency or glucose-6-phosphate dehydrogenase deficiency, also result in deficient defence mechanisms in the body, thereby causing hypersusceptibility to certain exposures.

Most research related to susceptibility has dealt with genetic predisposition. Other factors play a role as well and have been partly neglected. For example, individuals with a chronic disease may be more sensitive to an occupational exposure. Also, if a disease process or previous exposure to toxic chemicals has caused some subclinical organ damage, then the capacity to withstand a new toxic exposure is likely to be less. Biochemical indicators of organ function may in this case be used as susceptibility biomarkers. Perhaps the best example regarding hypersusceptibility relates to allergic responses. If an individual has become sensitized to a particular exposure, then specific antibodies can be detected in serum. Even if the individual has not become sensitized, other current or past exposures may add to the risk of developing an adverse effect related to an occupational exposure.

A major problem is to determine the joint effect of mixed exposures at work. In addition, personal habits and drug use may result in an increased susceptibility. For example, tobacco smoke usually contains a considerable amount of cadmium. Thus, with occupational exposure to cadmium, a heavy smoker who has accumulated substantial amounts of this metal in the body will be at increased risk of developing cadmium-related kidney disease.

Application in Occupational Health

Biomarkers are extremely useful in toxicological research, and many may be applicable in biological monitoring. Nonetheless, the limitations must also be recognized. Many biomarkers have so far been studied only in laboratory animals. Toxicokinetic patterns in other species may not necessarily reflect the situation in human beings, and extrapolation may require confirmatory studies in human volunteers. Also, account must be taken of individual variations due to genetic or constitutional factors.

In some cases, exposure biomarkers may not at all be feasible (e.g., for chemicals which are short-lived in vivo). Other chemicals may be stored in, or may affect, organs which cannot be accessed by routine procedures, such as the nervous system. The route of exposure may also affect the distribution pattern and therefore also the biomarker measurement and its interpretation. For example, direct exposure of the brain via the olfactory nerve is likely to escape detection by measurement of exposure biomarkers. As to effect biomarkers, many of them are not at all specific, and the change can be due to a variety of causes, including lifestyle factors. Perhaps in particular with the susceptibility biomarkers, interpretation must be very cautious at the moment, as many uncertainties remain about the overall health significance of individual genotypes.

In occupational health, the ideal biomarker should satisfy several requirements. First of all, sample collection and analysis must be simple and reliable. For optimal analytical quality, standardization is needed, but the specific requirements vary considerably. Major areas of concern include: preparation of the in- dividual, sampling procedure and sample handling, and measurement procedure; the latter encompasses technical factors, such as calibration and quality assurance procedures, and individual- related factors, such as education and training of operators.

For documentation of analytical validity and traceability, reference materials should be based on relevant matrices and with appropriate concentrations of toxic substances or relevant metabolites at appropriate levels. For biomarkers to be used for biological monitoring or for diagnostic purposes, the responsible laboratories must have well-documented analytical procedures with defined performance characteristics, and accessible records to allow verification of the results. At the same time, nonetheless, the economics of characterizing and using reference materials to supplement quality assurance procedures in general must be considered. Thus, the achievable quality of results, and the uses to which they are put, have to be balanced against the added costs of quality assurance, including reference materials, manpower and instrumentation.

Another requirement is that the biomarker should be specific, at least under the circumstances of the study, for a particular type of exposure, with a clear-cut relationship to the degree of exposure. Otherwise, the result of the biomarker measurement may be too difficult to interpret. For proper interpretation of the measurement result of an exposure biomarker, the diagnostic validity must be known (i.e., the translation of the biomarker value into the magnitude of possible health risks). In this area, metals serve as a paradigm for biomarker research. Recent research has demonstrated the complexity and subtlety of dose-response relationships, with considerable difficulty in identifying no-effect levels and therefore also in defining tolerable exposures. However, this kind of research has also illustrated the types of investigation and the refinement that are necessary to uncover the relevant information. For most organic compounds, quantitative associations between exposures and the corresponding adverse health effects are not yet available; in many cases, even the primary target organs are not known for sure. In addition, evaluation of toxicity data and biomarker concentrations is often complicated by exposure to mixtures of substances, rather than exposure to a single compound at the time.

Before the biomarker is applied for occupational health purposes, some additional considerations are necessary. First, the biomarker must reflect a subclinical and reversible change only. Second, given that the biomarker results can be interpreted with regard to health risks, then preventive efforts should be available and should be considered realistic in case the biomarker data suggests a need to reduce the exposure. Third, the practical use of the biomarker must be generally regarded as ethically acceptable.

Industrial hygiene measurements may be compared with applicable exposure limits. Likewise, results on exposure biomarkers or effect biomarkers may be compared to biological action limits, sometimes referred to as biological exposure indices. Such limits should be based on the best advice of clinicians and scientists from appropriate disciplines, and responsible administrators as “risk managers” should then take into account relevant ethical, social, cultural and economic factors. The scientific basis should, if possible, include dose-response relationships supplemented by information on variations in susceptibility within the population at risk. In some countries, workers and members of the general public are involved in the standard-setting process and provide important input, particularly when scientific uncertainty is considerable. One of the major uncertainties is how to define an adverse health effect that should be prevented—for example, whether adduct formation as an exposure biomarker by itself represents an adverse effect (i.e., effect biomarker) that should be prevented. Difficult questions are likely to arise when deciding whether it is ethically defensible, for the same compound, to have different limits for adventitious exposure, on the one hand, and occupational exposure, on the other.

The information generated by the use of biomarkers should generally be conveyed to the individuals examined within the physician-patient relationship. Ethical concerns must in particular be considered in connection with highly experimental biomarker analyses that cannot currently be interpreted in detail in terms of actual health risks. For the general population, for example, limited guidance exists at present with regard to interpretation of exposure biomarkers other than the blood-lead concentration. Also of importance is the confidence in the data generated (i.e., whether appropriate sampling has been done, and whether sound quality assurance procedures have been utilized in the laboratory involved). An additional area of special worry relates to individual hypersusceptibility. These issues must be taken into account when providing the feedback from the study.

All sectors of society affected by, or concerned with carrying out, a biomarker study need to be involved in the decision-making process on how to handle the information generated by the study. Specific procedures to prevent or overcome inevitable ethical conflicts should be developed within the legal and social frameworks of the region or country. However, each situation represents a different set of questions and pitfalls, and no single procedure for public involvement can be developed to cover all applications of exposure biomarkers.

 

Back

Read 9564 times Last modified on Tuesday, 26 July 2022 19:31
More in this category: Genetic Toxicity Assessment »

" 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.