Sunday, 16 January 2011 19:30

The United States Approach to Risk Assessment of Reproductive Toxicants and Neurotoxic Agents

Rate this item
(0 votes)

Neurotoxicity and reproductive toxicity are important areas for risk assessment, since the nervous and reproductive systems are highly sensitive to xenobiotic effects. Many agents have been identified as toxic to these systems in humans (Barlow and Sullivan 1982; OTA 1990). Many pesticides are deliberately designed to disrupt reproduction and neurological function in target organisms, such as insects, through interference with hormonal biochemistry and neurotransmission.

It is difficult to identify substances potentially toxic to these systems for three interrelated reasons: first, these are among the most complex biological systems in humans, and animal models of reproductive and neurological function are generally acknowledged to be inadequate for representing such critical events as cognition or early embryofoetal development; second, there are no simple tests for identifying potential reproductive or neurological toxicants; and third, these systems contain multiple cell types and organs, such that no single set of mechanisms of toxicity can be used to infer dose-response relationships or predict structure-activity relationships (SAR). Moreover, it is known that the sensitivity of both the nervous and reproductive systems varies with age, and that exposures at critical periods may have much more severe effects than at other times.

Neurotoxicity Risk Assessment

Neurotoxicity is an important public health problem. As shown in table 1, there have been several episodes of human neurotoxicity involving thousands of workers and other populations exposed through industrial releases, contaminated food, water and other vectors. Occupational exposures to neurotoxins such as lead, mercury, organophosphate insecticides and chlorinated solvents are widespread throughout the world (OTA 1990; Johnson 1978).

Table 1. Selected major neurotoxicity incidents

Year(s) Location Substance Comments
400 BC Rome Lead Hippocrates recognizes lead toxicity in the mining industry.
1930s United States (Southeast) TOCP Compound often added to lubricating oils contaminates “Ginger Jake,” an alcoholic beverage; more than 5,000 paralyzed, 20,000 to 100,000 affected.
1930s Europe Apiol (with TOCP) Abortion-inducing drug containing TOCP causes 60 cases of neuropathy.
1932 United States (California) Thallium Barley laced with thallium sulphate, used as rodenticide, is stolen and used to make tortillas; 13 family members hospitalized with neurological symptoms; 6 deaths.
1937 South Africa TOCP 60 South Africans develop paralysis after using contaminated cooking oil.
1946 Tetraethyl lead More than 25 individuals suffer neurological effects after cleaning gasoline tanks.
1950s Japan (Minimata) Mercury Hundreds ingest fish and shellfish contaminated with mercury from chemical plant; 121 poisoned, 46 deaths, many infants with serious nervous system damage.
1950s France Organotin Contamination of Stallinon with triethyltin results in more than 100 deaths.
1950s Morocco Manganese 150 ore miners suffer chronic manganese intoxication involving severe neurobehavioural problems.
1950s-1970s United States AETT Component of fragrances found to be neurotoxic; withdrawn from market in 1978; human health effects unknown.
1956 Endrin 49 persons become ill after eating bakery foods prepared from flour contaminated with the insecticide endrin; convulsions result in some instances.
1956 Turkey HCB Hexachlorobenzene, a seed grain fungicide, leads to poisoning of 3,000 to 4,000; 10 per cent mortality rate.
1956-1977 Japan Clioquinol Drug used to treat travellers’ diarrhoea found to cause neuropathy; as many as 10,000 affected over two decades.
1959 Morocco TOCP Cooking oil contaminated with lubricating oil affects some 10,000 individuals.
1960 Iraq Mercury Mercury used as fungicide to treat seed grain used in bread; more than 1,000 people affected.
1964 Japan Mercury Methylmercury affects 646 people.
1968 Japan PCBs Polychlorinated biphenyls leaked into rice oil; 1,665 people affected.
1969 Japan n-Hexane 93 cases of neuropathy occur following exposure to n-hexane, used to make vinyl sandals.
1971 United States Hexachlorophene After years of bathing infants in 3 per cent hexachlorophene, the disinfectant is found to be toxic to the nervous system and other systems.
1971 Iraq Mercury Mercury used as fungicide to treat seed grain is used in bread; more than 5,000 severe poisonings, 450 hospital deaths, effects on many infants exposedprenatally not documented.
1973 United States (Ohio) MIBK Fabric production plant employees exposed to solvent; more than 80 workers suffer neuropathy, 180 have less severe effects.
1974-1975 United States (Hopewell, VA) Chlordecone (Kepone) Chemical plant employees exposed to insecticide; more than 20 suffer severe neurologicalproblems, more than 40 have less severe problems.
1976 United States (Texas) Leptophos (Phosvel) At least 9 employees suffer severe neurological problems following exposure to insecticide during manufacturing process.
1977 United States (California) Dichloropropene (Telone II) 24 individuals hospitalized after exposure to pesticide Telone following traffic accident.
1979-1980 United States (Lancaster, TX) BHMH (Lucel-7) Seven employees at plastic bathtub manufacturing plant experience serious neurologicalproblems following exposure to BHMH.
1980s United States MPTP Impurity in synthesis of illicit drug found to cause symptoms identical to those of Parkinson’s disease.
1981 Spain Contaminated toxic oil 20,000 persons poisoned by toxic substance in oil, resulting in more than 500 deaths; many suffer severe neuropathy.
1985 United States and Canada Aldicarb More than 1,000 individuals in California and other Western States and British Columbia experience neuromuscular and cardiac problems following ingestion of melons contaminated with the pesticide aldicarb.
1987 Canada Domoic acid Ingestion of mussels contaminated with domoic acid causes 129 illnesses and 2 deaths; symptoms include memory loss, disorientation and seizures.

Source: OTA 1990.

Chemicals may affect the nervous system through actions at any of several cellular targets or biochemical processes within the central or peripheral nervous system. Toxic effects on other organs may also affect the nervous system, as in the example of hepatic encephalopathy. The manifestations of neurotoxicity include effects on learning (including memory, cognition and intellectual performance), somatosensory processes (including sensation and proprioreception), motor function (including balance, gait and fine movement control), affect (including personality status and emotionality) and autonomic function (nervous control of endocrine function and internal organ systems). The toxic effects of chemicals upon the nervous system often vary in sensitivity and expression with age: during development, the central nervous system may be especially susceptible to toxic insult because of the extended process of cellular differentiation, migration, and cell-to-cell contact that takes place in humans (OTA 1990). Moreover, cytotoxic damage to the nervous system may be irreversible because neurons are not replaced after embryogenesis. While the central nervous system (CNS) is somewhat protected from contact with absorbed compounds through a system of tightly joined cells (the blood-brain barrier, composed of capillary endothelial cells that line the vasculature of the brain), toxic chemicals can gain access to the CNS by three mechanisms: solvents and lipophilic compounds can pass through cell membranes; some compounds can attach to endogenous transporter proteins that serve to supply nutrients and biomolecules to the CNS; small proteins if inhaled can be directly taken up by the olfactory nerve and transported to the brain.

US regulatory authorities

Statutory authority for regulating substances for neurotoxicity is assigned to four agencies in the United States: the Food and Drug Administration (FDA), the Environmental Protection Agency (EPA), the Occupational Safety and Health Administration (OSHA), and the Consumer Product Safety Commission (CPSC). While OSHA generally regulates occupational exposures to neurotoxic (and other) chemicals, the EPA has authority to regulate occupational and nonoccupational exposures to pesticides under the Federal Insecticide, Fungicide and Rodenticide Act (FIFRA). EPA also regulates new chemicals prior to manufacture and marketing, which obligates the agency to consider both occupational and nonoccupational risks.

Hazard identification

Agents that adversely affect the physiology, biochemistry, or structural integrity of the nervous system or nervous system function expressed behaviourally are defined as neurotoxic hazards (EPA 1993). The determination of inherent neurotoxicity is a difficult process, owing to the complexity of the nervous system and the multiple expressions of neurotoxicity. Some effects may be delayed in appearance, such as the delayed neurotoxicity of certain organophosphate insecticides. Caution and judgement are required in determining neurotoxic hazard, including consideration of the conditions of exposure, dose, duration and timing.

Hazard identification is usually based upon toxicological studies of intact organisms, in which behavioural, cognitive, motor and somatosensory function is assessed with a range of investigative tools including biochemistry, electrophysiology and morphology (Tilson and Cabe 1978; Spencer and Schaumberg 1980). The importance of careful observation of whole organism behaviour cannot be overemphasized. Hazard identification also requires evaluation of toxicity at different developmental stages, including early life (intrauterine and early neonatal) and senescence. In humans, the identification of neurotoxicity involves clinical evaluation using methods of neurological assessment of motor function, speech fluency, reflexes, sensory function, electrophysiology, neuropsychological testing, and in some cases advanced techniques of brain imaging and quantitative electroencephalography. WHO has developed and validated a neurobehavioural core test battery (NCTB), which contains probes of motor function, hand-eye coordination, reaction time, immediate memory, attention and mood. This battery has been validated internationally by a coordinated process (Johnson 1978).

Hazard identification using animals also depends upon careful observational methods. The US EPA has developed a functional observational battery as a first-tier test designed to detect and quantify major overt neurotoxic effects (Moser 1990). This approach is also incorporated in the OECD subchronic and chronic toxicity testing methods. A typical battery includes the following measures: posture; gait; mobility; general arousal and reactivity; presence or absence of tremor, convulsions, lacrimation, piloerection, salivation, excess urination or defecation, stereotypy, circling, or other bizarre behaviours. Elicited behaviours include response to handling, tail pinch, or clicks; balance, righting reflex, and hind limb grip strength. Some representative tests and agents identified with these tests are shown in table 2.

Table 2. Examples of specialized tests to measure neurotoxicity

Function Procedure Representative agents
Neuromuscular
Weakness Grip strength; swimming endurance; suspension from rod; discriminative motor function; hind limb splay n-Hexane, Methylbutylketone, Carbaryl
Incoordination Rotorod, gait measurements 3-Acetylpyridine, Ethanol
Tremor Rating scale, spectral analysis Chlordecone, Type I Pyrethroids, DDT
Myoclonia, spasms Rating scale, spectral analysis DDT, Type II Pyrethroids
Sensory
Auditory Discriminant conditioning, reflex modification Toluene, Trimethyltin
Visual toxicity Discriminant conditioning Methyl mercury
Somatosensory toxicity Discriminant conditioning Acrylamide
Pain sensitivity Discriminant conditioning (btration); functional observational battery Parathion
Olfactory toxicity Discriminant conditioning 3-Methylindole methylbromide
Learning, memory
Habituation Startle reflex Diisopropylfluorophosphate (DFP)
Classical conditioning Nictitating membrane, conditioned flavour aversion, passive avoidance, olfactory conditioning Aluminium, Carbaryl, Trimethyltin, IDPN, Trimethyltin (neonatal)
Operant or instrumental conditioning One-way avoidance, Two-way avoidance, Y-maze avoidance, Biol watermaze, Morris water maze, Radial arm maze, Delayed matching to sample, Repeated acquisition, Visual discrimination learning Chlordecone, Lead (neonatal), Hypervitaminosis A, Styrene, DFP, Trimethyltin, DFP. Carbaryl, Lead

Source: EPA 1993.

These tests may be followed by more complex assessments usually reserved for mechanistic studies rather than hazard identification. In vitro methods for neurotoxicity hazard identification are limited since they do not provide indications of effects on complex function, such as learning, but they may be very useful in defining target sites of toxicity and improving the precision of target site dose-response studies (see WHO 1986 and EPA 1993 for comprehensive discussions of principles and methods for identifying potential neurotoxicants).

Dose-response assessment

The relationship between toxicity and dose may be based upon human data when available or upon animal tests, as described above. In the United States, an uncertainty or safety factor approach is generally used for neurotoxicants. This process involves determining a “no observed adverse effect level” (NOAEL) or “lowest observed adverse effect level” (LOAEL) and then dividing this number by uncertainty or safety factors (usually multiples of 10) to allow for such considerations as incompleteness of data, potentially higher sensitivity of humans and variability of human response due to age or other host factors. The resultant number is termed the reference dose (RfD) or reference concentration (RfC). The effect occurring at the lowest dose in the most sensitive animal species and gender is generally used to determine the LOAEL or NOAEL. Conversion of animal dose to human exposure is done by standard methods of cross-species dosimetry, taking into account differences in lifespan and exposure duration.

The use of the uncertainty factor approach assumes that there is a threshold, or dose below which no adverse effect is induced. Thresholds for specific neurotoxicants may be difficult to determine experimentally; they are based upon assumptions as to mechanism of action which may or may not hold for all neurotoxicants (Silbergeld 1990).

Exposure assessment

At this stage, information is evaluated on sources, routes, doses and durations of exposure to the neurotoxicant for human populations, subpopulations or even individuals. This information may be derived from monitoring of environmental media or human sampling, or from estimates based upon standard scenarios (such as workplace conditions and job descriptions) or models of environmental fate and dispersion (see EPA 1992 for general guidelines on exposure assessment methods). In some limited cases, biological markers may be used to validate exposure inferences and estimates; however, there are relatively few usable biomarkers of neurotoxicants.

Risk characterization

The combination of hazard identification, dose-response and exposure assessment is used to develop the risk characterization. This process involves assumptions as to the extrapolation of high to low doses, extrapolation from animals to humans, and the appropriateness of threshold assumptions and use of uncertainty factors.

Reproductive Toxicology—Risk Assessment Methods

Reproductive hazards may affect multiple functional endpoints and cellular targets within humans, with consequences for the health of the affected individual and future generations. Reproductive hazards may affect the development of the reproductive system in males or females, reproductive behaviours, hormonal function, the hypothalamus and pituitary, gonads and germ cells, fertility, pregnancy and the duration of reproductive function (OTA 1985). In addition, mutagenic chemicals may also affect reproductive function by damaging the integrity of germ cells (Dixon 1985).

The nature and extent of adverse effects of chemical exposures upon reproductive function in human populations is largely unknown. Relatively little surveillance information is available on such endpoints as fertility of men or women, age of menopause in women, or sperm counts in men. However, both men and women are employed in industries where exposures to reproductive hazards may occur (OTA 1985).

This section does not recapitulate those elements common to both neurotoxicant and reproductive toxicant risk assessment, but focuses upon issues specific to reproductive toxicant risk assessment. As with neurotoxicants, authority to regulate chemicals for reproductive toxicity is placed by statute in the EPA, OSHA, the FDA and the CPSC. Of these agencies, only the EPA has a stated set of guidelines for reproductive toxicity risk assessment. In addition, the state of California has developed methods for reproductive toxicity risk assessment in response to a state law, Proposition 65 (Pease et al. 1991).

Reproductive toxicants, like neurotoxicants, may act by affecting any of a number of target organs or molecular sites of action. Their assessment has additional complexity because of the need to evaluate three distinct organisms separately and together—the male, the female and the offspring (Mattison and Thomford 1989). While an important endpoint of reproductive function is the generation of a healthy child, reproductive biology also plays a role in the health of developing and mature organisms regardless of their involvement in procreation. For instance, loss of ovulatory function through natural depletion or surgical removal of oocytes has substantial effects upon the health of women, involving changes in blood pressure, lipid metabolism and bone physiology. Changes in hormone biochemistry may affect susceptibility to cancer.

Hazard identification

The identification of a reproductive hazard may be made on the basis of human or animal data. In general, data from humans are relatively sparse, owing to the need for careful surveillance to detect alterations in reproductive function, such as sperm count or quality, ovulatory frequency and cycle length, or age at puberty. Detecting reproductive hazards through collection of information on fertility rates or data on pregnancy outcome may be confounded by the intentional suppression of fertility exercised by many couples through family-planning measures. Careful monitoring of selected populations indicates that rates of reproductive failure (miscarriage) may be very high, when biomarkers of early pregnancy are assessed (Sweeney et al. 1988).

Testing protocols using experimental animals are widely used to identify reproductive toxicants. In most of these designs, as developed in the United States by the FDA and the EPA and internationally by the OECD test guidelines program, the effects of suspect agents are detected in terms of fertility after male and/or female exposure; observation of sexual behaviours related to mating; and histopathological examination of gonads and accessory sex glands, such as mammary glands (EPA 1994). Often reproductive toxicity studies involve continuous dosing of animals for one or more generations in order to detect effects on the integrated reproductive process as well as to study effects on specific organs of reproduction. Multigenerational studies are recommended because they permit detection of effects that may be induced by exposure during the development of the reproductive system in utero. A special test protocol, the Reproductive Assessment by Continuous Breeding (RACB), has been developed in the United States by the National Toxicology Program. This test provides data on changes in the temporal spacing of pregnancies (reflecting ovulatory function), as well as number and size of litters over the entire test period. When extended to the lifetime of the female, it can yield information on early reproductive failure. Sperm measures can be added to the RACB to detect changes in male reproductive function. A special test to detect pre- or postimplantation loss is the dominant lethal test, designed to detect mutagenic effects in male spermatogenesis.

In vitro tests have also been developed as screens for reproductive (and developmental) toxicity (Heindel and Chapin 1993). These tests are generally used to supplement in vivo test results by providing more information on target site and mechanism of observed effects.

Table 3 shows the three types of endpoints in reproductive toxicity assessment—couple-mediated, female-specific and male-specific. Couple-mediated endpoints include those detectable in multigenerational and single-organism studies. They generally include assessment of offspring as well. It should be noted that fertility measurement in rodents is generally insensitive, as compared to such measurement in humans, and that adverse effects on reproductive function may well occur at lower doses than those that significantly affect fertility (EPA 1994). Male-specific endpoints can include dominant lethality tests as well as histopathological evaluation of organs and sperm, measurement of hormones, and markers of sexual development. Sperm function can also be assessed by in vitro fertilization methods to detect germ cell properties of penetration and capacitation; these tests are valuable because they are directly comparable to in vitro assessments conducted in human fertility clinics, but they do not by themselves provide dose-response information. Female-specific endpoints include, in addition to organ histopathology and hormone measurements, assessment of the sequelae of reproduction, including lactation and offspring growth.

Table 3. Endpoints in reproductive toxicology

  Couple-mediated endpoints
Multigenerational studies Other reproductive endpoints
Mating rate, time to mating (time to pregnancy1)
Pregnancy rate1
Delivery rate1
Gestation length1
Litter size (total and live)
Number of live and dead offspring (foetal death rate1)
Offspring gender1
Birth weight1
Postnatal weights1
Offspring survival1
External malformations and variations1
Offspring reproduction1
Ovulation rate

Fertilization rate
Preimplantation loss
Implantation number
Postimplantation loss1
Internal malformations and variations1
Postnatal structural and functional development1
  Male-specific endpoints
Organ weights

Visual examination and histopathology

Sperm evaluation1

Hormone levels1

Developmental
Testes, epididymides, seminal vesicles, prostate, pituitary
Testes, epididymides, seminal vesicles, prostate, pituitary
Sperm number (count) and quality (morphology, motility)
Luteinizing hormone, follicle stimulating hormone, testosterone, oestrogen, prolactin
Testis descent1, preputial separation, sperm production1, ano-genital distance, normality of external genitalia1
  Female-specific endpoints
Body weight
Organ weights
Visual examination and histopathology

Oestrous (menstrual1) cycle normality
Hormone levels1
Lactation1
Development


Senescence (menopause1)

Ovary, uterus, vagina, pituitary
Ovary, uterus, vagina, pituitary, oviduct, mammary gland
Vaginal smear cytology
LH, FSH, oestrogen, progesterone, prolactin
Offspring growth
Normality of external genitalia1, vaginal opening, vaginal smear cytology, onset of oestrus behaviour (menstruation1)
Vaginal smear cytology, ovarian histology

1 Endpoints that can be obtained relatively noninvasively with humans.

Source: EPA 1994.

In the United States, the hazard identification concludes with a qualitative evaluation of toxicity data by which chemicals are judged to have either sufficient or insufficient evidence of hazard (EPA 1994). “Sufficient” evidence includes epidemiological data providing convincing evidence of a causal relationship (or lack thereof), based upon case-control or cohort studies, or well-supported case series. Sufficient animal data may be coupled with limited human data to support a finding of a reproductive hazard: to be sufficient, the experimental studies are generally required to utilize EPA’s two-generation test guidelines, and must include a minimum of data demonstrating an adverse reproductive effect in an appropriate, well-conducted study in one test species. Limited human data may or may not be available; it is not necessary for the purposes of hazard identification. To rule out a potential reproductive hazard, the animal data must include an adequate array of endpoints from more than one study showing no adverse reproductive effect at doses minimally toxic to the animal (EPA 1994).

Dose-response assessment

As with the evaluation of neurotoxicants, the demonstration of dose-related effects is an important part of risk assessment for reproductive toxicants. Two particular difficulties in dose-response analyses arise due to complicated toxicokinetics during pregnancy, and the importance of distinguishing specific reproductive toxicity from general toxicity to the organism. Debilitated animals, or animals with substantial nonspecific toxicity (such as weight loss) may fail to ovulate or mate. Maternal toxicity can affect the viability of pregnancy or support for lactation. These effects, while evidence of toxicity, are not specific to reproduction (Kimmel et al. 1986). Assessing dose response for a specific endpoint, such as fertility, must be done in the context of an overall assessment of reproduction and development. Dose-response relationships for different effects may differ significantly, but interfere with detection. For instance, agents that reduce litter size may result in no effects upon litter weight because of reduced competition for intrauterine nutrition.

Exposure assessment

An important component of exposure assessment for reproductive risk assessment relates to information on the timing and duration of exposures. Cumulative exposure measures may be insufficiently precise, depending upon the biological process that is affected. It is known that exposures at different developmental stages in males and females can result in different outcomes in both humans and experimental animals (Gray et al. 1988). The temporal nature of spermatogenesis and ovulation also affects outcome. Effects on spermatogenesis may be reversible if exposures cease; however, oocyte toxicity is not reversible since females have a fixed set of germ cells to draw upon for ovulation (Mattison and Thomford 1989).

Risk characterization

As with neurotoxicants, the existence of a threshold is usually assumed for reproductive toxicants. However, the actions of mutagenic compounds on germ cells may be considered an exception to this general assumption. For other endpoints, an RfD or RfC is calculated as with neurotoxicants by determination of the NOAEL or LOAEL and application of appropriate uncertainty factors. The effect used for determining the NOAEL or LOAEL is the most sensitive adverse reproductive endpoint from the most appropriate or most sensitive mammalian species (EPA 1994). Uncertainty factors include consideration of interspecies and intraspecies variation, ability to define a true NOAEL, and sensitivity of the endpoint detected.

Risk characterizations should also be focused upon specific subpopulations at risk, possibly specifying males and females, pregnancy status, and age. Especially sensitive individuals, such as lactating women, women with reduced oocyte numbers or men with reduced sperm counts, and prepubertal adolescents may also be considered.

 

Back

Read 7698 times Last modified on Tuesday, 26 July 2022 19:37

" 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.