&EPA United States Environmental Protection Agency Office of Research and Development Washington DC 20460 Draft Revisions to the Guidelines for Carcinogen Risk Assessment EPA/600/BP-92/003 August 1994 External Review Draft Review Draft (Do Not Cite or Quote) Notice This document is a preliminary draft. It has not been formally released by EPA and should not at this stage be construed to represent Agency policy. It is being circulated for comment on its technical accuracy and policy implications. ------- ------- DRAFT DO NOT QUOTE OR CITE EPA/600/BP-92/003 August 1994 External Review Draft DRAFT REVISIONS TO THE GUIDELINES FOR C ARCINOGEN RISK ASSESSMENT NOTICE THIS DOCUMENT IS A PRELIMINARY DRAFT. It has not been formally released by the U.S. Environmental Protection Agency and should not at this stage be construed to represent Agency policy. It is being circulated for comment on its technical accuracy and policy implications. Office of Health and Environmental Assessment Office of Research and Development U.S. Environmental Protection Agency Washington, D.C. Printed on Recycled Paper ------- DRAFT-DO NOT QUOTE OR CITE DISCLAIMER This document is an external draft for review purposes only and does not constitute Agency policy. Mention of trade names or commercial products does not constitute endorsement or recommendation for use. 11 07/25/94 ------- Workshop on Revising the Guidelines for Carcinogen Risk Assessment AGENCY: U.S. Environmental Protection Agency. ACTION: Notice of Meeting. SUMMARY: 'This notice announces a workshop sponsored by the U.S. Environmental Protection Agency's (EPA's) Office of Health and Environmental Assessment (OHEA) and Risk Assessment Forum. OHEA and the Risk Assessment Forum will convene a panel of experts to review a draft document entitled, Draft Revisions to the Guidelines for Carcinogen Risk Assessment (External Review Draft, August 1994). Discussion will focus on the proposed revisions to, the Agency's cancer risk assessment guidelines. DATES: The workshop will be held Monday, September 12, 1994, through Wednesday, September 14, 1994. The meeting will begin at S-:30_a,,m._ ^and end at ,5:00 ,p,m. on Monday,, and Tuesday, and wi3-3- begin at 9:00 a.m. and end at noon on Wednesday. Members of the public may attend as observers. ADDRESSES: The meeting will be held at the Hyatt Regency Hotel, 1800 Presidential Street, Reston, Virginia 22090, Tel: 703/709- 1234. Eastern Research Group, Inc., an EPA contractor, is providing logistical support for the workshop. To attend the workshop as an observer, contact Eastern Research Group, Inc., 110 Hartwell Avenue, Lexington, Massachusetts 02173, Tel: 617/674-7374 by August 31, 1994. Space is limited, so please register early. FOR FURTHER INFORMATION CONTACT: For technical inquiries, contact Dr. Harry.Teitelbaum, U.S. Environmental Protection ------- Cancer Guidelines Workshop Page 2 of 3 Agency (8101), 401 M Street, S.W., Washington, DC 20460, Tel: 202/260-6743. SUPPLEMENTARY INFORMATIONS In 1986, EPA published Guidelines for Carcinogen Risk Assessment (51 FR 33992; September 24, 1986). Scientific advances in both risk assessment and carcinogenesis have led EPA and the broader scientific community to new perspectives on cancer risk assessment and related new perspectives on EPA's Cancer Risk Assessment Guidelines. As part of its efforts to update and revise the 1986 Guidelines, the Agency will hold a workshop in Reston, Virginia, on September 12-14, 1994, to discuss the major changes contemplated for these guidelines. At this meeting, experts on cancer risk assessment and its associated sciences will comment on and discuss a working draft for revising the 1986 cancer risk assessment guidelines. The workshop aims to produce recommendations to the Agency on the use of default assumptions and wider application of mechanistic data in cancer risk assessment, among other things. To obtain a single copy of the draft document (paper or Word Perfect 5.1 disk), interested parties should contact the ORD publications office by telephone or FAX, CERI, U.S. Environmental Protection Agency, 26 West Martin Luther King Drive, Cincinnati, OH 45268, Tel: (513) 569-7562, FAX (513). 569-7566. Please provide your name and mailing address, the document title: Draft Revisions to the Guidelines for Carcinogen Risk Assessment ------- Cancer Guidelines Workshop Page 3 of 3 (External Review Draft, August 1994), and EPA document number EPA/600/BP-92/003 (for paper) or EPA/600/BP-92/003a (for disk) Date T Acting Assistant Administrator for Research and Development ------- ------- DRAFT-DO NOT QUOTE OR CITE CONTENTS Authors, Contributors, and Reviewers vii 1. INTRODUCTION 1 1.1. PURPOSE AND SCOPE OF THE GUIDELINES 1 1.2. ORGANIZATION AND .APPLICATION OF THE GUIDELINES ........ 1 1.2.1. Organization i 1.2.2. Application 2 1.3. OVERVIEW OF CANCER PROCESSES 3 1.4. USE OF DEFAULT ASSUMPTIONS 8 2. HAZARD ASSESSMENT 9 2.1. OVERVIEW OF HAZARD ASSESSMENT AND CHARACTERIZATION 9 2.2. ANALYSIS OF TUMOR DATA 11 2.2.1. Human Data 11 2.2.1.1. Types of Studies 11 2.2.1.2. Study Design and Conduct 12 2.2.1.3. Analysis of Causality 15 2.2.1.4. Conclusions 16 2.2.2. Animal Data 17 2.2.2.1. Long-Term Studies 17 2.2.2.2. Special Studies 21 2.2.2.3. Significance for Human Hazard 21 2.3. .ANALYSIS OF OTHER KEY EVIDENCE , 23 2.3.1. Physicochemical Properties 23 2.3.2. Structure-Activity Relationships (SAR) 24 2.3.3. Metabolism and Pharmacokinetics 25 2.3.4. Short-Term Studies , 26 2.3.4.1. Genotoxicity Information 27 2.3.4.2. Other Short-Term Test Information 28 2.3.4.3. Biomarker Information 30 2.3.4.4. Confidence in Conclusions 30 2.4. MODE OF ACTION-IMPLICATIONS FOR HAZARD CHARACTERIZATION AND DOSE-RESPONSE 31 2.5. HAZARD CHARACTERIZATION 33 2.5.1. Carcinogenic Potential for Humans 33 in 07/25/94 ------- r DRAFT-DO NOT QUOTE OR CITE CONTENTS (continued) 2.5.2. Conditions of Expression 34 2.5.3. Descriptions of Weight of Evidence 35 2.5.4. Hazard Narrative 37 3. DOSE-RESPONSE ASSESSMENT 38 3.1. RESPONSE DATA 39 3.2. DOSE DATA 41 3.2.1. Basic Analyses 42 3.2.2. Pharmacokinetic Analyses 43 3.2.3. Additional Considerations for Dose in Human Studies 44 3.3. SELECTION OF QUANTITATIVE APPROACH 45 3.3.1. Analysis in the Range of Observation 45 3.3.2. Extrapolation 46 3.3.3. Issues for Analysis of Human Studies 48 3.3.4. Use of Toxicity Equivalence Factors and Relative Potency Estimates . 49 3.4. DOSE-RESPONSE CHARACTERIZATION 50 4. EXPOSURE ASSESSMENT AND CHARACTERIZATION 52 5. RISK CHARACTERIZATION 53 5.1. PURPOSE 53 5.2. APPLICATION 54 5.3. CONTENT 54 5.3.1. Presentation 54 5.3.2. Strengths and Weaknesses 55 REFERENCES 56 APPENDIX A 68 IV 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE AUTHORS, CONTRIBUTORS, AND REVIEWERS This external review draft was prepared by a Technical Panel of the Risk Assessment Forum, Office of Research and Development (ORD). AUTHORS Jeanette Wiltse Richard Hill Vanessa Vu Arnold Kuzmack ORD/Office of Health and Environmental Assessment Office of Prevention, Pesticides, and Toxic Substances Office of Prevention, Pesticides, and Toxic Substances Office of Water CONTRIBUTORS AND REVIEWERS To be provided in the next draft. 07/25/94 ------- ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE 1. INTRODUCTION 1.1. PURPOSE AND SCOPE OF THE GUIDELINES These guidelines revise and replace United States Environmental Protection Agency (EPA) Guidelines for Carcinogen Risk Assessment published in 51 FR 33992, September 24, 1986. The guidelines provide EPA staff and decision makers with guidance and perspectives necessary to develop and use risk assessments. They also provide basic information to the public about the Agency's risk assessment methods. The guidelines encourage both consistency of procedures to support consistency in scientific components of Agency decision making and innovation to remain up-to-date in scientific thinking. In balancing these goals, the Agency relies on input from the general scientific community through established scientific peer review processes. The guidelines incorporate basic principles and science policies based on evaluation of the currently available information. As more is discovered about carcinogenesis, the need will arise to make appropriate changes hi risk assessment guidance. The Agency will revise these guidelines when extensive changes are due. In the interim, the Agency will issue special reports, after appropriate peer review, to supplement and update guidance on single topics, (e.g., U.S. EPA, 1991b) 1.2. ORGANIZATION AND APPLICATION OF THE GUIDELINES 1.2.1. Organization Publications of the Office of Science and Technology Policy (OSTP, 1985) and the National Research Council (NRC, 1983 and 1994) provide information and general principles about risk assessment. Risk assessment utilizes available scientific information bearing on the properties of an agent and its effects hi biological systems to provide an evaluation of the potential for harm as a consequence of environmental exposure to the agent. Risk assessment is one of the scientific analyses available for consideration, with other analyses, in decision making on environmental protection. The 1983 and 1994 NRC documents 07/25/94 ------- I 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE organize risk assessment information into four areas: hazard identification, dose-response assessment, exposure assessment, and risk characterization. This structure appears in these guidelines which additionally emphasize characterization of evidence and conclusions in each part of the assessment. The risk assessment questions addressed in these guidelines are: For hazard-Can the agent1 present a carcinogenic hazard to humans, and, if so, under what circumstances? For dose-responseAt what levels of exposure might effects occur? For exposure-What are the conditions of human exposure? For riskWhat is the character of the risk? How well do data support conclusions about the nature and extent of the risk? 1.2.2. Application The guidelines apply within the framework of policies provided by applicable EPA statutes and do not alter such policies. The guidelines cover assessment of available data. They do not imply that one land of data or another is prerequisite for regulatory action concerning any agent. Risk management applies directives of regulatory legislation, which may require consideration of potential risk, or solely hazard or exposure potential, along with social, economic, technical, and other factors in decision making. Risk assessments support decisions, but, to maintain their integrity as decision-making tools, are not influenced by consideration of the social or economic consequences of regulatory action. Not every EPA assessment has the same scope or depth. Agency staff often conduct screening-level assessments for priority setting or separate assessments of hazard or exposure for ranking purposes or to decide whether to invest resources in collecting data for a full assessment. Moreover, a given assessment of hazard and dose-response may be used with more than one exposure assessment that may be conducted separately and at different times as the need arises hi studying environmental problems hi various media. The guidelines 1The term "agent" refers generally to any chemical substance, mixture, or physical or biological entity being assessed, unless otherwise noted. 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 DRAFT-DO NOT QUOTE OR CITE apply to these various situations, in appropriate detail given the scope and depth of the particular assessment. When adopted by the Agency, the guidelines will apply prospectively to new assessments and to revisions of previous assessments prompted by new data that may alter previous conclusions. 1.3. OVERVIEW OF CANCER PROCESSES The following discussion provides background for considering different ways in which agents may be factors in carcinogenicity. The picture will change as research reveals more about carcinogenic processes. Nevertheless, it is apparent that several general modes of action are being elucidated from direct reaction with DNA to hormonal or other growth signaling processes. While the exact mechanism of action of an agent at the molecular level may not be clear from existing data, the available data will often provide support for deducing the general mode of action. Under these guidelines, using all of the data to arrive at a view of the mode of action supports both characterization of human hazard potential and assessment of dose-response relationships. Cancers are diseases of somatic mutation affecting growth and differentiation of cells. The genes that control cell growth, programmed cell death and cell differentiation are critical to normal development of tissues from embryo to adult metazoan organisms. These genes continue to be critical to maintenance of form and function of tissues in the adult (e.g., Meyn, 1993), and changes in them are essential elements of carcinogenesis (Hsu et al., 1991; Kakizuka et al., 1991; Bottaro et al., 1991; Kaplan et al., 1991; Sidransky et al., 1991; Salomon et al., 1990; Srivastava et al., 1990). The genes involved are among the most highly conserved in evolution as evidenced by the great homology of many of them in DNA sequence and function in organisms as phylogenetically distant as worms, insects, and mammals (Auger et al., 1989a, b; Kaplan, et al. 1991; Hollstein et al., 1991; Herschman, 1991; Strausfeld et al., 1991; Forsburg and Nurse, 1991). 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 Adult tissues, even those composed of rapidly replicating cells, maintain a constant 2 size and cell number (Nunez et al., 1991) by balancing three cell fates: (1) continued 3 replication; (2) differentiation to take on specialized functions; or (3) programmed cell death 4 (apoptosis) (Raff, 1992; Mailer, 1991; Naeve et al., 1991; Schneider et al., 1991; Harris, 5 1990). Neoplastic growth through clonal expansion can result from somatic mutations that 6 inactivate control over cell fate (Kakizuka et al., 1991; deThe et al., 1991; Sidransky et al., 7 1992; Nowell, 1976). ^ 8 Cancers may also be thought of as diseases of the cell cycle. For example, genetic 9 diseases that cause failure of cells to repair DNA damage prior to cell replication predispose 10 people to cancer. These changes are also frequently found in tumor cells in sporadic 11 cancers. These changes appear to be particularly involved at points in cell replication called 12 "checkpoints" where DNA synthesis or mitosis is normally stopped until DNA damage is 13 repaired or cell death induced (Tobey, 1975). A cell that bypasses a checkpoint may acquire 14 a heritable growth advantage. Similar effects on the cell cycle occur when mitogens such as 15 hormones or growth factors stimulate cell growth. Rapid replication in response to tissue 16 injury may also lead to unrepaired DNA damage that is a risk factor for carcinogenesis. 17 Normally a cell's fate is determined by a timed sequence of biochemical signals. 18 Signal transduction in the cell involves chemical signals that bind to receptors, generating 19 further signals in a pathway whose target in many cases is control of transcription of a 20 specific set of genes (Hunter, 1991; Cantley et al., 1991; Collum and Alt, 1990). Cells are 21 subject to growth signals from the same and distant tissues, e.g., endocrine tissues (Schuller, 22 1991) . In addition to hormones produced by endocrine tissues, numerous soluble 23 polypeptide growth factors have been identified that control normal growth and 24 differentiation (Cross and Dexter, 1991; Wellstein et al., 1990). The cells responsive to a 25 particular growth factor are those that express transmembrane receptors that specifically bind 26 the growth factor. 27 Solid tumors develop in stages operationally defined as initiation, promotion, and 28 progression (see, for example, Pilot and Dragan, 1991). These terms, which were coined in 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 the context of specific experimental designs, are used for convenience in discussing concepts, 2 but they refer to complex events that are not completely understood. During initiation, the 3 cell acquires a genetic change that confers a potential growth advantage. During promotion, 4 clpnal expansion of this altered cell occurs. Later, during progression, a series of genetic 5 and other biological events both enhance the growth advantage of the cells and enlist normal 6 host processes to support tumor development, and cells develop the ability to invade locally 7 and metastasize distally, taking on the characteristics of malignancy. Many endogenous and 8 exogenous factors are known to participate in the process as a whole. These include specific 9 genetic predispositions or variations in ability to detoxify agents, medical history (Harris, 10 1989; Nebreda et al., 1991), infections, chemicals, ionizing radiation, hormones and growth 11 factors, and immune suppression. Several such risk factors likely work together to cause 12 individual human cancers. 13 A cell that has been transformed, acquiring the potential to establish a line of cells 14 that grow to a tumor, will probably realize that potential only rarely. The process of 15 tumorigenesis in animals and humans is a multistep one (Bouk, 1990; Fearon and Vogelstein, 16 1990; Hunter, 1991; Kumar et al., 1990; Sukumar, 1989; Sukumar, 1990), and normal 17 physiological processes appear to be arrayed against uncontrolled growth of a transformed 18 cell (Weinberg, 1989). Powerful inhibition by signals from contact with neighboring normal 19 cells is one known barrier (Zhang et al., 1992). Another is the immune system (at least for 20 viral infection). How a cell with tumorigenic potential acquires additional properties that are 21 necessary to enable it to overcome these and other inhibitory processes is a subject of 22 ongoing research. For known human carcinogens studied thus far, there is an often decades- 23 long latency between exposure to carcinogenic agents and development of tumors (Fidler and 24 Radinsky, 1990; Tanaka et al., 1991; Thompson et al., 1989). This latency is also typical of 25 development of tumors in individuals with genetic diseases that make them cancer prone 26 (Meyn, 1993; Srivastava et al., 1990). 27 The importance of genetic mutation in the carcinogenic process calls for special 28 attention to assessing agents that cause such mutations. Much of the screening and testing of 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 agents for carcinogenic potential has been driven by the idea of identifying this mode of 2 action. Cognizance of and emphasis on other modes of action such as ones that act at the 3 level of growth signalling within or between cells, through cell receptors, or that indirectly 4 cause genetic change, comes from more recent research. There are not yet standardized tests 5 for many modes of action, but pertinent information may be available in individual cases. 6 Agents of differing characteristics influence cancer development: inorganic and 7 organic, naturally occurring and synthetic, of inanimate or animate origin, endogenous or 8 exogenous, dietary and non-dietary. The means by which these agents act to influence 9 carcinogenesis are variable, and reasoned hazard assessment requires consideration of the 10 multiple ways chemicals influence cells in experimental systems and in humans. Agents 11 exert genotoxic effects either by interacting directly with DNA or by indirect means through 12 intermediary substances (e.g., reactive oxygen species) or processes. Most DNA interactive 13 chemicals are electrophilic or can become electrophilic when metabolically activated. 14 Electrophilic molecules may bind covalentiy to DNA to form adducts and this may lead to 15 depurination, depyrimidation or produce DNA strand breaks; such lesions can be converted 16 to mutations with a round of DNA synthesis and cell division. Other DNA-interactive 17 chemicals may cause the same result by intercalation into the DNA helix. Still other 18 chemicals may methylate DNA, changing gene expression. Non-DNA-reactive chemicals 19 produce genotoxic effects by many different processes. They may affect spindle formation 20 or chromosome proteins, interfere with normal growth control mechanisms, or affect 21 enzymes involved with ensuring the fidelity of DNA synthesis (e.g., topoisomerase), 22 recombination, or repair. 23 The "classical" chemical carcinogens in laboratory rodent studies are agents that 24 consistently produce gene mutations and structural chromosome aberrations in short-term 25 tests. A large data base reveals that these mutagenic substances commonly produce tumors 26 at multiple sites and in multiple species (Ashby and Tennant, 1991). Most of the carcinogens 27 identified in human studies, aside from hormones, are also gene or structural chromosome 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 mutagens (Tennant and Ashby, 1991). Most of these compounds or their metabolites contain 2 electrophilic moieties that react with DMA. 3 Numerical chromosome aberrations, gene amplification and the loss of gene 4 heterozygosity are also found in animal and human tumor cells that may arise from initiating 5 events or during progression. There is reason to believe that accumulation of additional 6 genetic changes is favored by selection in the evolution of tumor cells because they confer 7 additional growth advantages. Exogenous agents may function at any stage of carcinogenesis 8 (Barrett, 1993). Some aberrations may arise as a consequence of genomic instability arising 9 from tumor suppressor gene mutation, e.g., p53 (Harris and Hollstein, 1993). The frequent 10 observation in tumor cells that both of a pair of homologous chromosomes have identical 11 mutation spectra in tumor suppressor genes suggests an ongoing, endogenous process of gene 12 conversion. Currently, there is a paucity of routine test methods to screen for events such as 13 gene conversion or gene amplification, and knowledge regarding the ability of particular 14 agents of environmental interest to induce them is, for the most part, wanting. Work is 15 underway to characterize, measure, and evaluate their significance (Bianchi et al., 1991; 16 Cowell et al., 1991; see Schwab, 1992; Travis et al., 1991). 17 Several kinds of mechanistic studies aid in risk assessment. Comparison of DNA 18 lesions in tumor cells taken from humans to the lesions that a tumorigenic agent causes in 19 experimental systems, can permit inferences about the association of exposure to the agent 20 and an observed human effect (Vahakangas et al., 1992; Hollstein et al., 1991; Hayward et 21 al., 1991). An agent that is observed to cause mutations experimentally may be inferred to 22 have potential for carcinogenic activity (U.S. EPA, 1991a). If such an agent is shown to be 23 carcinogenic in animals, the inference that its mode of action is through mutagenicity is 24 strong. A carcinogenic agent that is not mutagenic in experimental systems, but is mitogenic 25 or affects hormonal levels or causes toxic injury followed by compensatory growth may be 26 inferred to have effects on growth signal transduction or to have secondary carcinogenic 27 effects. The strength of these inferences depends in each case on the nature and extent of all 28 the available data. 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 Differing modes of action at the molecular level have different dose-response 2 implications for the activity of agents. The carcinogenic activity of a direct-acting mutagen 3 should be a function of the probability of its reaching and reacting with DNA. The activity 4 of an agent that interferes at the level of signal pathways with many potential receptor targets 5 should be a function of multiple reactions. The activity of an agent that acts by causing 6 toxicity followed by compensatory growth should be a function of the toxicity. 7 8 1.4. USE OF DEFAULT ASSUMPTIONS 9 The National Research Council (NRC), in its 1983 report on the science of risk 10 assessment (NRC, 1983), recognized that default assumptions are necessarily made in risk 11 assessments when gaps in general knowledge or in data available for a particular agent are 12 encountered. These default assumptions are inferences based on general scientific knowledge 13 of the phenomena in question and are also matters of policy. In its 1994 report on risk 14 assessment, the NRC supported continued use of default assumptions (NRC, 1994). The 15 1994 report recommended that the EPA explain the science and policy underlying the 16 defaults and provide general criteria for departing from them. (Descriptions of major 17 defaults and their rationales appear in the responses to comments accompanying this 18 document. NOTE: the responses will appear with the Federal Register proposal of revisions 19 and do not accompany this review draft.) 20 Under these guidelines, it is the policy of the EPA to continue to use default 21 assumptions. Instead of considering them to be positions from which departure is justified, it 22 is the policy of these guidelines that default assumptions are indeed default positions, and that 23 they are not used unless supported by the circumstances of a particular case. In each case in 24 which the risk assessor decides that it is appropriate to use a default, the use is explained and 25 the lack or inadequacy of data justifying resort to a default is described. 26 Throughout these guidelines, major defaults are noted in the context of risk 27 assessment subjects. Most major defaults covered in the guidelines arise as a consequence of 28 either using animal toxicity data as a model for human toxicity or in the extrapolation of 8 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE observed dose-response relationships to lower dose levels associated with environmental - exposures of interest. Other default assumptions may be applied in practice that are not within the scope of coverage of the guidelines. When these are applied, they are similarly explained. An example of such a default is use of an exposure factor such as food or water consumption, typical of an adult in the United States. Such factors are often taken from published compendia that contain numerous exposure factors based on general population data. These factors are used to apply to specific populations. 2. HAZARD ASSESSMENT 2.1. OVERVIEW OF HAZARD ASSESSMENT AND CHARACTERIZATION Hazard Assessment The purpose of hazard assessment is to review and evaluate data pertinent to two questions: (1) whether an agent may pose a carcinogenic hazard to human beings, and (2) under what circumstances an identified hazard may be expressed. The ingredients of hazard assessment are reviews of a variety of kinds of data that may range from observations of tumors to analysis of structure-activity relationships. The purpose of the assessment is not simply to assemble these separate reviews; its purpose is to construct a total case analysis examining the biological story the data reveal as a whole about carcinogenic effects, mode of action of the agent, and implications of these for human hazard and dose-response evaluation. For example, interpretation of studies on human effects considers structure-activity relationships and experimental data on animal effects. Similarly, interpretation of animal tumors considers other relevant data such as effects at the cellular level, comparative metabolism, and non-cancer toxicity. The reliability of conclusions comes from the combined strength and coherence of inferences appropriately drawn from all of the available evidence. If studies reveal that an agent affects carcinogenic processes under one set of circumstances, the next question is whether one can infer that the effects will occur under 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 other or all circumstances. For instance, are effects seen in one species likely to occur in 2 another species? Are effects that occur by one route and level of exposure likely to occur by 3 another route and level? It would take an extraordinary amount of work to empirically 4 address every question involved in these extrapolations. Realistically, hazard assessments 5 must address these issues with the data that are practicably obtainable, combined with 6 inferences drawn from a basic understanding of biological and chemical processes relevant to 7 carcinogenesis and to exposure. Extrapolation of responses from one species to another and 8 from high to low dose can be much stronger if there is information on the mode of action of 9 the agent. Such information can enable greater concentration on particular lines of evidence. 10 To the extent data permit, hazard assessment addresses the mode of action question both as a 11 part of identifying human hazard potential and as an initial step in considering appropriate 12 approaches to dose-response assessment. 13 14 Hazard Characterization 15 The hazard characterization develops conclusions on the questions of human hazard 16 potential and circumstances of its expression. Developing conclusions involves integrating 17 the salient outcomes of analyses of all of the data. This, in turn, involves considering the 18 inferences drawn from data, their strengths and weaknesses in light of the uncertainties of the 19 available data and the state of current scientific knowledge. Presentation of the results of 20 hazard characterization includes: 21 summarizing the evaluations of hazard data, 22 expressing the reasoning from the data to conclusions, and 23 explaining significant strengths or limitations of the conclusions given the limits of 24 available data and knowledge. 25 The discussions that follow cover analysis of tumor data, both animal and human, and 26 analysis of other key evidence about properties and effects that relate to carcinogenic 27 potential. The'discussion covers aspects of evidence pertinent to mode of action and how 28 mode of action can influence thinking about the dose-response relationship. The last part of 10 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 DRAFT-DO NOT QUOTE OR CITE Section 2 describes hazard characterization. It discusses the characterization, how the likelihood of carcinogenic hazard is categorized, and the use of a hazard narrative as a summary. 2.2. ANALYSIS OF TUMOR DATA Evidence of carcinogenicity comes from finding tumor increases in humans or in laboratory animals exposed to a given agent or from finding tumors following exposure to structural analogues to the compound under review. The significance of the observed or anticipated tumor effects is evaluated in reference to all of the other key evidence on the agent. This section contains guidance for analyzing human and animal studies to decide whether there is an association between exposure to an agent or a structural analogue and occurrence of tumors. (The use of the term "tumor" here is generic, meaning malignant neoplasms or a combination of malignant and corresponding benign neoplasms.) 2.2.1. Human Data 2.2.1.1. Types of Studies Human data may come from studies that select and compare exposed and unexposed (or less-exposed) populations or from investigations of cases in specific settings, e.g., workplace or clinical. Formal studies include cohort, case-control, proportionate ratio, clinical trial, and correlational studies. Each has well known strengths and weaknesses that affect interpretation of results (Kelsey et al., 1986; Lilienfeld and Lilienfeld, 1979; Mausner and Kramer, 1985; Rothman, 1986). Reports of cancer cases can identify associations particularly when there are unique features such as association with an uncommon tumor (e.g., vinyl chloride and angiosarcoma or diethylstilbestrol and clear cell carcinoma of the vagina). 11 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 2.2.1.2. Study Design and Conduct 2 In cancer epidemiologic studies, unlike laboratory experiments, the exposures of the 3 study and the control (referent) populations are not controlled. Because most studies are 4 retrospective, and cancer effects are latent for many years, the exposure estimates are usually 5 a reconstruction of what exposures may have been many years in the past. Moreover, the 6 fact that most human cancers have more than one potential cause creates both design and 7 analytical problems of distinguishing among possible causes for an observed effect. These 8 features mean that drawing causal inferences depends on an analysis of many factors. The 9 following discussions cover factors included in an analysis of human data: 10 11 Population Issues 12 The ideal comparison would be between two populations that differ only in exposure 13 to the agent in question. Since this is seldom the case, it is important to identify any sources 14 of bias that are inherent in a study's design or data collection methods. Bias can arise from 15 several sources, including non-comparability between populations of factors such as general 16 health (McMichael, 1976), diet, lifestyle or geographic location; differences in the way case 17 and control individuals recall past events; differences in data collection that result in unequal 18 ascertainment of health effects in the populations; and unequal follow-up of individuals. 19 Both acceptance of studies for assessment and judgment of their strength or weakness 20 depends on identifying sources of bias and their effects on results. 21 22 Exposure Issues 23 Questions to address about exposure are: What can one reliably conclude about the 24 level, duration, route, and frequency of exposure of individuals in one population as 25 compared to another? How sensitive are study results to uncertainties in these parameters? 26 Surrogates are often used to reconstruct exposure parameters when historical 27 measurements are not available. These may take the form of attributing exposures to job 28 classifications in a workplace or to broader occupational or geographic groupings. Use of 12 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE surrogates carries a potential for misclassification in that individuals may be counted in the incorrect exposure groups. Misclassification generally leads to reduced ability of a study to detect differences between study and referent populations. When either current or historical monitoring data are available, the exposure evaluation includes consideration of the error bounds of the monitoring and analytic methods and whether the data are from routine or accidental exposures. The potentials for misclassification and measurement errors are amenable to both qualitative and quantitative analysis. These are essential analyses for judging a study's results since exposure estimation is the most critical, but weakest, part of a retrospective study. Biological markers of exposure or effect potentially offer excellent measures of exposure (Hulka and Margolin, 1992; Peto and Darby, 1994). Validated markers of exposure such as alkylated hemoglobin from exposure to ethylene oxide (Callemen et al., 1986; van Sittert et al., 1985) or urinary arsenic (Enterline et al., 1987), can greatly improve estimates of dose. Markers closely identified with effects promise to greatly increase the ability of studies to distinguish real effects from bias at low levels of relative risk between populations (Taylor et al., 1994; Biggs et al., 1993) and to resolve problems of confounding risk factors. In deciding whether there is an association between health effects and exposure to an agent, the evaluation gives studies with more precise and specific exposure estimates greater weight. Confounding Factors A confounding variable is a risk factor, independent of the putative agent, that is distributed unequally among the exposed and unexposed populations. Adjustment for possible confounding factors can occur either in the design of the study (e.g., matching on critical factors) or in the statistical analysis of the results. The latter may not be possible due to the presentation of the data or because needed information was not collected during the study. In this case, indirect comparisons may be possible. For example, in the absence of 13 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 data on smoking status among individuals in the study population, an examination of the 2 possible contribution of cigarette smoking to increased lung cancer risk may be based on 3 information from other sources such as the American Cancer Society's longitudinal studies 4 (Hammand, 1966; Garfmkel and Silverberg, 1991). The effectiveness of adjustments 5 contributes the ability to draw inferences from a study. 6 Different studies involving exposure to an agent may have different confounding 7 factors. If consistent increases in cancer risk are observed across a collection of studies with 8 randomly distributed confounding factors, the inference that the agent under investigation 9 was the etiologic factor is strong, even though complete adjustment for confounding factors 10 cannot be made and no single study supports a strong inference. 11 Biological evidence on confounding factors, such as animal data, structure-activity 12 relationships, and metabolism data, is analyzed. The analysis compares such evidence on the 13 agent under study with evidence on identified confounders to test the plausibility of 14 associating observed human effects either with the agent or with confounding factors. 15 16 Sensitivity 17 Sensitivity, or the ability of a study to detect real effects, is a function of several 18 factors. Greater size of the study population(s) (sample size) increases sensitivity as does 19 greater exposure (levels and duration) of the population members. Because of the often 20 lengthy latency period in cancer development, sensitivity also depends on whether adequate 21 time has elapsed since exposure began for effects to occur. A unique feature that can be 22 ascribed to effects of a particular agent (such as a tumor type that is seen only rarely in the 23 absence of the agent) can increase sensitivity by permitting separation of bias and 24 confounding factors from real effects. Similarly, a biomarker particular to the agent can 25 permit these distinctions. These are all factors to explore in statistical analysis of the data. 26 14 07/25/94 . ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE 2.2.1.3. Analysis of Causality Statistical Considerations The analysis applies appropriate statistical methods to ascertain whether or not there is any significant association between exposure and effects. A description of the method or methods includes the reasons for their selection. Statistical analyses of the potential effects of bias or confounding factors are part of addressing the significance of an association, or lack of one, and whether a study is able to detect any effect. The analysis augments examination of the results for the whole population with exploration of the results for groups with comparatively greater exposure or time since first exposure. This may support identifying an association or establishing a dose-response trend. When studies show no association, such exploration may apply to determining an upper limit on potential human risk for consideration alongside results of animal tumor effects studies. Statistical methods for examining several studies in combination are commonly termed "meta-analysis." Meta-analysis evaluates whether study results differ randomly from the hypothesis of no effect (Mann, 1990). If an effect is not present, the observed results ought to appear to be randomly distributed and cancel each other when studies are combined. Meta-analysis is an appropriate tool to use if several studies are available. Before applying meta-analysis, several issues are addressed to decide if the analysis will be meaningful. These include: controlling for bias and confounding prior to combining studies; establishing and meeting appropriate criteria for study inclusion; assigning weights to individual studies; arid anticipating possible publication and aggregation bias (Greenland, 1987; Peto, 1992). Criteria for Causality A causal interpretation is enhanced for studies to the extent that they meet the criteria described below. None of the criteria is conclusive by itself, and the only criterion that is essential is the temporal relationship. These criteria are modelled after those developed by Bradford Hill in the examination of cigarette smoking and lung cancer (Rothman, 1986), and they need to be interpreted in the light of all other information on the agent being assessed. 15 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 Temporal relationship: The disease occurs within a biologically reasonable time after 2 initial exposure. This feature must be present if causality is to be considered. The 3 initial period of exposure to the agent is the accepted starting point in most 4 epidemiologic studies. 5 Consistency: Associations occur in several independent studies of a similar exposure 6 in different populations, or associations occur consistently for different subgroups in 7 the same study. 8 Magnitude of the association: A causal relationship is more credible when the risk 9 estimate is large and precise (narrow confidence intervals). 10 Biological gradient: The risk ratio increases with increasing exposure or dose. A 11 strong dose-response relationship across several categories of exposure, latency, and 12 duration is supportive for causality given that confounding is unlikely to be correlated 13 with exposure. The absence of a dose-response relationship is not by itself evidence 14 against a causal relationship. 15 Specificity of the association: The likelihood of a causal interpretation is increased if 16 an exposure produces a specific effect (one or more tumor types also found in other 17 studies) or if a given effect has a unique exposure. 18 Biological plausibility: The association makes sense in terms of biological 19 knowledge. Information is considered from animal toxicology, pharmacokinetics, 20 structure-activity relationship analysis, and short-term studies of the agent's influence 21 on events in the carcinogenic process considered. 22 Coherence: The cause-and-effect interpretation is in logical agreement with what is 23 known about the natural history and biology of the disease, i.e., the entire body of 24 knowledge about the agent. 25 26 2.2.1.4. Conclusions 27 Critical evaluation of each human study includes the exposure-effect relationship, 28 exposure assessment, selection and comparison of groups, sample size, and handling of 16 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE latency, confounders, and bias as discussed under section 2.2.1.2. The weight of a study in providing inferences about human carcinogenicity depends upon this evaluation, the statistical confidence in conclusions, and biological plausibility of findings. Greater plausibility and stronger inferences come from meeting criteria for causality. The plausibility of an exposure-effect finding can be increased or decreased by other relevant findings such as evidence of structure-activity relationships with other agents, studies of mode of action, understanding of metabolic pathways, and animal toxicology findings. The above considerations of the studies available are integrated in a summary of results. 2.2.2. Animal Data Various kinds of whole animal test systems are currently used or are under development for evaluating potential carcinogenicity. Standardized, long-term studies involving chronic exposure for most of the lifespan of an animal are generally accepted for evaluation of tumor effects (Tomatis et al., 1989; Rail, 1991; Allen et al., 1988; but see Ames and Gold, 1990). Other studies of special design are useful for observing formation of preneoplastic lesions or assaying agents for specific modes of action. 2.2.2.1. Long-Term Studies Current standardized long-term studies in rodents test at least 50 animals in each of three treatment group and in the control group usually for 2 years (US EPA, 1983a,b,c). Other, similar protocols have been and continue to be used by many laboratories. Studies are examined for production of tumors and preneoplastic lesions, as well as for toxicity reactions that may give clues to the mode of action of the agent in producing tumors. Analyses of study results are by dose, sex, species and route of exposure. Analysis of toxicity covers both its potential role in carcinogenicity and its separate potential for causing^ effects in humans. The assessment may identify general toxicity as a more important end point for consideration than carcinogenicity. 17 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 All available studies of tumor effects in whole animals are considered, at least 2 preliminarily. The analysis discards studies judged to be wholly inadequate in protocol or 3 conduct and carries forward the remainder. Studies for analysis include both those that are 4 optimal in terms of current protocols and conduct, and studies whose adequacy is limited in 5 some respect. Limitations may be in aspects such as duration, dosing, or sensitivity. Such 6 limited studies can contribute as their deficiencies permit. 7 Evaluation of tumor effects includes both biological significance and statistical 8 significance (Haseman, 1984, 1985, 1990). 9 10 Statistical Significance of Animal Responses 11 Statistical analysis of a long-term study considers each tumor type separately. Benign 12 and malignant lesions of the same cell type, usually within a single tissue or organ, are 13 combined (McConnell, 1986). Trend tests and pairwise comparison tests are the 14 recommended tests for determining whether chance, rather than a treatment-related effect, is 15 a plausible explanation for an apparent increase in tumor incidence. A trend test such as the 16 Cochran-Armitage test (Snedecor and Cochran, 1967) asks whether the results in all dose 17 groups together increase as dose increases. By convention, a trend is statistically significant 18 if the probability that an effect seen would have occurred by chance is 5% or less 19 (p<0.05). A pairwise comparison test such as the Fisher exact test (Fisher, 1932) asks 20 whether an incidence in one dose group is increased over the control group. Again, by 21 convention, a statistically significant comparison is one for which p<0.05 that the increased 22 incidence is due to chance. Significance in either kind of test is sufficient to reject the 23 hypothesis that chance accounts for the result. A statistically significant response may or 24 may not be biologically significant and vice versa. 25 The standard comparison is tumor effect in dosed animals as compared with 26 concurrent control animals. Additional insights about both statistical and biological 27 significance can come from an examination of historical control data (Haseman et al., 1984). 28 If such data are used, the discussion needs to address several issues that affect comparability 18 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE of historical and concurrent control data. Among these issues are the following: genetic drift in the laboratory strains; differences in pathology examination at different times and in different laboratories (e.g., in criteria for evaluating lesions; variations in the techniques for preparation or reading of tissue samples among laboratories); comparability of animals from different suppliers. It is most desirable to compare historical and current data from the same laboratory and supplier, gathered closely in time. Always, the discussion of results covers issues of comparability. Historical control data can add to the analysis particularly by enabling identification of uncommon tumor types or high spontaneous incidence of a tumor in a given strain. Identification of common or uncommon situations prompts further thought about the meaning of the response in the current study in context with other observations in animal studies and with other evidence about the carcinogenic potential of the agent. These other sources of information may reinforce or weaken the significance given to the response in the hazard assessment. Evaluation of Animal Responses Observation of tumor effects under varying circumstances (e.g., test species or strain, route of administration, sex, replication in different laboratories) lends support to significance of findings for animal carcinogenicity. Progression of lesions observed in subchronic studies or in the chronic study, greater ratio of malignant to benign tumors, dose- response trend, or reduced latency of neoplastic lesions are among the findings that also lend support. Finding a very uncommon tumor type suggests biological significance even if the increase in incidence is not statistically significant, particularly if the increase is dose-related. The high dose in long-term studies is generally selected to provide maximum statistical ability to detect treatment-related effects. Two or more additional doses, reasonably spaced, are at some fraction of the high dose. The high dose, in EPA requirements, is one that, in an animal lifetime, produces some toxic effects without either unduly affecting mortality from effects other than cancer, or producing significant adverse 19 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 effects on the animals' health (see generally NRC, 1993b). Dose selections for long-term 2 studies are based on results of subchronic studies along with any available information on the 3 disposition, metabolism, and pharmacokinetics of the test agent. The aims in selecting dose 4 levels are both adequate sensitivity and the opportunity to observe tumor effects specific to 5 the agent. Failure to reach an adequately high dose reduces the sensitivity of the study. On 6 the other hand, excessive general toxicity, or toxicity in a target tissue for tumor effects, 7 raises the question whether tumor effects are specific to the agent or are nonspecific effects 8 secondary to the toxicity. There is no completely satisfactory answer to the possible conflict 9 between these aims. For instance, reducing the high dose to avoid any toxicity would reduce 10 the sensitivity of a protocol that is at best able to detect an increase of tumor incidence of 11 10%, if there is no spontaneous background incidence (Haseman, 1983). Reliable detection 12 of a 1 % increase would require using thousands of animals, which is not a feasible option. 13 Any of the following treatment-related findings may indicate that other, noncancer 14 toxicity may confound results of the cancer studies: 15 A 10%, or greater, increase in mortality due to toxicity 16 Significant toxicity manifested by a decrement in body weight greater than 10 to 15% 17 (not due to reduced food consumption from impalatability) 18 Significant toxicity manifested by clinical signs, hematological or chemistry measures, 19 or changes in organ weight, morphology, and histopathology that may interact with 20 the carcinogenic process or obscure interpretation of the results 21 Absence of tumor effects in an adequately sensitive study that is well conducted is accepted 22 as a negative finding. Studies with undue effects on mortality or health that show no tumor 23 effects are accepted as negative, if lower doses are appropriately spaced and mortality does 24 not remove too many animals from the study. Studies of inadequate sensitivity may be used 25 to bound the range of potential risk. Studies that show tumor effects only at doses with 26 undue effects on mortality or health are compromised and may or may not contribute to the 27 analysis, depending on interpretation in the context of other study results and other lines of 28 evidence. The effect of toxicity is examined, even when it is not excessive. In all cases, the 20 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE strength of inferences to be drawn about human carcinogenic potential from the statistical, and biological significance of animal tumor effects is judged in conjunction with all other available data. 2.2.2.2. Special Studies Specialized short-term studies often use protocols that screen for effects, usually preneoplastic effects, in a single tissue. The selected tissue is, in a sense, the test system. Certain systems of this kind are thought to be particularly informative about carcinogenic potential of defined chemical classes or agents, or modes of action (reviewed in Vainio et al., 1992). Other experimental systems such as strain A mice that are bred to be particularly sensitive may also provide some information. Nevertheless, limitations in experimental protocol, such as short duration, limited histology, or lack of complete development of tumors, make results from these studies correspondingly limited in their contribution to assessment. Their results are appropriate for identifying subjects of further research and for considering the effects seen in context with other evidence, especially regarding potential i modes of action. Transgenic animals carrying introduced proto-oncogenes, or activated oncogenes or tumor suppressor genes are promising experimental systems for probing mode of action questions when experience that supports their interpretation is in place. The strength of inferences that such studies support rests on their contribution to the congruity of evidence about an agent. 2.2.2.3. Significance for Human Hazard These guidelines adopt the science policy position that tumor findings in animals indicate that an agent may produce such effects in humans. Moreover, the absence of tumor findings in well-conducted, long-term animal studies in at least two species provides reasonable assurance that an agent may not be a carcinogenic concern for humans. Each of these is a default assumption that may be adopted, when appropriate, after evaluation of tumor data and other key evidence. 21 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 Several kinds of observations from animal studies can contribute to the judgment 2 whether animal responses indicate a significant carcinogenic hazard to humans. Significance 3 is a function of the number of factors present and, for a factor such as malignancy, the 4 severity of the observed pathology. The following observations add significance: 5 uncommon tumor types 6 tumors at multiple sites 7 tumors by more than one route of administration 8 tumors in multiple species, strains, or both sexes 9 progression of lesions from preneoplastic to benign to malignant 10 reduced latency of neoplastic lesions 11 metastases 12 unusual magnitude of tumor response 13 proportion of malignant tumors 14 dose-related increases 15 These features or their absence also may provide useful suggestions about a possible * 16 mode(s) of action of an agent. For example, multisite and multispecies effects are often 17 associated with genotoxic agents. Effects restricted to one sex may suggest an influence 18 connected with gender-related biological differences such as hormonal status. Late onset of 19 tumors that are primarily benign, or reversal of lesions on cessation of exposure, may point 20 to a growth-promoting mode of action. Similarly, a general increase in incidence of a 21 tumor with a high spontaneous background with no increase in malignancy, may indicate a 22 growth-promoting activity. The possibility that an agent may act differently in different 23 tissues or have more than one mode of action in a single tissue is kept in mind. 24 Site concordance of tumor effects between animals and humans is an issue to be 25 considered in each case. Thus far, there is evidence that growth control mechanisms at the 26 level of the cell are homologous among mammals, but there is no evidence that these 27 mechanisms are site concordant. Moreover, agents observed to produce tumors in both 28 humans and animals have produced tumors either at the same (e.g., vinyl chloride) or 22 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE different sites (e.g., benzene) (NRC, 1994). Hence, site concordance is not assumed a priori. On the other hand, certain processes with consequences for particular tissue sites (e.g., disruption of thyroid function) may lead to an anticipation of site concordance. Relevance of tumor responses in nonhuman species to human hazard is an integral judgment in analysis of bioassay results. The assumption of relevance is less appropriate when a body of evidence supports a mode of action in animals that would not be seen in humans under the same conditions of exposure. The Agency will undertake analyses of relevance issues as needed in reports to be published from time to time (e.g., U.S. EPA, 1991b). 2.3. ANALYSIS OF OTHER KEY EVIDENCE Certain structural, chemical, and biological attributes of an agent provide key information about its potential to cause or influence carcinogenic events. These attributes along with comparative studies between species also provide clues as to the potential mode of carcinogenic action and the potential dose-response relationship. The following sections provide guidance for analyses of these attributes. 2.3.1. Physicochemical Properties Physicochemical properties affect an agent's absorption, tissue distribution (bioavailability), biotransformation, or degradation in the body. These dispositions are important determinants of hazard potential. Properties to analyze include, but are not limited to, the following: molecular weight, size, and shape; valence state; physical state (gas, liquid, solid); water or lipid solubility that can influence retention and tissue distribution; and potential for chemical degradation or stabilization in the body. An agent's potential for chemical reaction with cellular components, particularly with DNA and proteins, is also important. The agent's molecular size and shape, electrophilicity, and charge distribution are considered to decide whether they would facilitate such reactions. 23 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 2.3.2. Structure-Activity Relationships (SAR) 2 Results of SAR analysis may strengthen or weaken the concern for an agent's 3 carcinogenic potential, depending on confidence in the SAR analysis. SAR analysis supports 4 evaluation of carcinogenic potential of both tested and untested chemicals. 5 Currently, SAR analysis is most useful for chemicals that are believed to produce 6 carcinogenesis, at least initially, through covalent interaction with DNA (i.e., DNA-reactive, 7 mutagenic, electrophilic, or proelectrophilic chemicals) (Ashby and Tennant, 1991). The 8 following parameters are useful in comparing an agent to its structural analogues and 9 congeners that have tumor effects and effects on related biological processes such as receptor 10 binding, genotoxicity, and general toxicity (Woo and Arcos, 1989): 11 nature and reactivity of the electrophilic moiety or moieties present 12 potential to form electrophilic reactive intermediate(s) through chemical, 13 photochemical, or metabolic activation 14 contribution of the carrier molecule to which the electrophilic moiety(ies) is attached 15 physicochemical properties (e.g., physical state, solubility, octanol-water partition 16 coefficient, half-life in aqueous solution) 17 structural and substructural features (e.g., electronic, stearic, molecular geometric) 18 metabolic pattern (e.g., metabolic pathways and activation and detoxification ratio) 19 possible exposure route(s) of the agent 20 Suitable SAR analysis of non-DNA-reactive chemicals and of DNA-reactive chemicals 21 that do not appear to bind covalentiy to DNA requires knowledge or postulation of the 22 probable mode(s) of action of closely related carcinogenic structural analogues (e.g., 23 receptor-mediated, cytotoxicity related). Examination of the physicochemical and 24 biochemical properties of the agent may then allow one to assess the likelihood of its activity 25 by that mode of action. 26 24 07/25/94 . ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE 2.3.3. Metabolism and F'harmacokinetics Studies of the absorption, distribution, biotransformation and excretion of agents permit comparisons among species to assist in determining the implications of animal responses for human hazard assessment, to support identification of active metabolites, to identify changes in distribution and metabolic pathway or pathways over a dose range and between species, and to make comparisons among different routes of exposure. If data are available (e.g., blood/tissue partition coefficients and pertinent physiological parameters of the species of interest), physiologically based pharmacokinetic models can be constructed to assist in determination of tissue dosimetry, species-to-species extrapolation of dose, and route-to-route extrapolation (Connolly and Andersen, 1991; see section 3.2.2.). Without such data, it is necessary to assume that pharmacokinetic and metabolic processes are qualitatively comparable between species if this appears generally reasonable. Adequate metabolism and pharmacokinetic data can be applied toward the following as data permit. Confidence in conclusions is greatest when in vivo data are available. Identifying metabolites and reactive intermediates of metabolism and determining whether one or more of these intermediates are likely to be responsible for the observed effects. This information on the reactive intermediates will appropriately focus SAR analysis, analysis of potential modes of action, and estimation of internal dose in dose-response assessment (D'Souza et al., 1987; Krewski el al., 1987). Identifying and comparing the relative activities of metabolic pathways in animals with those in humans. This analysis can provide insights for extrapolating results of animal studies to humans. Describing anticipated distribution within the body, and possibly identifying target organs. Use of water solubility, molecular weight, and structure analysis can support qualitative inferences about anticipated distribution and excretion. In addition, describing whether the agent or metabolite of concern will be excreted rapidly or slowly or will be stored in a particular tissue or tissues to be mobilized later can 25 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 identify issues in comparing species and formulating dose-response .assessment 2 approaches. 3 Identifying changes in pharmacokinetics and metabolic pathways with increases in 4 dose. These changes may result in the formation and accumulation of toxic products 5 following saturation of detoxification enzymes. These studies have an important role 6 in providing a rationale for dose selection in carcinogenicity studies. In addition, they 7 may be important in estimating a dose over a range of high to low exposure for the 8 purpose of dose-response assessment. 9 Determining bioavailability via different routes of exposure by analyzing uptake 10 processes under various exposure conditions. This analysis supports identification of 11 hazard for untested routes. In addition, use of physicochemical data (e.g., octanol- 12 water partition coefficient information) can support an inference about the likelihood 13 of dermal absorption (Flynn, 1990). 14 In all of these areas, attempts are made to clarify and describe as much as possible 15 the variability to be expected because of differences in species, sex, age, and route of 16 exposure. Utilization of pharmacokinetic information takes into account that there may be 17 subpopulations of individuals who are particularly vulnerable to the effects of an agent 18 because of pharmacokinetic or metabolic differences (genetically or environmentally 19 determined). 20 21 2.3.4. Short-Term Studies 22 Results from short-term in vitro and in vivo studies of effects that relate to 23 carcinogenic events are useful in the interpretation of epidemiological and animal data on 24 tumor effects. They are often the primary source of information about possible modes of 25 action. 26 A large number of short-term assays exist for examining biological activities relevant 27 to the carcinogenic process (e.g., mutagenesis, tumor promotion, aberrant intercellular 28 communication, increased cell proliferation, malignant transformation, immunosuppression). 26 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 * DRAFT-DO NOT QUOTE OR CITE These assess a variety of end points; confidence in the inferences to be drawn from results of each assay varies according to experience with the test system and its interpretation. Both the relevance of tested end points to potential carcinogenicity and confidence in interpretation are issues explicitly evaluated. 2.3.4.1. Genotoxicity Information "Genotoxicity" is an inclusive term that refers to effects of agents that interact with or alter the genome. "Mutagenicity" constitutes the part of genotoxicity associated with heritable changes in DNA structure or content. Mutations, including changes in DNA sequence and gene amplification, and structural and numerical chromosome aberrations are attributes of tumor cells and of predisposing genetic diseases. Recent studies on oncogenes provide additional evidence for the linkage between mutation and cancer (Bishop, 1991). Activation of proto oncogenes to oncogenes can be triggered, for example, by point mutations, DNA insertions, or chromosomal translocation (Bishop, 1991). In addition, the inactivation of tumor suppressor genes can occur by chromosomal deletion or loss or by mitotic recombination (Bishop, 1989; Varmus, 1989; Stanbridge and Cavenee, 1989). Mutagenic endpoints are, therefore, of obvious interest. The process by which an agent brings about mutations is also of interest, in part because different processes have different implications for the dose-response relationship. Agents may react directly with DNA as evidenced by DNA adducts, strand breakage, and intercalation between bases. Alternatively, they may react with other cell components, with DNA damage as an indirect consequence, e.g., interfere with spindle formation or with repair or recombination processes. Different numbers of chemical reactions during critical periods of the cell cycle are inherent in direct versus indirect processes, with implications for linearity or nonlinearities in the dose-response relationship. The EPA has published testing protocols and guidelines for detection of mutagenicity (U.S. EPA, 199la). A method to portray data graphically is the genetic activity profile methodology (Garrett et al., 1984; Waters et al., 1988). 27 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 A higher level of confidence that an agent is a mutagen is assigned to agents that 2 consistently induce sequence or structural changes in DNA in a number of test systems. 3 Although important information can be gained from in vitro assays, a higher level of 4 confidence is given to a data set that includes in vivo data because many agents require 5 metabolic conversion to an active intermediate for biological activity. Metabolic activation 6 systems can be incorporated into in vitro assays, but they may not mimic mammalian 7 metabolic activation and inactivation. If available, human genetic toxicity end points relevant 8 to carcinogenesis are important in vivo data. 9 Because mutagenic carcinogens have been observed to induce tumors across species 10 and at multiple sites, evidence of both mutagenicity and tumor responses in multiple species 11 or sexes significantly increases concern for the human carcinogenic potential of an agent. 12 Agents that induce mutations in the germinal cells of animals are similarly of concern. 13 About 30 agents, many of which are carcinogenic to humans, have been shown to induce 14 germ cell mutations in animals and may be anticipated to do so in somatic cells. Absence of 15 mutagenicity in several test systems, preferably including in vivo tests for genotoxicity in the 16 tumor target tissue (e.g., DNA alkylation, unscheduled DNA synthesis) suggests that another 17 mode of action may be important (Ashby, J., chapters in Vainio et al., 1992). The absence 18 of evidence of mutagenicity in an adequate data base and the lack of responses in a chronic 19 rodent bioassay gives reasonable confidence that an agent is not a human cancer hazard 20 unless there is human evidence to the contrary. 21 22 2.3.4.2. Other Short-Term Test Information 23 Nongenotoxic agents are also known to affect carcinogenesis. For example, 24 hormones and growth factors or agents that mimic them alter DNA functions that are critical 25 in the cell cycle. Other agents may cause cell toxicity which results in compensatory cell 26 replication or selectively promotes growth of initiated cells. Dose-response relationships for 27 some agents may be tied to their cell toxicity. 28 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE The relationship of dose and response is more complex for agents that affect the function of cell cycle controls, encountering issues of human variability in endocrine and exocrine balance and background exposures (U.S. EPA, 1994). Clues about nongenotoxic modes of action can come from tests for increased cell proliferation, cell transformation, aberrant intercellular communication, receptor mediated effects, or changes in gene transcription. Cell proliferation plays a key role at each stage in the carcinogenic process, and it is well established that increased rates of cell proliferation are associated with increased cancer risk. An agent may produce cellular toxicity with compensatory cell proliferation or act as a mitogen directly or indirectly (e.g., by affecting endocrine function). Increased risk may be due to bypassing DNA repair checkpoints or to interference with apoptosis or differentiation resulting in an increase in the number of proliferating cells at risk of spontaneous and induced genetic damage. Evidence for an increased rate of cell division may be determined by measuring the mitotic index, or by supplying a specific DNA precursor to cells (e.g., 3H- thymidine or bromodeoxyuridine) and counting the percentage of cells that have incorporated the precursor into replicating DNA, or by immunodetection of proliferation-specific antigens. These analyses can be carried out in vitro, or during prechronic animal studies, or as part of long-term animal studies. Assays for measuring perturbation of gap-junctional intercellular communication may provide an indication of carcinogenicity, especially promotional activity (Yamasaki, 1990). Cell transformation assays have been widely used for studying mechanistic aspects of chemical carcinogenesis because in vitro cell transformation is considered to be relevant to the in vivo carcinogenic process. Both of these kinds of assays have been proposed as tests for identifying nongenotoxic carcinogens (Swierenga, S.H.H. and Yamasaki, H., chapter in Vainioetal., 1992) There is as yet a paucity of standard tests for effects specifically on the cell cycle, although as the fields of cell cycle and cancer research merge, more cell cycle study methods may become available for screening agents for such effects. 29 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 2.3.4.3. Biomarker Information 2 Several genotoxicity end points and other measurements can serve as biological 3 markers of events in biological systems or samples. In some cases, these molecular or 4 cellular effects (e.g., DNA or protein adducts, structural chromosomal aberrations, levels of 5 thyroid stimulating hormone) can be measured in blood, body fluids, and cancer target 6 organs to serve as biomarkers of exposure (Callemen et al., 1978; Birner et al., 1990). As 7 such they can do the following: 8 act as an internal, surrogate measure of chemical dose, 9 help identify doses at which elements of the carcinogenic process are operating, 10 aid in interspecies extrapolations when data are available from experimental animal 11 and human cells, and 12 under certain circumstances, provide insights into the shape of the dose-response 13 curve below levels where tumor incidences are observed (e.g., Choy, 1993). 14 Genetic and other findings (like changes in proto-oncogenes and tumor suppressor 15 genes in neoplastic tissue or measures of endocrine disruption) can indicate the potential for 16 disease and as such serve as biomarkers of effect. They, too, can be used in different ways: 17 The spectrum of genetic changes in tumors following chemical administration to 18 experimental animals can be determined and compared to those in spontaneous tumors 19 in control animals, in animals exposed to other agents, and in persons exposed to the 20 agent under study. 21 As with biomarkers of exposure, it may be justified in some cases to use these 22 endpoints to provide insight into the shape of the dose-response curve at doses below 23 those at which tumors are induced. 24 25 2.3.4.4. Confidence in Conclusions 26 All data from short-term studies become part of the overall evaluation. It is 27 recognized that agents will not typically produce uniform responses in all short-term tests. 28 The reviewer is often left with a mix of responses in different tests. To the extent data 30 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 permit, the reviewer makes judgments as to genotoxic and nongenotoxic effects, the degree 2 to which the effects may influence observed tumor formation, and the influence of effects in 3 judging human hazard potential and dose-response implications. Considerations and elements 4 of confidence in such judgments include, but are not limited to, the following: 5 spectrum of endpoints relevant to carcinogenicity 6 number of studies of each endpoint 7 consistency of results in different test systems and different species 8 in vivo as well as in vitro observations 9 reproducibility of results in a test system 10 existence of a dose-response relationship for effects 11 , tests conducted in accordance with generally accepted protocols . 12 degree of consensus and general experience among scientists regarding interpretation 13 of the significance and specificity of the tests 14 At the end of the evaluation, when conclusions are offered, the conclusions are accompanied 15 by a statement of the confidence supported by the data base. 16 17 2.4. MODE OF ACTION-IMPLICATIONS FOR HAZARD CHARACTERIZATION 18 AND DOSE-RESPONSE 19 The analysis of the entire range of data reviewed in the assessment of tumor effects 20 studies and other key evidence is the support for arriving at a judgment about the potential 21 mode(s) of action of an agent. The purpose for analyzing the mode of action is to make a 22 reasoned judgment about the ways agents appear to be producing carcinogenic effects. This 23 judgment affects the ways of characterizing hazards to humans and means of evaluating 24 potential dose-response relationships. Given the gaps in the general understanding of 25 carcinogenesis, these analyses and judgments do not lay out detailed molecular mechanisms. 26 Nevertheless, commonly available data support general views as to physical, chemical, and 27 biological factors that appear to be influencing the carcinogenic process and are 28 acknowledged by scientists as playing a role. For hazard characterization, these analyses 31 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 address the relevance of the animal tumor model to humans and the conditions of expression 2 of potential hazard (see, for example, Appendix A, Narratives 1 and 4). For dose-response 3 assessment, these analyses help to guide the development of biologically based models and, 4 in the absence of detailed information, to select among different default techniques for low- 5 dose extrapolation. 6 The analysis reviews all the relevant information on the agent including observed 7 tumor responses and other key evidence. The entire list of factors to be considered in this 8 analysis would be beyond the scope of these guidelines to list. Some of the important ones 9 include the following: 10 number of tumor sites, sexes, studies, and species affected or unaffected 11 nature of tumor sites, e.g., responsive to endocrine influence or not 12 route of exposure effects 13 target organ or systemic effects (e.g., target organ toxicity, urinary chemical changes 14 associated with stones, effects on immune surveillance) 15 progression of lesions from preneoplastic to benign to malignant 16 early or late appearance of tumors after exposure 17 proportion of malignant to benign tumors 18 tumors invading locally, metastasizing, producing death 19 tumors at sites in laboratory animals with high or low spontaneous historical incidence 20 biomarker of effect in tumor cells, such as mutation spectra (like or unlike 21 spontaneous tumors), DNA or protein adducts, chromosome changes 22 shape of the dose response in the range of tumor observation 23 structural relationship of the agent or its metabolites to mutagenic carcinogens 24 structural relationship of the agent or its metabolites to agents that are non-DNA- 25 reactive, or influence other processes (e.g., hormone disruption) 26 genotoxic effects 27 effects on cell proliferation (e.g., mitogenic effects, effects on differentiation or 28 apoptosis, response to toxicity) 32 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24, 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE effects on intercellular communication evidence of initiating or promoting activity in two-stage assays in vitro or in vivo Dose-response default positions include low-dose linear extrapolation, nonlinear, and both procedures. Both procedures are warranted when (a) tumors at different sites appear to have different modes of action (see, for example, Appendix A, Narrative 4), (b) there is evidence in support of both default procedures (see, for example, Appendix A, Narrative 5), and (c) there is considerable uncertainty as to potential mode of action (see, for example, Appendix A, Narratives 3 and 6). Many combinations of data elements are possible. For example, a data set that might lead to a conclusion that a linear dose-response default is reasonable might include: tumors at multiple sites, sexes, and species, with structural alerts and good evidence of DNA- reactivity in short-term tests (e.g., Appendix A, Narrative 7). Finding that there are tumor responses in two species at sites that have high spontaneous rates of tumor incidence, the effect at each site being to accelerate appearance of tumors without altering the spontaneous proportion of benign to malignant tumors coupled with finding no structural alerts or evidence of genotoxicity in extensive short-term testing, would support a nonlinear dose- response default procedure. 2.5. HAZARD CHARACTERIZATION 2.5.1. Carcinogenic Potential for Humans The hazard characterization summarizes and evaluates the analyses of pertinent data concerning carcinogenic hazard potential and applies them to make a conclusion as to carcinogenicity to human beings. The conclusion regards both the general population and subpopulations identified as particularly sensitive. Evaluation addresses the significant strengths and weaknesses of existing data. It also explains the reasoning that supports data interpretation, including the use of default assumptions, and weighs the coherence of implications of different kinds of data. The central purpose of evaluation is to explore and explain how the available empirical evidence may relate to biological activity in humans. 33 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE No single kind of evidence can reveal the whole biological story otherwise told by bringing together all of the available information. Moreover, the kinds of evidence that are the most telling are different from case to case. Some illustrations of this are well known: Metabolism information~the presence or absence of a single enzymewas the key to explaining why certain aromatic amines cause bladder cancer in dogs and humans, but not in rodents. The activity and high levels of a small protein, alpha 2u globulin, in male rats was the key to finding that a particular kidney tumor in these animals has no relevant parallel in humans. Appendix A contains several examples highlighting key roles for other kinds, of information: Narrative 4 illustrates that data on an agent's homology in biological effects and chemical structure to a carcinogenic analogue can support the conclusion that the agent is likely to be carcinogenic, although there are no tumor effects data on the agent itself. Narrative 1 illustrates how information on physiological perturbation and physical irritation can explain mode of action sufficiently to define dose as a limiting factor on potential expression of carcinogenic effects. Narrative 6 shows that information on absorption and toxicity can lead to different conclusions about carcinogenic potential by different routes of exposure. 2.5.2. Conditions of Expression A characterization qualitatively describes the conditions under which the agent's effects may be expressed in human beings. The description includes routes of exposure and dose levels and durations of exposure. Discussion of approaches to quantitative assessment of internal or delivered dose according to route of exposure appears in section 3 below. Qualitative characterization describes whether the route of exposure is likely to be a limiting factor on expression of effects. The supporting data include studies hi which effects of the agent or analogues on animals or humans have been observed by differing routes, or physical-chemical properties, 34 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 DRAFT-DO NOT QUOTE OR CITE or pharmacokinetics studies. For certain agents there may be enough known about properties of their class to make confident conclusions regarding absorption by different routes. For others, more data on the specific agent may be necessary as conclusions may be less confident, or not possible, without such information. Discussion of these issues is part of the characterization. Relationship of dose level, pattern, and duration of exposure to effects can theoretically take many forms. Hazard assessment examines all of the kinds of evidence to see what processes or events in carcinogenesis a specific agent appears to affect. This information is used to relate the mode of action in general terms to the approach to quantitative dose-response assessment. The mode of action may imply a linear or a non- linear dose-response relationship, or a threshold of dose below which effects will not occur in an individual. The implications of available information about human variability in sensitivity or with respect to threshold effects are important elements. These implications relate to both the qualitative characterization of hazard and the quantitative assessment of dose-response. 2.5.3. Descriptions of Weight of Evidence The hazard narrative, described in the following subsection, presents the weight of evidence in terms of likelihood of human carcinogenicity using descriptors. The descriptors are not meant to replace an explanation of the nuances of the biological evidence, but rather to summarize it. Each descriptor spans a wide variety of data sets and weights of evidence. There will always be gray areas, gradations, and borderline cases. The narrative preserves and presents this complexity, which is an essential part of the hazard characterization. Applying a descriptor is a matter of judgment and cannot be reduced to a formula. Risk managers should consider the entire range of information included in the narrative rather than focus simply on the descriptor. 35 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE Note that one agent may fit more than one descriptor, if, for instance, the agent were likely to be carcinogenic by one route of exposure and not by another. Use of the descriptors is illustrated hi Appendix A. The descriptions below are intended only as guidance and provide some typical examples; the examples are not exhaustive or comprehensive. "Likely" or "Known" These descriptors are appropriate when the evidence provides a reasonable assurance of carcinogenic potential for human beings and supports proceeding with the risk assessment. "Likely" is the descriptive term generally used. "Known" is used when the weight of evidence gives especially high assurance because either an association between carcinogenicity and a specific route of exposure is drawn from human data, or conclusions from other kinds of data give confidence that is equal to having human data. "Cannot be determined" "Cannot be determined" is appropriate whenever support for a conclusion about carcinogenic potential for human beings is not sufficient to proceed with the assessment. Where appropriate, the narrative explains the situation. Examples may include the following, among others. The evidence raises a concern for carcinogenic effects, but falls short of supporting a conclusion about the likelihood of effects. The narrative provides a summary of the research or testing needed to explore the issue further. The added descriptor, "testing candidate" is appropriate in these cases to flag the agent for attention by testing programs. The data are inadequate to perform an assessment. The information is inconclusive or conflicting, e.g., some evidence is suggestive of carcinogenic effects, but other equally pertinent evidence does not confirm any concern. 36 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 . 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE "Not Likely" "Not likely" is appropriate whenever evidence about an agent generally, or about a condition of exposure, is satisfactory for deciding that there is not a basis for concern. Again, the narrative explains the conclusion. Examples may include the following: The agent has been adequately characterized empirically, and the conclusion is negative, or the only positive data are not considered relevant to human beings. Adequate empirical characterization generally includes well conducted, long-term animal studies on an agent or its structural analogue with consistent findings from analysis of other key evidence. The evidence shows that under certain conditions of exposure, no expression of carcinogenic effects is anticipated. The agent's carcinogenic potential is categorized as "not likely" for those conditions, e.g., a route of exposure or a defined dose level. The agent has been adequately characterized empirically, and the only positive indication of effects was seen under experimental circumstances that are implausible for raising an environmental concern, e.g., injection of a polymer. 2.5.4. Hazard Narrative A narrative summarizes the results of hazard characterization. The narrative, optimally two pages or less in length, explains an agent's human carcinogenic potential and the conditions of its expression. If data do not allow a conclusion, the narrative explains this determination. Examples of narratives appear in Appendix A below as guidance for format and content. The items regularly included are these: name of agent and Chemical Abstracts Services number, if available a brief identification of the kinds of data available conclusions (by route of exposure) about human carcinogenicity, described as "known" or " likely," "not likely," or "cannot be determined" summary of tumor data, human or animal, on the agent and/or its structural analogues, their relevance, and biological plausibility 37 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 Other key evidence, e.g., structure-activity data, pharmacokinetics and metabolism, 2 short-term studies, other relevant toxicity data 3 effect of route of exposure 4 discussion of possible mode(s) of action and appropriate dose-response approaches) 5 strengths and weaknesses of the evidence and assumptions significant to the case 6 The following items may be added if justified: 7 depiction of the evidence as particularly strong or weak as compared to other cases. 8 a description of one or more research approaches or tests that can be anticipated to 9 resolve a critical question identified in the assessment. 10 11 3. DOSE-RESPONSE ASSESSMENT 12 13 Dose-response assessment addresses both the relationship of dose2 to degree of 14 response observed empirically and the nature of this relationship at environmental exposure 15 levels of interest below the range of observation. Analysis of responses includes tumor 16 responses in human and animal studies and the agent's effects on macromolecules involved in 17 growth control or other toxic effects that may play a role in carcinogenesis. Analyses of 18 dose use available metabolism and pharmacokinetic data to identify appropriate measures of 19 applied dose and, as data permit, improve the analysis by identifying measures of internal or 20 delivered dose. If empirical data are from animal studies, the analysis extrapolates animal 21 doses to human equivalent doses. Depending on the availability of data, dose-response 22 assessments are carried out for any effect associated with an agent's carcinogenicity to assess 23 potential for effects from environmental exposure. 24 2For this discussion, "exposure" means contact of an agent with the outer boundary of an 25 organism. "Applied dose" means the amount of an agent presented to an absorption barrier 26 and available for absorption. "Internal dose" means the amount crossing an absorption 27 barrier (e.g., the exchange boundaries of skin, lung, and digestive tract) through uptake 28 processes. "Delivered dose" for an organ or cell means the amount available for interaction 29 with that organ or cell (U.S. EPA, 1992a). 38 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 3.1. RESPONSE DATA 2 Response data of interest for analysis include tumor incidence data from human or 3 animal studies. They also include data on other responses as they relate to an agent's 4 carcinogenic effects, for instance, effects on growth control processes or cell macromolecules 5 or other toxic effects. Tumor incidence data is ordinarily the basis of dose-response 6 assessment, but availability of other response data can augment such assessment or provide 7 separate assessments of other important effects. Data on carcinogenic processes underlying 8 tumor effects may be used to support biologically based models. If confidence is high in the 9 linkage of a precursor effect and the tumor effect, the assessment of tumor incidence may be 10 extended to lower dose levels by linking it to the assessment of the precursor effect 11 (Swenberg et al., 1987). Linking analyses may not be appropriate; even so, the assessment 12 for a precursor effect may provide a view of the likely shape of the dose-response curve for 13 tumor incidence below the range of tumor observation (Cohen1 and Ellwein, 1990; Choy, 14 1993). The effects of an agent on cell macromolecules may be used as markers of 15 carcinogenic effect for which a separate dose-response assessment may be done, or such 16 effects may provide markers of exposure in support of dose analysis. In some cases, the 17 dose-response relationship for a response other than tumor incidence may be more useful 18 than tumor incidence to assess potential effects associated with environmental exposure. For 19 example, if it is concluded that the carcinogenic effect is secondary to another toxic effect, 20 the dose-response for the other effect will likely be more pertinent for risk assessment. The 21 dose-response relationship for a precursor effect may also be more useful, for example, 22 disruption of hormone activity when this is considered the key mode of action of an agent. 23 These kinds of supplementary analyses are presented and evaluated whenever data are 24 available to support them. 25 If adequate positive human epidemiologic data are available, they provide an 26 advantageous basis for analysis since concerns about interspecies extrapolation do not arise. 27 Adequacy of human exposure data for quantification is an important consideration in deciding 28 whether epidemiologic data are the optimal basis for analysis in a particular case. Positive 39 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 data are analyzed to estimate response to environmental exposure in the observed range. 2 (U.S. EPA, 1992a). Extrapolation to lower environmental exposure ranges is carried out, as 3 needed. Analysis of associated responses as described in the paragraph above supplements 4 analyses of human tumor data as well as those of animal tumor data. If adequate exposure 5 data exist in a well-designed and well-conducted epidemiologic study that detects no effects, 6 it may be possible to obtain an upper-bound estimate of the potential human risk to provide a 7 check on plausibility of available estimates based on animal tumor or other responses. 8 When animal studies are used, response data from a species that responds most like 9 humans should be used, if information to this effect exists. If this is unknown, and an agent 10 has been tested in several experiments involving different animal species, strains, and sexes 11 at several doses and different routes of exposure, all of the data sets are considered and 12 compared, and a judgment is made as to the data to be used to best represent the observed 13 tumor incidence data Snd important biological features such as mode of action. Appropriate 14 options for presenting results include use of a single data set, combining data from different 15 experiments (Stiteler et al., 1993; Vater et ah, 1993), showing a range of results from more 16 than one data set, showing results from analysis of more than one tumor response based on 17 differing modes of action, representing total response in a single experiment by combining 18 animals with tumors, or a combination of these options. The approach judged to best 19 represent the data is presented with the rationale for the judgment, including the biological 20 and statistical considerations involved. The following are some points to consider: 21 high quality of study protocol and execution 22 malignancy of neoplasms 23 latency of onset of neoplasia, 24 number of data points to define the relationship of dose and response 25 background incidence in test animal 26 most sensitive responding species 27 data on a related effect (e.g., DNA adduct formation) are available to augment the 28 tumor effect data 40 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 DRAFT-DO NOT QUOTE OR CITE Analyses of carcinogenic effects other than tumor incidence are similarly presented and evaluated for their contribution to a best judgment on how to represent the biological data for dose-response assessment. 3.2. DOSE DATA Whether animal experiments or epidemiologic studies are the sources of data, questions need to be addressed in arriving at an appropriate measure of dose and, in case of animal data, a measure of dose appropriate to humanshuman equivalent dosethat is matched to the anticipated route of environmental exposure. Among these are: (1) Is the parent compound, a metabolite, or both, active in the process? and (2) How well do data at hand measure available dose and support pharmacokinetic modeling of internal or delivered dose? In practice, there may be little or no information on the concentration or identity of the active form at a target; being able to compare the applied and delivered doses between routes and species is the ideal, but is rarely attained. Even so, the objective is to use available data to reach a measure of internal or delivered dose if possible. Even if pharmacokinetic and metabolic data are sufficient to derive a measure of delivered dose to the target, the dose-response relationship is also affected by kinetics of reactions at the target (pharmacodynamics) and by other steps in the development of neoplasia. With few exceptions, these processes are currently undefined, and this is a limitation to the ultimate aim of dose analysis which is to have a measure of effective dose to the target. The following discussion assumes that the analyst will have data of varying detail in different cases about pharmacokinetics and metabolism. Discussed below are approaches to basic data that are most frequently available, as well as approaches and judgments for improving the analysis based on additional data. 41 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 3.2.1. Basic Analyses 2 When there are insufficient data available to compare dose between species, the 3 default assumption is that delivered doses through oral exposure are related to applied dose 4 by a power of body weight. This assumption rests on the similarities of mammalian 5 anatomy, physiology, and biochemistry generally observed across species. This assumption 6 is more appropriate at low applied dose concentrations where sources of nonlinearity, such as 7 saturation or induction of enzyme activity, are less likely to occur. To derive a human 8 equivalent oral dose from animal data, the default procedure is to scale daily applied doses 9 experienced for a lifetime in proportion to body weight raised to the 3/4 power (W3/4). 10 Equating exposure concentrations in parts per million units for food or water is an alternative 11 version of the same default procedure because daily intakes of these are in proportion to 12 \y3/4. The rationale for this factor rests on the empirical observation that rates of 13 physiological processes consistently tend to maintain proportionality with W3/4. A more 14 extensive discussion of the rationale and data supporting the Agency's adoption of this 15 scaling factor can be found in (U.S. EPA, 1992b). Information such as blood levels or 16 exposure biomarkers that are available for interspecies comparison is used to improve the 17 analysis when possible. 18 The default procedure to derive a human equivalent concentration of inhaled particles 19 and gases is described in U.S. EPA (in press) and Jarabek (in press). The methodology 20 estimates respiratory deposition of inhaled particles and gases and provides methods for 21 estimating internal doses of gases with different absorption characteristics. The method is 22 able to incorporate additional pharmacokinetics and metabolism to improve the analysis if 23 such data are available. 24 The differences in biological processes among routes of exposure (oral, inhalation, 25 dermal) can be great because of, for example, first pass effects and differing results from 26 different exposure patterns. There is no generally applicable method for accounting for these 27 differences in uptake processes in quantitative route-to-route extrapolation of dose-response 28 data in the absence of good data on the agent of interest. Therefore, route-to-route 42 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE extrapolation of dose data relies on a case-by-case analysis of available data. When good data on the agent itself are limited, an extrapolation analysis can be based on expectations from physical chemical properties of the agent, properties and route-specific data on structurally analogous compounds, or in vitro or in vivo uptake data on the agent. Route-to- route uptake models may be applied if model parameters are suitable for the compound of interest. Such models are currently considered interim methods; further model development and validation is awaiting the development of more extensive data (see generally, Gerrity and Henry, 1990). 3.2.2. Pharmacokinetic Analyses Physiologically based mathematical models are potentially the most comprehensive way to account for pharmacokinetic processes affecting dose. Models build on physiological compartmental modeling and attempt to incorporate the dynamics of tissue perfusion and the kinetics of enzymes involved in metabolism of an administered compound. A comprehensive model requires the availability of empirical data on the carcinogenic activity contributed by parent compound and metabolite or metabolites and data by which to compare kinetics of metabolism and elimination between species. A discussion of issues of confidence accompanies presentation of model results (Monro, 1992). This includes considerations of model validation and sensitivity analysis that stress the predictive performance of the model. Another assumption made when a delivered dose measure is used in animal-to-human extrapolation of dose-response data is that the pharmacodynamics of the target tissue(s) will be the same in both species. This assumption is discussed, and confidence in accepting it is considered in presenting results. Pharmacokinetic data can improve dose-response assessment by accounting for sources of change in proportionality of applied to internal or de21ivered dose at various levels of applied dose. Many of the sources of potential nonlinearity involve saturation or induction of enzymatic processes at high doses. An analysis that accounts for nonlinearity (for instance, due to enzyme saturation kinetics) can assist in avoiding over- 43 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 estimation or under-estimation of low dose-response otherwise resulting from extrapolation 2 from a sublinear or supralinear part of the experimental dose-response curve. (Gillette, 3 1983). Pharmacokinetic processes tend to become linear at low doses, an expectation that is 4 more robust than low dose linearity of response (Hattis, 1990). Accounting for 5 pharmacokinetic nonlinearities allows better description of the shape of the curve at higher 6 levels of dose, but cannot determine linearity or nonlinearity of response at low dose levels 7 (Lutz, 1990; Swenberg etal., 1987). 8 9 3.2.3. Additional Considerations for Dose in Human Studies 10 The applied dose in a human study has uncertainties because of the exposure 11 fluctuations that humans experience compared with the controlled exposures received by 12 animals on test. In a prospective cohort study, there is opportunity to monitor exposure and 13 human activity patterns for a period of time that supports estimation of applied dose (U.S. 14 EPA, 1992a). In a retrospective cohort study, exposure may be based on monitoring data, 15 but is often based on human activity patterns and levels reconstructed from historical data, 16 contemporary data, or a combination of the two. Such reconstruction is accompanied by 17 analysis of uncertainties considered with sensitivity analysis in the estimation of dose 18 (Wyzga, 1988; U.S. EPA, 1986a). These uncertainties can also be assessed for any 19 confounding factor, for which a quantitative adjustment of dose-response data is made (U.S. 20 EPA, 1984). 21 Exposure levels of groups of people in the study population often are represented by 22 an average when they are actually distributed in a range. The full extent of data are 23 analyzed and portrayed in the dose-response analysis when possible (U.S. EPA, 1986a). 24 The cumulative dose of an agent is commonly used when modeling human data. This 25 can be done, as in animal studies, with a default assumption in the absence of data that 26 support a different dose surrogate. Given data of sufficient quality, dose rate or peak 27 exposure can be used as an alternative surrogate to cumulative dose. 28 44 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22' 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE 3.3. SELECTION OF QUANTITATIVE APPROACH The goal in choosing an approach is to achieve the closest possible correspondence between the approach and the view of the agent's mode(s) of action. For this purpose, it is appropriate to analyze the dose-response of observed tumor incidence and other carcinogenic effects as discussed above, in section 3.1. Pharmacokinetic analysis or interspecies scaling or other appropriate methods are used to derive human-equivalent measures of the animal- administered dose. If the hazard assessment describes more than one mode of action as plausible and persuasive given the data available, corresponding alternative approaches for dose-response analysis are considered. 3.3.1. Analysis in the Range of Observation The analysis first addresses responses in the observed range. The responses include tumor incidence as well as other responses relevant to carcinogenicity. The latter may enlighten judgment about extrapolation or extend the range of observation (see section 3.3.1.). If data are sufficient to support a biologically based model specific to the agent, this is the first choice for both the observed tumor data and for extrapolation to the range of environmental exposures of interest. Dose-response models based on general concepts of a mode of action are next in amount of information required. For a specific agent, model parameters are obtained from laboratory studies. Examples are the two-stage models of initiation plus clonal expansion and progression developed by Moolgavkar and Knudson (1981) and Chen and Farland (1991). Such models require extensive data to build the form of the model as well as to estimate how well it conforms with the observed carcinogenicity data. Theoretical values for parameters, e.g., theoretical cell proliferation rates, are not used to enable application of such a model (Portier, 1987). If data are not available for such a model to be applied to observed tumor incidence data, a multistage model is appropriate for curve-fitting as a default approach (Zeise et al., 1987), unless characteristics of the data make another model more appropriate. For instance, when longitudinal data on tumor 45 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 development are available, time-to-tumor or survival models may be appropriate and 2 necessary to fit the data. Analyses of responses other than tumor effects are individually 3 designed and used to inform the extrapolation approach or used in lieu of tumor data if they 4 better portray the biology. 5 The ED10 (dose associated with a 10% response) of the observed dose-response for 6 tumor incidence (or another effect related to carcinogenesis) or is identified. This level is 7 adopted as a matter of Agency policy in order to remain consistent and comparable from case 8 to case; the rationale supporting this choice is that 10% is the limit of sensitivity of most 9 studies of tumor incidence. Determination of the ED10 is a matter of judgment. Because 10 statistical considerations (e.g., the number and spacing of dose levels, sample sizes, and the 11 precision and accuracy of dose measurements) can affect the precision of model estimates, 12 upper and lower confidence limits are stated with the ED,0. The divergence between upper 13 and lower confidence bounds provides a sense of the precision with which the modeling can 14 make projections in that range. The ED10 provides a point of departure for extrapolation of 15 tumor incidence data (or a related effect) and is an estimate that can be used for comparison 16 with similar analyses of the observed range of noncancer effects of an agent (U.S. EPA, 17 199 If) or to support hazard ranking among carcinogenic agents. It is also employed in the 18 dose-response and risk characterizations as a basis for describing the magnitude of 19 extrapolation needed to reach the range of environmental exposure of interest. 20 21 3.3.2. Extrapolation 22 If a biologically based or mode of action-based model has been used to portray the 23 observed data and confidence in the model is good, it may be extended to the level of 24 environmental exposure of interest. The reliability of the model is appropriately examined 25 by considering the point at which the upper and lower confidence bound on the curve diverge 26 to an unacceptable extent. If data are insufficient to support this extension, extrapolation 27 relies on a default procedure reflecting the general mode(s) of action supported by the 28 available biological information. The Agency adopts the three default procedure options .46 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 DRAFT-DO NOT QUOTE OR CITE described below as matters of policy based on current theory of the likely shapes of dose- response curves for differing modes of action. If a carcinogenic agent acts by accelerating the same carcinogenic process that leads to the background occurrence of cancer, the added effect on the population at low doses marginally above background level is expected to be linear. Above background level, the population response may continue to be linear (e.g., in the case of an agent acting directly on DNA), or be nonlinear reflecting complex physiological processes or events caused by multiple reactions (e.g., numerical chromosomal changes following reactions with spindle proteins, interrupting spindle formation). If the agent acts by mode of action with no endogenous counterpart, a population response threshold may exist (Crump et al., 1976; Peto, 1978; Hoel, 1980; Lutz, 1990). The Agency reviews each assessment as to the evidence on mode of action and other biological or statistical evidence that indicates the suitability of a particular procedure. In all cases, a rationale is included to justify the use of the selected procedure. Linear If the mode of action being considered leads to an expected linear low dose-tumor incidence relationship, a straight line is drawn from the EDi0 to the origin (zero dose, zero response) (Flamm and Winbush, 1984 ; Gaylor and Kodell, 1980; Krewski et al., 1984). This approach is generally conservative of public health, particularly in the absence of information about the extent of human variability in sensitivity to effects. Nonlinear The mode of action being considered may project that the dose-response relationship is nonlinear or is most influenced by individual differences in sensitivity. In this case, a model incorporating nonlinearity may be used to provide estimates of the proportion of the population at risk for specific doses of interest, e.g., 1/1000, 1/10000 lifetime risk levels. If an appropriate model is not available, a margin of exposure analysis may be used as 47 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 described below. The margin of exposure is the EDJO divided by the environmental exposure 2 of interest. 3 The mode of action may be one that involves a population threshold, e.g., the 4 carcinogenicity may be a secondary effect of toxicity that is itself a threshold phenomenon. 5 In these cases, the risk is not extrapolated as a probability of an effect at low doses. 6 Instead, a margin of exposure presentation is made in the risk characterization. The 7 environmental exposures of interest may be actual levels, projected levels after control, or 8 potential environmental standards. The risk manager decides whether a given margin of 9 exposure is adequate under applicable management policy criteria. The risk assessment 10 provides supporting information to assist the decision maker. Analysis to support 11 consideration of the margin of exposure conveys as much as possible about the anticipated 12 dose-response relationship in humans. This includes consideration of comparative sensitivity 13 of animals and humans, where animal data are the observed data, and consideration of human 14 variability in sensitivity to the putative mode(s) of action. 15 16 Both Linear and Nonlinear 17 Both linear and nonlinear procedures may be used in a particular case. If differing 18 modes of action appear to be involved in effects at different target tissues, the tissues may be 19 dealt with separately by linear and nonlinear procedures. If a mode of action analysis finds 20 equal support for modes of action with differing implications, both linear and nonlinear 21 procedures may be used. 22 If a dose-response assessment relies on data for an endpoint other than cancer, for 23 example, endocrine disruption, the assessment is individually designed and a rationale is 24 presented. The margin of exposure procedure is used when appropriate. 25 26 3.3.3. Issues for Analysis of Human Studies 27 Issues and uncertainties arising in dose-response assessment based on epidemiological 28 studies are analyzed in each case. Several sources of uncertainty need to be addressed in the 48 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE dose-response analysis. Consideration needs to be given to the data on the exposure and mortality experience of the study population and of the population that will represent the background incidence of the neoplasm(s) involved. In this area, there are potentials for mistakes or uncertainty in the data or adjustments to the data concerning the occurrence or level of exposure of the population members, mortality experience of a population, incomplete follow-up of individuals, exposure (or not) of individuals to confounding causes, or consideration of latency of response. These are assessed by analyzing the sensitivity of dose-response study results to errors where data permit. Other kinds of uncertainty can occur because of small sample size which can magnify the effects of misclassification or change assumptions about statistical distribution that underlie tests of statistical significance (Wyzga, 1988). These uncertainties are discussed. Where possible, analyses of the sensitivity of results to the potential variability in the data in these areas are performed. The suitability of various available mathematical procedures for quantifying risk attributed to exposure to the study agent is discussed. These methods (e.g., absolute risk, relative risk, excess additive risk) account differently for duration of exposure and background risk, and one or more can be used in the analysis as data permit. The use of several of these methods is encouraged when they can be used appropriately in order to gain perspectives on study results. 3.3.4. Use of Toxicity Equivalence Factors and Relative Potency Estimates A toxicity equivalence factor (TEF) procedure is one used to derive quantitative dose- response estimates for agents that are members of a category or class of agents. TEFs are based on shared characteristics that can be used to order the class members by carcinogenic potency when cancer bioassay data are inadequate for this purpose (U.S. EPA, 1991c). The ordering is by reference to the characteristics and potency of a well-studied member or members of the class. Other class members are indexed to the reference agent(s) by one or more shared characteristic to generate their TEFs. The TEFs are usually indexed at increments of a factor of 10. Very good data may permit a smaller increment to be used. 49 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 Shared characteristics that may be used are, for example, receptor-binding characteristics, 2 results of assays of biological activity related to carcinogenicity or structure-activity 3 relationships. 4 TEFs are generated and used for the limited purpose of assessment of agents or 5 mixtures of agents in environmental media when better data are not available. When better 6 data become available for an agent, its TEF should be replaced or revised. Criteria for 7 constructing TEF's are given in U.S. EPA (1991b). The criteria call for data that are 8 adequate to support summing doses of the agents in mixtures. 9 Relative potencies can be similarly derived and used for agents with carcinogenicity 10 or other supporting data. The criteria for these basically the same as for TEF's with the 11 exception that data of a quality that will support summing doses of agents in a mixture are 12 not required. 13 The uncertainties associated with both TEF's and relative potencies are explained 14 whenever they are used. 15 16 3.4. DOSE-RESPONSE CHARACTERIZATION 17 The conclusions of dose-response analysis are presented in a characterization section. 18 Because alternative approaches may be plausible and persuasive in selecting dose data, 19 response data, or extrapolation procedures, the characterization presents the judgments made 20 in such selections. The results for the approach or approaches chosen are presented with a 21 rationale for the one (or more than one if they are equally supported) that is considered to 22 best represent the available data and best correspond to the view of the mechanism of action 23 developed in the hazard assessment. 24 The exploration of significant uncertainties in data for dose and response and in 25 extrapolation procedures is part of the characterization. A distinction is made in the 26 presentation between model uncertainty and measurement uncertainty. Model uncertainty is 27 an uncertainty about a basic biological question. For example, a linear dose-response 28 extrapolation may have been made based on tumor and other key evidence supporting the 50 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE view that an agent's mode of action is through mutagenicity. Discussion of the confidence in this approach is appropriately done qualitatively, it is not amenable to useful quantitative uncertainty analysis. Measurement uncertainties deal with numbers representing statistical or analytical measures of variance or error in data or estimates. Uncertainties in measurement are described quantitatively, if practicable, through sensitivity analysis and statistical uncertainty analysis. With the recent expansion of readily available computing capacity, computer methods are being adapted to create simulated biological data that are comparable with observed information. These simulations can be used for sensitivity analysis, for example, to analyze how small, plausible variations in the observed data could affect dose- response estimates. These simulations can also provide information about experimental uncertainty in dose-response estimates, including a distribution of estimates that are compatible with the observed data. Because these simulations are based on the observed data, they cannot assist in evaluating the extent to which the observed data as a whole are idiosyncratic rather than typical of the true situation. If quantitative analysis is not possible, significant measurement uncertainties are described qualitatively. In either case, the discussion highlights those uncertainties that are specific to the agent being assessed, as distinct from those that are generic to most assessments. Numerical dose-response estimates are presented to one significant figure and qualified as to whether they represent central tendency or statistical upperbounds, and whether the method used is Inherently more likely to over-estimate, or under-estimate (Krewski etal., 1984). In cases where a mode of action or other feature of the biology has been identified that has special implications for early-life exposure, differential effects by sex, or other concerns for sensitive subpopulations, these are explained. Similarly, any expectations that high dose-rate exposures may alter the risk picture for some portion of the population are described. These and other perspectives are recorded to guide exposure assessment and risk characterization. Special problems arise when the human exposure situation of concern suggests exposure regimens, e.g., route and dosing schedule that are substantially different 51 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 from those used in the relevant animal studies. The cumulative dose received over a 2 lifetime, expressed as average daily exposure prorated over a lifetime, is generally 3 considered an appropriate measure of exposure to a carcinogen particularly for an agent that 4 acts by damaging DNA, if consistent with available data. The assumption is made that a 5 high dose of a carcinogen received over a short period of time is equivalent to a 6 corresponding low dose spread over a lifetime; this is based on theoretical considerations. 7 This approach becomes more problematic as the exposures in question become more intense 8 but less frequent, or when there is evidence that the agent acts by a mode of action involving 9 dose-rate effects. These issues are explored and pointed out for attention in the exposure 10 assessment and risk characterization. 11 12 4. EXPOSURE ASSESSMENT AND CHARACTERIZATION 13 14 Guidelines for exposure assessment of carcinogenic and other agents are published in 15 U.S. EPA (1992a) and are to be used in conjunction with these cancer risk assessment 16 guidelines. The exposure characterization is a key part of the exposure assessment; it is the 17 summary explanation of the exposure assessment. Major points of exposure characterization 18 are reiterated here to be considered in the cancer risk assessment context. The 19 characterization 20 provides a statement of purpose, scope, level of detail, and approach used in the 21 assessment; 22 presents the estimates of exposure and dose by pathway and route for individuals, 23 population segments, and populations in a manner appropriate for the intended risk 24 characterization; 25 provides an evaluation of the overall quality of the assessment and the degree of 26 confidence the authors have in the estimates of exposure and dose and the conclusions 27 drawn; and 52 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 II 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE communicates the results of exposure assessment to the risk assessor, who can then use the exposure characterization, along with the characterization of the other risk assessment elements, to develop a risk characterization. In general, the magnitude, duration, and frequency of exposure provide fundamental information for estimating the concentration of the carcinogen to which the organism is exposed. These data are generated from monitoring information, modeling results, or reasoned estimates. The potential for exposure via ingestion, inhalation, and dermal penetration from relevant sources of exposures, including multiple avenues of intake from the same source, are appropriate matters to consider in cancer risk assessment. 5. RISK CHARACTERIZATION 5.1. PURPOSE The risk characterization summarizes and integrates the major results of the risk assessment in a way that makes them understandable for all interested readers. Since the risk characterization provides a transition between risk assessment and risk management, one of its objectives is to be an appraisal of the science that the risk manager can use, along with other decision making resources, to make public health decisions. A complete characterization presents the risk assessment as an integrated, and balanced, picture of the analysis of the hazard, dose response, and exposure. It is the risk analyst's obligation to communicate not only summaries of the evidence and results, but also perspectives on the quality of available data and the degree of confidence to be placed in the risk estimates. These perspectives on the science include explaining the constraints of available data and the state of knowledge about the phenomena studied. All of this is in aid of a major aim of characterization which is to explain not only significant scientific issues, but also the significant science and science policy choices made when alternative interpretations of data exist. 53 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 5.2. APPLICATION 2 A risk characterization is a necessary part of any Agency report on risk, whether the 3 report is a preliminary one prepared to support allocation of resources toward further study 4 or a comprehensive one prepared to support regulatory decisions. In the former case, the 5 detail and sophistication of the characterization are appropriately small in scale, in the latter 6 case, appropriately extensive. Even if only parts of a risk assessment (hazard and dose- 7 response analyses for instance) are covered in a document, the risk characterization matches 8 the extent of coverage of the document. 9 10 5.3. CONTENT 11 Each of the following subjects should be covered in the risk characterization. 12 13 5.3.1. Presentation 14 The presentation of the results of the assessment should fulfill the aims as outlined in 15 the purpose section above. The summary draws from the key points of the individual 16 characterizations of hazard, dose response, and exposure analysis performed separately under 17 these guidelines. The summary integrates these characterizations into an overall risk 18 characterization (AIHC, 1989). 19 The presentation of results clearly explains the numerical estimates of risk. For 20 example, when estimates of individual risk or population risk (incidence) are used, there are 21 several features of such estimates that risk managers need to understand. They include, for 22 instance, whether the numbers represent average exposure circumstances or maximum 23 potential exposure. The size of the population considered to be at risk and the distribution of 24 individuals' risks within the population should be given. When risks to a sensitive 25 subpopulation have been identified and characterized, the explanation covers the special 26 characterization of this population. 54 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 DRAFT-DO NOT QUOTE OR CITE The presentation identifies and explains the significant issues encountered when the data support alternative interpretation, describes the choice made and the scientific or science policy rationale for the choice. 5.3.2. Strengths and Weaknesses The risk characterization summarizes the kinds of data brought together in the analysis and the reasoning upon which the assessment rests. The description conveys the major strengths and weaknesses of the assessment that arise from availability of data and the current limits of understanding of the process of cancer causation. Health risk is a function of the three elements of hazard, dose response, and exposure. Confidence in the results of a risk assessment is, thus, a function of confidence in the results of the analyses of each element. The important issues and interpretations of data are explained, and the risk manager is given a clear picture of consensus or lack of consensus that exists about significant aspects of the assessment. In addition, the peer-reviewed conclusions of other governmental or international bodies are provided for information. Whenever more than one view of the weight of evidence or dose-response characterization is supported by the data and the policies of these guidelines, and when choosing between them is difficult, the views are presented together. If one has been selected over another, the rationale is given; if not, both are presented as plausible alternative results. If a quantitative uncertainty analysis of data is appropriate, it is presented in the risk characterization; in any case, qualitative discussion of important uncertainties is appropriate. 55 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE REFERENCES 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 Allen, B. C.; Crump, K. S.; Shipp, A. M. (1988) Correlation between carcinogenic potency of chemicals in animals and humans. Risk Anal. 8: 531-544. American Industrial Health Council (AIHC). (1989) Presentation of risk assessment of carcinogens. Report of a Washington, D.C., ad hoc study group on risk assessment presentation. Ames, B. N.; Gold, L. S. (1990) Too many rodent carcinogens: mitogenesis increases mutagenesis. Science 249: 970-971. Ashby, J.; Tennant, R. W. (1991) Definitive relationship among chemical structure, carcinogenicity and mutagenicity for 301 chemicals tested by the U.S. NTP. Mutat. Res. 257: 229-306. Auger, K. R.; Carpenter, C. L.; Cantley, L. C.; Varticovski, L. (1989a) Phosphatidylinositol 3-kinase and its novel product, phosphatidylinositol 3-phosphate, are present in Saccharomyces cerevisiae. J. Biol. Chem. 264: 20181-20184. Auger, K. R.; Sarunian, L. A.; Soltoff, S. P.; Libby, P.; Cantley, L. C. (1989b) PDGF- dependent tyrosine phosphorylation stimulates production of novel polyphosphoinositides in intact cells. Cell 57: 167-175. Barrett, J. C. (1993) Mechanisms of multistep carcinogenesis and carcinogen risk assessment. Environ. Health Perspect. 100: 9-20. Biggs, P. J., et al. (1993) Mutagenesis 8: 275-283. Birner, et al. (1990) Biomonitoring of aromatic amines. Ill: Hemoglobin binding and benzidine and some benzidine congeners. Arch. Toxicol. 64(2): 97-102. Bishop, J. M. (1991) Molecular themes in oncogenesis. Cell 64: 235-248. Bishop, J. M. (1989) Oncogens and clinical cancer. In: Weinberg, R.A., ed. Oncogens and the molecular origins of cancer. Cold Spring Laboratory Press, pp. 327-358. Bottaro, D. P.; Rubin, J. S.; Faletto, D. L.; Chan, A. M. L.; Kmieck, T. E.; Vande Woude, G. F.; Aaronson, S. A. (1991) Identification of the hepatocyte growth factor receptor as the c-rnet proto-oncogene product. 56 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25' 26 27 28 29 30 31 32 33' 34 35 36 37 38 39 40 41 42 Bouk, N. (1990) Tumor angiogenesis: the role of oncogenes and tumor suppressor genes. Cancer Cells 2: 179-183. Callemen, C. J.; Ehrenberg, L.; Jansson, B.; Osterman-Golkar, S.; Segerback, D.; Svensson, K; Wachtmeister, C. A. (1978) Monitoring and risk assessment by means of alkyl groups in hemoglobin in persons occupationally exposed to ethylene oxide. J. Environ. Pathol. Toxicol. 2: 427-442. Callemen, et al. (1986) Cantley, L. C.; Auger, K. R.; Carpenter, C.; Duckworth, B.; Graziani, A.; Kapeller, R.; Soltoff, S. (1991) Oncogenes and signal transduction. Cell 64: 281-302. Chen, C.; Farland, W. (1991) Incorporating cell proliferation in quantitative cancer risk assessment: approaches, issues, and uncertainties. In: Butterworth, B.; Slaga, T.; Farland, W.; McClain, M. (eds.) Chemical induced cell proliferation: Implications for risk assessment. New York: Wiley-Liss; pp. 481-499. Choy, W. N. (1993) A review of the dose-response induction of DNA adducts by aflatoxin 62 and its implications to quantitative cancer-risk assessment. Mutat. Res. 296: 181- 198. Cohen, S. W.; Ellwein, L. B. (1990) Cell proliferation in carcinogenesis. Science 249: 1007- 1011. Collum, R. G.; Alt, F. W. (1990) Are myc proteins transcription factors? Cancer Cells 2: 69-73. Connolly, R. B.; Andersen, M. E. (1991) Biological based pharmacodynamic models: tools for lexicological research and risk assessment. Ann. Rev. Pharmacol. Toxicol. 31: 503-523. Cross, M.; Dexter, T. (1991) Growth factors in development, transformation, and tumorigenesis. Cell 64: 271-280. Crump, K. S.; Hoel, D. G.; Langley, C. H.; Peto, R. (1976) Fundamental carcinogenic processes and their implications for low dose risk assessment. Cancer Res. 36: 2973- 2979. deThe, H.; Lavau, C.; Marchio, A.; Chomiehne, C.; Degos, L.; Dejean, A. (1991) The PML-RARoe fusion mRNA generated by the t (15; 17) translocation in acute promyelocytic leukemia encodes a functionally altered RAR. Cell 66: 675-684. 57 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 D'Souza, R. W.; Francis, W. R.; Bruce, R. D.; Andersen, M. E. (1987) Physiologically based pharmacokinetic model for ethylene chloride and its application in risk assessment. In: Pharmacokinetics in risk assessment. Drinking Water and Health. Vol. 8. Washington, DC: National Academy Press. Enterline, et al. (1987) Fearon, E.; Vogelstein, B. (1990) A genetic model for colorectal tumorigenesis. Cell 61: 959-767. Fidler, I. J.; Radinsky, R. (1990). Genetic control of cancer metastasis. J. Natl. Cancer Inst. 82: 166-168. Flamm, W. G.; Winbush, J. S. (1984) Role of mathematical models in assessment of risk and in attempts to define management strategy. Fund. Appl. Toxicol. 4: S395-S401. Flynn, G. L. (1990) Physicochemical determinants of skin absorption. In: Gerrity, T. R.; Henry, C. J., eds. Principles of route to route extrapolation for risk assessment. New York: Elsevier Science Publishing Co.; pp. 93-127. Forsburg, S. L.; Nurse, P. (1991) Identification of a Gl-type cyclin pugl+ in the fission yeast Schizosaccharomyces pombe. Nature 351: 245-248. Garfmkel, L; Silverberg, E. (1991) Lung cancer and smoking trends in the United States over the past 25 years. Cancer 41: 137-145. Garrett, N. E.; Stack, H. F.; Gross, M. R.; Waters, M. D. (1984) An analysis of the spectra of genetic activity produced by known or suspected human carcinogens. Mutat. Res. 134: 89-111. Gaylor, D. W.; Kodell, R. L. (1980) Linear interpolation algorithm for low-dose risk assessment of toxic substances. J. Environ. Pathol. Toxicol. 4: 305-312. Gerrity, T. R.; Henry, C., eds. (1990) Principles of route to route extrapolation for risk assessment. New York: Elsevier Science Publishing Co. Gillette, J. R. (1983) The use of pharmacokinetics in safety testing. Safety evaluation and regulation of chemicals 2. 2nd Int. Conf., Cambridge, MA; pp. 125-133. Greenland, S. (1987) Quantitative methods in the review of epidemiologic literature. Epidemiol. Rev. 9:1-29. 58 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 Hammand, E. C. (1966) Smoking in relation to the death rates of one million men and women. In: Haenxzel, W., ed. Epidemiological approaches to the study of cancer and other chronic diseases. National Cancer Institute Monograph No. 19 Washington DC. ' ' Harris, C. C. (1989) Interindividual variation among humans in carcinogen metabolism, DNA adduct formation and DNA repair. Carcinogenesis 10: 1563-1566. Harris, C. C.; Hollstein, M. (1993) Clinical implications of the p53 tumor suppressor gene N. Engl. J. Med. 329: 1318-1327. Harris, H. (1990) The role of differentiation in the suppression of malignancy. J Cell Sci 97: 5-10. Haseman, J. K. (1983) Issues: a reexamination of false-positive rates for carcinogenesis studies. Fund. Appl. Toxicol. 3: 334-339. Haseman, J. K. (1984) Statistical issues in the design, analysis and interpretation of animal carcinogenicity studies. Environ. Health Perspect. 58: 385-392. Haseman, J. K. (1985) Issues in carcinogenicity testing: dose selection. Fund. Appl. Toxicol 5: 66-78. Haseman, J. K. (1990) Use of statistical decision rules for evaluating laboratory animal carcinogenicity studies. Fund. Appl. Toxicol. 14: 637-648. Haseman, J. K.; Huff, J.; Boorman, G.A. (1984) Use of historical control data in carcinogenicity studies in rodents. Toxicol. Pathol. 12: 126-135. Hattis, D. (1990) Pharmacokinetic principles for dose-rate extrapolation of carcinogenic risk from genetically active agents. Risk Anal. 10: 303-316. Hayward, N. K.; Walker, G. J.; Graham, W.; Cooksley, E. (1991) Hepatocellular carcinoma mutation. Nature 352: 764. Herschman, H. R. (1991) Primary response genes induced by growth factors or promoters. Ann. Rev. Biochem. 60: 281-319. Hoel, D. G. (1980) Incorporation of background in dose-response models. Fed. Proc Fed Am. Soc. Exp. Biol. 39: 73-75. 59 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 Hollstein, M.; Sidransky, D.; Vogelstein, B.; Harris, C. C. (1991) p53 mutations in human cancers. Science 253: 49-53. Hsu, I. C.; Metcaff, R. A.; Sun, T.; Welsh, J. A.; Wang, N. L; Harris, C. C. (1991) Mutational hotspot in human hepatocellular carcinomas. Nature 350: 427-428. Hulka, B. S.; Margolin, B. H. (1992) Methodological issues in epidemiologic studies using biological markers. Am. J. Epidemiol. 135: 122-129. Hunter, T. (1991) Cooperation between oncogenes. Cell 64: 249-270. Jarabek, (1994) in press Kakizuka, A.; Miller, W. H.; Umesono, K.; Warrell, R. P.; Frankel, S. R.; Marty, V. V. V. S.; Dimitrovsky, E.; Evans, R. M. (1991). Chromosomal translocation t(15;17) in human acute promyelocyte leukemia fuses RARoc with a novel putative transcription factor, pml. Cell 66: 663-674. Kelsey, J. L.; Thompson, W. D.; Evans, A. S. (1986) Methods in observational epidemiology. New York: Oxford University Press. Kodell, R. L.; Park, C. N. (in press) Linear extrapolation in cancer risk assessment. ILSI Risk Science Institute. . Krewski, D.; Murdoch, D. J.; Withey, J. R. (1987). The application of pharmacokinetic data in carcinogenic risk assessment. In: Pharmacokinetics in risk assessment. Drinking Water and Health. Vol. 8. Washington, DC: National Academy Press; pp. 441-468 Krewski, D.; Brown, C.; Murdoch, D. (1984) Determining "safe" levels of exposure: Safely factors of mathematical models. Fund. Appl. Toxicol. 4: S383-S394. Kumar, R.; Sukumar, S.; Barbacid, M. (1990) Activation of ras oncogenes preceding the onset of neoplasia. Science 248: 1101-1104. Lilienfeld, A. M.; Lilienfeld, D. (1979) Foundations of epidemiology, 2nd ed. New York: Oxford University Press. Lutz, W. K. (1990) Dose-response relationship and low dose extrapolation in chemical carcinogenesis. Carcinogenesis 11: 1243-1247. Mailer, J. L. (1991) Mitotic control. Curr. Opin. Cell Biol. 3:269-275. 60 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 ' 30 31 32 33 34 35 36 37 38 39 40 41 DRAFT-DO NOT QUOTE OR CITE Mann, C. (1990) Meta-analysis in a breech. Science 249: 476-480. Mausner, J. S.; Kramer, S. (1985) Epidemiology, 2nd ed. Philadelphia: W. B. Saunders Company. McConnell, E. E.; Solleveld, H. A.; Swenberg, J. A.; Boorman, G. A. (1986) Guidelines for combining neoplasms for evaluation of rodent carcinogenesis studies J Nafl Cancer Inst. 76: 283-289. McMichael, A. J. (1976) Standardized mortality ratios and the "healthy worker effect": scratching beneath the surface. J. Occup. Med. 18: 165-168. Meyn, M. S. (1993) High spontaneous intrachromosomal recombination rates in ataxia- telangiectasia. Science 260: 1327-1330. Moolgavkar, S. H.; Knudson, A. G. (1981) Mutation and cancer: a model for human carcinogenesis. J. Natl. Cancer Inst. 66: 1037-1052. Monro, A. (1992) What is an appropriate measure of exposure when testing drugs for carcinogenicity in rodents? Toxicol. Appl. Pharmacol. 112: 171-181. Naeve, G. S.; Sharma, A.; Lee, A. S. (1991) Temporal events regulating the early phases of the mammalian cell cycle. Curr. Opin. Cell Biol. 3: 261-268. National Research Council (NRC). (1983) Risk assessment in the federal government: managing the process. Committee on the Institutional Means for Assessment of Risks to Public Health, Commission on Life Sciences, NRC. Washington, DC: National Academy Press. National Research Council (NRC). (1993a) Pesticides in the diets of infants and children. Committee on Pesticides in the Diets of Infants and Children, Commission on Life Sciences, NRC. Washington, DC: National Academy Press. National Research Council (NRC). (1993b) Issues in risk assessment. Committee on Risk Assessment Methodology, Commission on Life Sciences, NRC. Washington, DC: National Academy Press. National Research Council (NRC). (1994) Science and judgment in risk assessment. Committee on Risk Assessment of Hazardous Air Pollutants, Commission on Life Sciences, NRC. Washington, DC: National Academy Press. 61 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 National Toxicology Program (NTP). (1984) Report of the Ad Hoc Panel on Chemical Carcinogenesis Testing and Evaluation of the National Toxicology Program, Board of Scientific Counselors. Washington, DC: U.S. Government Printing Office. 1984-421- 132: 4726. Nebreda, A. R.; Martin-Zanca, D.; Kaplan, D. R.; Parada, L. P.; Santos, E. (1991) Induction by NGF of meiotic maturation ofxenopus oocytes expressing the trk proto- oncogene product. Science 252: 558-561. Nowell, P. (1976) The clonal evolution of tumor cell populations. Science 194: 23-28. Nunez, G.; Hockenberry, D.; McDonnell, J.; Sorenson, C. M.; Korsmeyer, S. J. (1991). Bcl-2 maintains B cell memory. Nature 353: 71-72. Office of Science and Technology Policy (OSTP). (1985) Chemical carcinogens: review of the science and its associated principles. Federal Register 50: 10372-10442. Peto, J. (1992) Meta-analysis of epidemiological studies of carcinogenesis. In: Mechanisms of carcinogenesis in risk assessment. IARC Sci. Pubs. No. 116, Lyon, France: International Agency for Research on Cancer: 571-577. Peto, J.; Darby, S. (1994) Radon risk reassessed. Nature 368: 97-98. Peto, R. (1978) Carcinogenic effects of chronic exposure to very low levels of toxic substances. Environ. Health Perspect. 22: 155-161. Pitot, H.; Dragan, Y. P. (1991) Facts and theories concerning the mechanisms of carcinogenesis. FASEB J. 5: 2280-2286. Portier, C. (1987) Statistical properties of a two-stage model of carcinogenesis. Environ. Health Perspect. 76: 125-131. Raff, M. C. (1992) Social controls on cell survival and cell death. Nature 356: 397-400. Rail, D. P. (1991) Carcinogens and human health: part 2. Science 251: 10-11. Rothman, K. T. (1986) Modern epidemiology. Boston: Little, Brown and Company. Salomon, D. S.; Kim, N.; Saeki, T.; Ciardiello, F. (1990) Transforming growth factor a- an oncodevelopmental growth factor. Cancer Cells 2: 389-397. 62 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 Schneider, C.; Gustincich, S.; DelSal, G. (1991) The complexity of cell proliferation control in mammalian cells. Curr. Opin. Cell Biol. 3: 276-281. Schuller, H. M. (1991) Receptor-mediated mitogenic signals and lung cancer. Cancer Cells 3: 496-503. Sidransky, D.; Mikkelsen, T.; Schwechheimer, K.; Rosenblum, M. L.; Cavanee, W.; Vogelstein, B. (1992) Clonal expansion of p53 mutant cells is associated with brain tumor progression. Nature 355: 846-847. Sidransky, D.; Von Eschenbach, A.; Tsai, Y.C.; Jones, P.; Summerhayes, I.; Marshall, P.; Paul, M.; Green, P.; Hamilton, P.P.; Vogelstein, B. (1991) Identification of p53 gene mutations in bladder cancers and urine samples. Science 252: 706-710. Srivastava, S.; Zou, Z.; Pirollo, K.; Blattner, W.; Chang, E. (1990) Germ-line transmission of a mutated p53 gene in a cancer-prone family with Li-Fraumeni syndrome. Nature 348(6303): 747-749. Stanbridge, E. J.; Cavenee, W. K. (1989) Heritable cancer and tumor suppressor genes: A , tentative connection. In: Weinberg, R. A., ed. Oncogenes and the molecular origins of cancer, p. 281. Stiteler, W. H.; Knauf, L. A.; Hertzberg, R. C.; Schoeny, R. S. (1993) A statistical test of compatibility of data sets to a common dose-response model. Reg. Tox. Pharmacol. 18: 392-402. Strausfeld, U.; Labbe, J. C.; Fesquet, D.; Cavadore, J. C.; Dicard, A.; Sadhu, K.; Russell, P.; Dor'ee, M. (1991) Identification of a Gl-type cyclin pud 4- in the fission yeast Schizosaccharomyces pombe. Nature 351: 242-245. Sukumar, S. (1989) ras oncogenes in chemical carcinogenesis. Curr. Top. Microbiol. Immunol. 148: 93-114. Sukumar, S. (1990) An experimental analysis of cancer: role of ras oncogenes in multistep carcinogenesis. Cancer Cells 2: 199-204. Swenberg, J. A.; Richardson, F. C.; Boucheron, J. A.; Deal, F. H.; Belinsky, S. A.; Charbonneau, M.; Short, B. G. (1987) High to low dose extrapolation: critical determinants involved in the dose-response of carcinogenic substances. Environ. Health Perspect. 76: 57-63. 63 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 Tanaka, K.; Oshimura, M.; Kikiuchi, R.; SeM, M.; Hayashi, T; Miyaki, M. (1991) Suppression of tumorigenicity in human colon carcinoma cells by introduction of normal chromosome 5 or 18. Nature 349: 340-342. Taylor, J. H.; Watson, M. A.; Devereux, T. R.; Michels, R. Y.; Saccomanno, G.; Anderson, M. (1994) Lancet 343: 86-87. Tennant, R. W.; Ashby, J. (1991) Classification according to chemical structure, mutagenicity to Salmonella and level of carcinogenicity of a further 39 chemicals tested for carcinogenicity by the U.S. National Toxicology Program. Mutat. Res. 257: 209-277. Thompson, T. C.; Southgate, J.; Kitchener, G.; Land, H. (1989) Multistage carcinogenesis induced by ras and wye oncogenes in a reconstituted organ. Cell 56: 917-3183. Tobey, R. A. (1975) Different drugs arrest cells at a number of distinct stages in G2. nature 254: 245-247. Tomatis, L.; Aitio, A.; Wilbourn, J.; Shuker, L. (1989) Jpn. J, Cancer Res. 80: 795-807. Travis, C. C.; McClain, T.W.; Birkner, P. D. (1991) Diethylnitrosamine-induced hepatocarcinogenesis in rats: a theoretical study. Toxicol. Appl. Pharmacol. 109: 289- 309. U.S. Environmental Protection Agency. (1983a) Good laboratory practices standards- toxicology testing. Federal Register 48: 53922. U.S. Environmental Protection Agency. (1983b) Hazard evaluations: humans and domestic animals. Subdivision F. Springfield, VA: NTIS. PB 83-153916. U.S. Environmental Protection Agency. (1983c) Health effects test guidelines. Springfield, VA: NTTS. PB 83-232984. U.S. Environmental Protection Agency. (1984) Estimation of the public health risk from exposure to gasoline vapor via the gasoline marketing system. Washington, DC: Office of Health and Environmental Assessment. U.S. Environmental Protection Agency. (1986a) Health assessment document for beryllium. Washington, DC: Office of Health and Environmental Assessment. EPA/600/ . 64 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 DRAFT-DO NOT QUOTE OR CITE U.S. Environmental Protection Agency. (1986b) The risk assessment guidelines of 1986.. Washington, DC: Office of Health and Environmental Assessment. EPA/600/8- 87/045. U.S. Environmental Protection Agency. (1989a) Interim procedures for estimating risks associated with exposures to mixtures of chlorinated dibenzo-p-dioxins and - dibenzofurans (CDDs and CDFs) and 1989 update. Washington, DC: Risk Assessment Forum. EPA/625/3-89/016. U.S. Environmental Protection Agency. (1989b) Workshop on EPA guidelines for carcinogen risk assessment. Washington, DC: Risk Assessment Forum. EPA/625/3-89/015. U.S. Environmental Protection Agency. (1989c) Workshop on EPA guidelines for carcinogen risk assessment: use of human evidence. Washington, DC: Risk Assessment Forum EPA/625/3-90/017. U.S. Environmental Protection Agency. (199la) Pesticide Assessment Guidelines: Subdivision F, hazard evaluation: human and domestic animals. Series 84, Mutagenicity. PB91-158394, 540/09-91-122. Office of Pesticide Programs. U.S. Environmental Protection Agency. (1991b) Alpha-2u-globulin: association with chemically induced renal toxicity and neoplasia in the male rat. Washington, DC: Risk Assessment Forum. EPA/625/3-91/019F. U.S. Environmental Protection Agency. (199 Ic) Workshop report on toxicity equivalency factors for polychlcrinated biphenyl congeners. Washington, DC: Risk Assessment Forum. EPA/625/3-91/020. U.S. Environmental Protection Agency. (1991f) Guidelines for developmental toxicity risk assessment. Federal Register 56(234): 63798-63826. U.S. Environmental Protection Agency. (1992a) Guidelines for exposure assessment. Federal Register 57(104): 22888-22938 U.S. Environmental Protection Agency. (1992b) Draft report: A cross-species scaling factor for carcinogen risk assessment based on equivalence of mg/kg3/4/day. Federal Register 57(109): 24152-24173. U.S. Environmental Protection Agency. (1992c, August) Health assessment for 2,3,7,8- tetrachlorodibenzo-j3>-dioxin (TCDD) and related compounds (Chapters 1 through 8). Workshop Review Drafts. EPA/600/AP-92/001a through OOlh. 65 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 U.S. Environmental Protection Agency. (1994) Estimating exposure to dioxin-like compounds. Prepared by the Office of Health and Environmental Assessment, Office of Research and Development, Washington, DC. External Review. Draft, 3 vol. EAP/600/6-88/005Ca, Cb, Cc. U.S. Environmental Protection Agency, (in press) Inhalation RfC methodology: an inhalation dosimetry method. Vahakangas, K. H.; Samet, J. M.; Metealf, R. A.; Welsh, J. A.; Bennett, W. P.; Lane, D. P.; Harris, C. C. (1992) Mutation of p53 and ras genes in radon-associated lung cancer from uranium miners. Lancet 339: 576-578. Vainio, H.; Magee, P.; McGregor, D.; McMichael, AJ. (1992) Mechanisms of carcinogenesis in risk identification. IARC Sci. Pubs. No. 116. Lyon, France, International Agency for Research on Cancer. Van Sittert, N. J.; De Jong, G.; Clare, M.G.; Davies, R.; Dean, B.J.; Wren, L.R.; Wright, A.S. (1985) Cytogenetic, immunological, and hematological effects in workers in an ethylene oxide manufacturing plant. Br. J. Indust. Med. 42:19-26. Varmus, H. (1989) An historical overview of oncogenes. Cold Spring Harbor Laboratory Press, pp. 3-44. Vater, S. T.; McGinnis, P. M.; Schoeny, R. S.; Velazquez, S. (1993) Biological considerations for combining carcinogenicity data for quantitative risk assessment. Reg. Toxicol. Pharmacol. 18: 403-418. Weinberg, R. A. (1989) Oncogenes, antioncogenes, and the molecular bases of multistep carcinogenesis. Cancer Res. 49: 3713-3721. Wellstein, A.; Lupu, R.; Zugmaier, G.; Flamm, S. L.; Cheville, A.L.; Bovi, P. D.; Basicico, C.; Lippman, M. E.; Kern, F.G. (1990) Autocrine growth stimulation by secreted Kaposi fibroblast growth factor but not by endogenous basic fibroblast growth factor. Cell Growth Differ. 1: 63-71. Woo, Y. T.; Arcos, J. C. (1989) Role of structure-activity relationship analysis in evaluation of pesticides for potential carcinogenicity. In: Ragsdale, N. N.; Menzer, R. E., eds. Carcinogenicity and pesticides: principles, issues, and relationship. ACS Symposium Series No. 414. San Diego: Academic Press; pp. 175-200. Wyzga, R. E. (1988) The role of epidemiology in risk assessments of carcinogens. Adv. Mod. Environ. Toxicol. 15: 189-208. 66 07/25/94 ------- 1 2 3 4 5 6 7 8 DRAFT-DO NOT QUOTE OR CITE Yamasaki, H. (1990) Gap junctional intercellular communication and carcinogenesis. Carcinogenesis 11: 1051-1058. Zeise, L.; Wilson, R.; Crouch, E. A. C. (1987) Dose-response relationships for carcinogens: a review. Environ. Health Perspect. 73: 259-308. Zhang, K.; Papageorge, A. G.; Lowry, D. R. (1992) Mechanistic aspects of signalling through ras in NIH 3T3 cells. Science 257: 671-674. 67 OH/25194 ------- DRAFT-DO NOT QUOTE OR CITE 1 APPENDIX A: EXAMPLES OF NARRATIVES 2 3 Narrative # 1 4 CAS#XXXX 5 6 The main kinds of evidence available for consideration of potential human 7 carcinogenicity of this metal conjugated aliphatic phosphonate are animal studies of 8 tumorigenicity, short-term tests of genotoxicity and animal studies of toxicity. No human 9 data are available. The animal studies provide a reasonable basis for understanding the 10 reasons for carcinogenic effects of the compound in laboratory animals and their 11 applicability for judging potential human carcinogenicity. 12 Metal conjugate caused a statistically significant increase in the incidence of urinary 13 bladder hyperplasia and tumors (urinary bladder transitional cell papillomas and carcinomas) 14 in male, but not female, Charles River CD rats at 30,000 ppm in the diet in a long-term 15 study. Some high dose animals had urinary tract stones. No tumors, hyperplasia or stones 16 were seen in two lower dose groups (2,000 and 8,000 ppm) in the same study. A study in 17 Charles River CD-I male and female mice at similar dietary doses to those hi the rat study 18 showed no tumor response or urinary tract effects. A two-year study in dogs at doses up to 19 40,000 ppm showed no urinary tract effects. A major metabolite of metal conjugate that 20 contains the phosphonate moiety caused no tumor response or urinary tract effects in a well- 21 conducted two-year bioassay hi rats. 22 In a 90-day study hi the same rat strain, observations of urinary tract function and 23 histopathology revealed a pattern of physiological changes hi both males and females at doses 24 in the same range as the chronic bioassay (8,000, 30,000, 50,000 ppm). These included 25 proteinuria, increased blood phosphorus and urinary calcium, diuresis, and a sharp drop in 26 urine pH. The calcium effects and diuresis were seen only at doses at or above 30,000 ppm, 27 and were more pronounced hi the males. Histopathology evaluation revealed a pattern of 28 cytotoxicity hi the urinary tract with hyperplasia hi the urinary bladder that was seen at doses 68 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 DRAFT-DO NOT QUOTE OR CITE at or above 30,000 ppm and significantly more extensive in the males. In males, but not females, urinary bladder stones containing calcium and phosphorus were observed in association with bladder hyperplasia. Treatment of male rats for at least eight weeks followed by a return to normal diet for up to 21 weeks showed significant reversibility in the frequency of both stones and bladder hyperplasia. A three-generation reproductive study hi rats showed urinary tract toxicity consistent with the results of the 90-day study: stones, cytotoxicity, and hyperplasia at 24,000 ppm; no urinary tract effects at 6,000 or 12,000 ppm. Short-term studies including an in vivo mouse micronucleus test, gene mutation tests in Salmonella, and DNA damage tests hi yeast indicate that metal conjugate is not genotoxic. Confidence hi the extent and nature of this data set is considered medium for supporting this conclusion. Metal conjugate is not hi a structural chemical category characterized as having carcinogenic effects hi the urinary tract. The results of the tumorigenicity, toxicity, and genotoxicity studies support a conclusion that the male rat tumors were the result of high dose toxicity hi the urinary tract. It is clear that formation of stones hi the male rat caused profound hyperplasia which, in turn, led to tumor formation. The corollary would be: No stones, no tumors. It is concluded that metal conjugate is likely to be carcinogenic to humans only under conditions that repeat the pattern of effects seen in the male rat at high doses and not likely without those conditions. Likewise, it is unlikely to be a human carcinogen under conditions hi which bladder stone formation is unlikely to develop. Certain observations suggest that humans are much less sensitive to this particular carcinogenic process than are male rats. First, of the several species and sexes studied at high doses, only the male rat responded with effects hi the urinary tract. Second, in rodents, bladder stones of any composition including inert materials like glass frequently lead to tumor development. Third, there is little indication that humans with bladder stones have a corresponding tumor response in the bladder. As the data support the view that carcinogenicity would be secondary to toxicity of the chemical, it is recommended that metal conjugate dose-response and human exposure 69 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 DRAFT-DO NOT QUOTE OR CITE analyses of high exposure scenarios be developed to support a margin of exposure approach to human risk characterization. Uncertainties include (1) incomplete data on the relationship between stone formation and neoplasia in the human bladder, (2) whether the observations in the rat are biologically relevant to humans. Overall, the conclusions drawn about the process of carcinogenesis in the male rat are strongly based. The inference that this process would occur in humans is weak. Narrative # 2 CAS# XXX The potential for human carcinogenicity of aromatic alkene (ar-alkene) cannot be determined from the available data. However, evaluation of animal studies and short-term studies on ar-alkene and its metabolite suggest an hypothesis as to potential carcinogenicity that requires further research, and the chemical is a testing candidate. Epidemiologic studies performed on ar-alkene are considered inadequate for evaluation. In a two-year gavage bioassay hi F344/N rats and B6C3F1 mice, male mice showed a statistically significant, increased incidence of combined adenomas and carcinomas of the lung. However, the NTP, sponsor of the bioassay, considered the response equivocal because of experimental design problems. Two inhalation studies hi Sprague-Dawley female rats showed increased incidence of mammary tumors which was considered equivocal because of lack of a dose-response trend and high historical incidence of this response. Studies hi these rats by gavage or ip injection were negative. The metabolite is rapidly broken down at the pH of the glandular stomach; thus, no internal dose is expected from administration by this route. Ar-alkene is mutagenic hi bacterial test systems only with metabolic activation. Both positive and negative responses were seen hi mammalian cells in vitro. The metabolite is 70 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE clearly mutagenic in numerous in vitro tests. Both ar-alkene and its metabolite produce inconsistent results in in vivo test systems. Ar-alkene is fat soluble and readily absorbed by all exposure routes. It is distributed generally in the body. The kinetics of conversion of ar-alkene to its metabolite and the bioavailability of the metabolite have not been well characterized. ,If ar-alkene has carcinogenic potential, the long-term animal studies and the mutagenicity tests suggest that this potential will depend on the bioavailability and activity of the metabolite. At present, the data on these factors are not strong enough to support a conclusion. Depending on the pharmacokinetics of ar-alkene and its metabolite, carcinogenicity may result from bioavailability of the metabolite at the site of administration or at internal tissues sites. Further study, preferably by inhalation, is needed. Narrative #3 CAS# XXX Information on this chlorinated cyclic hydrocarbon includes case histories of exposed humans, tumor findings in the liver of mice but not hi rats, and the absence of genotoxic effects hi short-term tests. Some persons with aplastic anemia have reported exposure to the chemical; some of these people have also developed leukemia. No other information on humans exists as to potential carcinogenic effects. Multiple feeding studies in males and females of several strains of mice indicate that exposure to the chemical leads to hepatocellular tumors. Tumors were not found at other anatomical sites in mice or at any site in two chronic rat feeding studies. The chemical is a potent inducer of liver microsomal enzymes at doses associated with liver cancer in mice. Chemical administration leads to liver enlargement due hi part to increases in liver cell size; this enlargement persists until tumor development. The extent to which the chemical leads to increases in cell proliferation has not been established. Some 71 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 studies indicate that the chemical may increase the amount of reactive oxygen radicals and 2 lipid peroxidation at high doses. It promotes liver tumors in mice following initiation by a 3 genotoxic agent at doses associated with tumors from exposure to the chemical alone. 4 Testing for genotoxicity has generally led to negative findings, and there are no 5 structural alerts. The chemical does not increase the frequency of gene mutations in bacteria, 6 fungi and algae; there is a questionable increase in cultured mouse lymphoma cells. No or 7 marginal increases in structural aberrations were noted in two studies of cultured Chinese 8 hamster cells and in a study of Syrian hamster bone marrow cells in vivo; there was no 9 increase in sister chromatid exchanges in the cultured hamster cells. 10 Uncertainties include the finding that the only indication of a carcinogenic response 11 comes from the mouse liver, the most common cancer site in rodents and the one that is 12 uniquely positive with many other chlorinated hydrocarbons. Other sites in the mouse and 13 all of them in the rat fail to indicate a carcinogenic potential. There is no significant 14 genotoxic potential or structural indicators of carcinogenicity. Cancer responses are 15 associated with high doses that lead to induction of liver microsomal enzymes and potential 16 reactive oxygen species. 17 It is hard to interpret the carcinogenic findings; there are cancer responses in the 18 mouse liver, but they occur at high doses of this nongenotoxic compound that are associated 19 with demonstrable effects on the liver, doses that are far above anticipated human exposures. 20 Overall, the evidence is on the border between a likely human carcinogen and cannot be 21 determined. Further mechanistic work is needed to differentiate these alternatives. Until 22 such time, it will be assumed that the chemical is likely to be a human carcinogen. Cancer 23 risks should be evaluated by a margin of exposure evaluation. 24 72 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 DRAFT-DO NOT QUOTE OR CITE Case Study # 4 CAS# XXX There are no tumor data on this bis-benzenamine. Evaluation of its carcinogenic potential comes from tumor studies on structural analogues and toxicity information on the chemical and its analogues. The chemical is considered likely to be carcinogenic to humans by all routes of exposure. Mechanistic information helps to associate findings in animals with those in exposed humans. Close structural analogues produce thyroid follicular cell tumors and hepatocellular tumors hi rats and mice following ingestion. The thyroid tumors are associated with known perturbations in thyroid-pituitary functioning. These compounds inhibit the accumulation of iodide into the thyroid gland, apparently due to inhibition of the enzyme that synthesizes the thyroid hormones. Accordingly, blood levels of thyroid hormones decrease, which induce the pituitary gland to produce more TSH, a hormone that stimulates the thyroid to produce more of its hormones. The thyroid gland becomes larger due to increases in the size of individual cells and their proliferation and, upon chronic administration, tumors develop. Thus, thyroid tumor development in rats is significantly influenced t>y disruption in the thyroid-pituitary axis. Human and rat thyroid glands respond similarly to short-term exposure. Worker exposure has not been well characterized or quantified, but recent medical monitoring of workers exposed over a several year period has uncovered decreases in thyroid hormones, increases hi TSH and symptoms of hypothyroidism. A urinary metabolite of the chemical has been monitored in the workers. Its concentration was only about 2-fold lower than that hi rats receiving the chemical for 28-days, which led to similar changes in thyroid and pituitary hormones. In addition, the dose of the chemical given to rats hi this study was essentially the same as that of an analogue that had produced thyroid tumors hi rats. Although the human thyroid responds as does that of rodents following limited exposure, it is 73 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 not well established that thyroid-pituitary imbalance leads to cancer in humans as it does in 2 rodents. 3 The chemical is an aromatic amine, a member of a class of chemicals that has 4 regularly produced carcinogenic effects in rodents; some have produced cancer in humans. 5 Structural analogues are genotoxic; they produce gene mutations in cultured bacteria and 6 mammalian cells and structural chromosome aberrations and DNA damage in mammalian 7 cultured cells; these are characteristics of chemicals that produce cancer hi multiple 8 anatomical sites and species. The genotoxic effects could influence the development of 9 tumors in the thyroid and liver. 10 Given the information at hand on potential thyroid and liver tumors hi rodents among 11 close structural analogues, it is reasonable to conclude that the chemical is likely to be a 12 human carcinogen. Biological information on the compound leads one hi differing directions 13 as to how to quantitate potential cancer risks. The information on disruption on thyroid- 14 pituitary status argues for using a margin of exposure evaluation, whereas that bearing on the 15 chemical's being an aromatic amine and having gene and structural chromosome mutation 16 activity, coupled with a lack of mode of action information on the liver tumors, points 17 toward a low dose linear approach. In recognition of these differences, it is appropriate to 18 quantitate thyroid tumors using both a nonlinear or margin of exposure and linear techniques; 19 liver tumors should be evaluated by the linear approach. Tumor data on the close analogues 20 should be utilized in projections of potential risks from exposure. Given the absence of 21 direct tumor information on the chemical per se, potential risks should only be presented as 22 screening estimates with rough order-of-magnitude values. The chemical can be absorbed by 23 the oral, inhalation and dermal routes of exposure. 24 Uncertainties include (1) the lack of carcinogenicity studies on the chemical, (2) the 25 employment of data on structural analogs, (3) the lack of established information on the 26 relevance of thyroid-pituitary unbalance for human carcinogenicity, and (4) the different 27 potential mechanisms that may influence tumor development and potential risks. Overall, 28 this is a strong inferential case for potential human carcinogenicity. 74 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE Narrative #5 CAS#XXXX The main evidence available for consideration of potential human carcinogenicity of chlorinated alkene (cl-alkene) includes animal studies of tumorigenicity, short-term tests of genotoxicity, and studies of metabolism and toxicity of this and related chlorinated aliphatics. Epidemiologic studies have been made of workers exposed to cl-alkene, but the studies do not demonstrate either an association or lack of one between cancer and exposure to cl- alkene. The chemical is considered likely to be carcinogenic to humans by all routes of exposure. Administration of cl-alkene by gavage (corn oil vehicle) or by inhalation to B6C3F1 mice in long-term studies caused statistically significant, dose-related increases in hepatocellular carcinomas hi both sexes as well as reduced time-to-tumor hi the gavage studies. Doses were 0, 100, 200 ppm hi the inhalation study; time-weighted average doses hi the gavage studies were 536 or 1072 mg/kg and 300 or 600 mg/kg to male and female mice, respectively. In long-term inhalation studies hi F344/N rats (0, 200, 400 ppm) tubular cell hyperplasia was observed hi male rats and hi one high dose female. Renal tubule neoplasms were observed in male rats. The response was not statistically significant by pairwise comparison, but there was a statistically significant, positive dose-response trend. The historical control incidence of renal tubule tumor in F344/N males is less than 1/2%. The probability of observing two such rare tumors in 50 animals is less than 0.001. A second response observed hi the rat study was an increased incidence of mononuclear cell leukemia. Analyses of the incidence of disease in the study and supplementary analyses of the progression of the disease showed that there was a statistically significant, increased incidence in low dose males and females, in high dose males and, marginally, hi high dose females. In addition, there was a significant increase in extent of the disease hi treated animals of both sexes and a significant shortening of the tune of disease onset in females. In long-term studies hi Sprague-Dawley rats by gavage and inhalation, slight, but not 75 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 DRAFT-DO NOT QUOTE OR CITE statistically significant, increases in kidney tumors were noted; there were no observations of mononuclear cell leukemia. In chronic and subchronic studies, cl-alkene has demonstrated nephrotoxicity in both rats and mice. Cl-alkene has been tested in numerous test systems for mutagenic effects; results indicate that the compound is not genotoxic. Primary metabolites of cl-alkene also test as not genotoxic. A pathway of metabolism identified in the rat generates a secondary mutagenic metabolite in the rat kidney at high doses; this pathway is also active in humans. In general, cl-alkene shares chemical characteristics of related chlorinated ethanes and ethenes in toxicity and tumorigenicity particularly, but not exclusively, toward the mouse liver and rat kidney, and in lack of DNA reactivity or mutagenicity in short-term studies. Similarly, the primary metabolites of these compounds are not notably genotoxic. More than one mechanism has been proposed by which cl-alkene might cause each tumor response; available data do not clearly support any of the various mechanistic views. If a single mechanism accounts for the compound's activity, it is not genotoxicity. The mononuclear leukemia and liver responses in rodents suggest a general accelerating influence on underlying neoplastic processes. The kidney response might be associated with the toxic effects of cl-alkene in the kidney or with mutagenic activity of the secondary metabolite; the data do not show an answer. As a whole the data do not point to the linearity at low doses generally expected of mutagenic compounds, nor are the data strong enough to describe how cl-alkene might have a threshold or non-linear dose response relationship at low doses. It is recommended that two assessments of dose response and of exposure to cl- alkene be developed; one using a model that assumes linearity at low dose, a second to support a margin of exposure approach to human risk characterization. Uncertainties include (1) how the compound acts to influence carcinogenic processes in animals, and how it might act in humans, and (2) implications of the data for dose response assessment. 76 07/25/94 ------- 1 2. 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17, 18 19 20 21 22 23 24 25 26 27 28 DRAFT-DO NOT QUOTE OR CITE Narrative # 6 CAS#XXX Human and animal studies indicate that this metal is likely to be carcinogenic to humans by inhalation and not likely to be carcinogenic via oral or dermal exposure. Absorption of compounds of the metal varies significantly by route of exposure: inhalation > oral > > dermal. Toxic mechanisms are not established, but interference with essential metals and complexes with certain proteins may be operative. There is some evidence for genotoxic effects in cultured cells, but it is not expected that the metal would directly react with DNA. Several epidemiologic studies of workers exposed to the metal in the air have been conducted. Increases in lung and prostatic cancer have been reported in some but not all studies. Interpretation is hindered by the presence of concomitant confounding workplace exposures (e.g., arsenic) and influences of smoking. In the only rodent inhalation study, rats developed significantly increased incidences of lung cancer. In contrast to these findings, multiple feeding and drinking water studies in rodents have failed to indicate carcinogenic responses from daily doses up to over 100-times those in the inhalation study. Increases in testicular interstitial cell tumors have been found in rodent injection studies of soluble metal salts. These compounds lead to necrosis of the testicular germ cell epithelium due to changes in the blood supply with corresponding increases in hormones that stimulate the interstitial cells; such studies are not considered to be applicable to human exposures. Compounds of the metal vary significantly in their solubility: the metal and many salts are very insoluble, whereas a few of the salts are readily soluble. Following inhalation, about 50% of the metal is absorbed into the body, while only about 5% is absorbed from the gut (10-fold lower than via inhalation). Dermal absorption is insignificant. The metal may have genotoxic potential as well -as other toxic properties; both should be considered in evaluating potential risks of exposure. It competes and interferes with the 77 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 functioning of a number of requked metals; it also can bind to certain parts of proteins 2 (sulfhydryl groups) and interfere with their activity. In genotoxicity studies, it is generally 3 negative for gene mutations hi bacteria but positive hi cultured mammalian cells. Although 4 conflicting observations for structural chromosome aberrations have been found in human 5 lymphocytes cultured from exposed workers and hi cell lines exposed in culture, negative 6 findings have been noted hi mouse bone marrow studies. It is not expected that the metal 7 would directly interact with and damage cellular DNA. The evidence does not support a 8 conclusion as to the mode of action; it is recommended that both a linear and nonlinear or 9 margin of exposure approach be taken to dose-response assessment. 10 11 Narrative # 7 12 CAS # XXX 13 14 There is strong evidence that short-chain alkene (SCA) is likely to be a human 15 carcinogen via the inhalation pathway. This conclusion is based on an inhalation animal 16 bioassay (involving both sexes of two species) of SCA showing an unambiguous positive 17 responses and is supported by genotoxicity and metabolic data. We can infer that SCA is 18 also likely to be a human carcinogen for the ingestion and dermal pathways. Some human 19 studies were positive, but methodological problems make them inconclusive. 20 In the animal bioassay, rats and mice were exposed hi air on a schedule simulating 21 occupational exposure. Exposure levels were 625 and 1,250 ppm for mice, and 1,000 and 22 8,000 ppm for rats. Statistically significant increases hi tumor rates were observed hi 23 multiple sites hi both sexes of both species. In the case of the mice, early high mortality due 24 to the tumors was observed. 25 SCA and its metabolites are positive in several genotoxicity tests, including mutage- 26 nicity tests hi bacteria with metabolic activation. Several metabolites of SCA are alkylating 27 agents and are directing acting mutagens hi bacterial tests. Moreover, metabolism experi- 28 ments hi both rats and mice using radio-labeled SCA show binding to nucleoproteins and 78 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 DRAFT-DO NOT QUOTE OR CITE DNA. SCA is also structurally related to other compounds which are positive in animal -bio- assays. There have been five occupational epidemiology studies involving inhalation exposure to SCA. Three of them showed statistically significant risks of cancer; however, hi two of them, workers were also exposed to solvents and the third did not control for smoking. The two negative studies were flawed by low power and possible bias. Thus, the human studies considered together are inconclusive. The direct evidence for carcinogenicity of SCA is based on inhalation exposure. However, since the observed tumors in the animal bioassay occurred at several sites distant from the portal of entry, and since the compound can be absorbed through the gastrointestinal tract and the skin, we assume that it is also likely to be a human carcinogen through these pathways as well. Based on pharmacokinetic, metabolism and genotoxicity data, linear extrapolation of response to low doses is felt to be appropriate. Studies of the pharmacokinetics of SCA hi rats and mice indicate first-order kinetics below a saturation level. Metabolites of SCA are genotoxic and are likely to be carcinogenic, and, although the metabolic pathway is saturable, tumors were observed in the bioassay at exposure levels lower than the saturation level. Although the case for carcinogenicity of SCA is very strong, there are some uncertainties. Since the epidemiological data are inconclusive, we face the usual uncertainties in extrapolating from animals to humans. We have an unusually strong set of mechanistic data supporting linear extrapolation, but uncertainties hi extrapolating from high to low doses remain, as always. 79 07/25/94 ------- DRAFT-DO NOT QUOTE OR CITE 1 Narrative # 8 2 CAS # XXX 3 4 Organophosphate has been tested in several animal bioassays, one of which produced 5 positive results but was possibly flawed, and others which were negative or provide limited 6 support for those results. Genotoxicity and metabolism data also include some that support a 7 concern about carcinogenicity but do not constitute a consistent pattern. No human data are 8 available. Thus, from the data, one cannot determine the likelihood of human 9 carcinogenicity. 10 Three chronic feeding studies have been performed with rats. In a study with 11 Osborne-Mendel rats, statistically significant increases were observed hi combined benign 12 and malignant kidney tumors in both dose groups for males and hi high-dose females, as 13 compared with pooled controls. A statistically significant trend hi malignant pancreatic 14 tumors and benign thyroid tumors was found hi males only. Concerns were raised in audits 15 of the test facility where this study was performed. In a second study with Sprague-Dawley 16 rats, a non-statistically-significant increase hi benign thyroid tumors was observed in males. 17 In a third study with Wistar rats, a statistically significant increasing trend was observed in 18 benign pancreatic tumors in males. 19 Two studies have been performed with mice. In the first, no excess of tumors was 20 found, but the study was flawed. In the second, statistically significant increases were 21 observed of benign lung tumors in males and of malignant lymphomas in females, both at a 22 low dose, but similar increases were not found at two higher doses. 23 Evaluating the rat data together, we see that the strongest result was observed in the 24 Osborne-Mendel rat study, about which doubts have been raised hi an audit. In the Sprague- 25 Dawley rat study, an increase, although not statistically significant, was observed in the same 26 type of tumor hi the same sex as one of the Osborne-Mendel rat responses. Similarly, in the 27 Wistar rat study, a statistically significant trend was observed hi the same sex and site as one 28 of the Osborne-Mendel rat responses, although only benign tumors were seen in this study. 80 07/25/94 ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 DRAFT-DO NOT QUOTE OR CITE The latter two studies have limited weight in reaching a conclusion that the compound is carcinogenic in humans, since only benign tumors were found, but they tend to confirm the results of the first study because the tumors were observed hi the same sites. The sort of problems noted hi the audit of the facility where the Osborne-Mendel rat study was performed - poor record keeping, possible misdosing of the animals - are more likely to mask a positive result than to create a false positive, but they do raise a concern about reliance on the results. The two studies hi mice can be discounted, the first because of flawed methodology, the second because the response was seen only at one low dose. In genotoxicity testing, the compound was found to cause unscheduled DNA synthesis, but other tests were negative or (hi one case) equivocal. This forms the basis for some concern, but is not strong enough or consistent enough to contribute significantly to a judgment on human carcinogenicity. The compound is not structurally related to known human or animal carcinogens. In summary, there are one positive, but possibly flawed, animal feeding study and two other animal studies showing weakly positive results hi the same sites. No human data are available, and the other data are inconclusive. This set of data as a whole does not support a determination of human carcinogenic potential. 81 07/25/94 3S-U.S. GOVERNMENT PRINTING OFFICE: 1994 - 550-064/80017 ------- ------- ------- m TJ p CD O CO ill f a- 3-g1 "" S CO O 6 ------- |