DRAFT - DO NOT CITE OR QUOTE EPA/635/R-09/001D www.epa.gov/iris TOXICOLOGICAL REVIEW OF 1,1,2,2-TETRACHLOROETHANE (CAS No. 79-34-5) In Support of Summary Information on the Integrated Risk Information System (IRIS) July 2010 NOTICE This document is an Interagency Science Discussion/Final Agency Review draft. This information is distributed solely for the purpose of pre-dissemination peer review under applicable information quality guidelines. It has not been formally disseminated by EPA. It does not represent and should not be construed to represent any Agency determination or policy. It is being circulated for review of its technical accuracy and science policy implications. U.S. Environmental Protection Agency Washington, DC ------- DISCLAIMER This document is a preliminary review draft for review purposes only. This information is distributed solely for the purpose of pre-dissemination peer review under applicable information quality guidelines. It has not been formally disseminated by EPA. It does not represent and should not be construed to represent any Agency determination or policy. Mention of trade names or commercial products does not constitute endorsement or recommendation for use. DRAFT - DO NOT CITE OR QUOTE ------- CONTENTS - TOXICOLOGICAL REVIEW OF 1,1,2,2-TETRACHLOROETHANE (CAS No. 79-34-5) LIST OF TABLES vi LIST OF FIGURES ix LIST OF ABBREVIATIONS AND ACRONYMS x FOREWORD xn AUTHORS, CONTRIBUTORS, AND REVIEWERS xm 1. INTRODUCTION 1 2. CHEMICAL AND PHYSICAL INFORMATION 3 3. TOXICOKINETICS 5 3.1. ABSORPTION 5 3.1.1. Oral Exposure 5 3.1.2. Inhalation Exposure 6 3.2. DISTRIBUTION 6 3.3. METABOLISM 7 3.4. ELIMINATION 10 3.5. PHYSIOLOGICALLY BASED TOXICOKINETIC MODELS 11 4. HAZARD IDENTIFICATION 13 4.1. STUDIES IN HUMANS—EPIDEMIOLOGY, CASE REPORTS, CLINICAL CONTROLS 13 4.1.1. Oral Exposure 13 4.1.2. Inhalation Exposure 13 4.2. SUBCHRONIC AND CHRONIC STUDIES AND CANCER BIOASSAYS IN ANIMALS—ORAL AND INHALATION 16 4.2.1. Oral Exposure 16 4.2.1.1. Subchronic Studies 16 4.2.1.2. Chronic Studies 27 4.2.2. Inhalation Exposure 32 4.2.2.1. Subchronic Studies 32 4.2.2.2. Chronic Studies 34 4.3. REPRODUCTIVE/DEVELOPMENTAL STUDIES—ORAL AND INHALATION 35 4.3.1. Oral Exposure 35 4.3.2. Inhalation Exposure 38 4.4. OTHER DURATION- OR ENDPOINT-SPECIFIC STUDIES 38 4.4.1. Acute Studies (Oral and Inhalation) 39 4.4.1.1. Oral Studies 39 4.4.1.2. Inhalation Studies 40 4.4.2. Short-term Studies (Oral and Inhalation) 42 4.4.2.1. Oral Studies 42 4.4.2.2. Short-term Inhalation Studies 48 4.4.3. Acute Injection Studies 49 111 DRAFT - DO NOT CITE OR QUOTE ------- 4.4.4. Immunotoxicological Studies 50 4.5. MECHANISTIC DATA AND OTHER STUDIES IN SUPPORT OF THE MODE OF ACTION 50 4.5.1. Genotoxicity 51 4.5.2. Short-Term Tests of Carcinogenicity 54 4.6. SYNTHESIS OF MAJOR NONCANCER EFFECTS 56 4.6.1. Oral 56 4.6.1.1. Human Data 56 4.6.1.2. Animal Data 56 4.6.2. Inhalation 62 4.6.2.1. Human Data 62 4.6.2.2. Animal Data 64 4.6.3. Mode of action Information 69 4.7. EVALUATION OF CARCINOGENICITY 70 4.7.1. Summary of Overall Weight of Evidence 70 4.7.2. Synthesis of Human, Animal, and Other Supporting Evidence 71 4.7.3. Mode of action Information 73 4.8. SUSCEPTIBLE POPULATIONS AND LIFE STAGES 74 4.8.1. Possible Childhood Susceptibility 74 4.8.2. Possible Gender Differences 74 4.8.3. Other Susceptible Populations 75 5. DOSE-RESPONSE ASSESSMENTS 76 5.1. ORAL REFERENCE DOSE (RfD) 76 5.1.1. Subchronic Oral RfD 76 5.1.1.1. Choice of Principal Study and Critical Effect—with Rationale and Justification 76 5.1.1.2. Methods of Analysis—Including Models (PBPK, BMD, etc.) 79 5.1.1.3. RfD Derivation—Including Application of Uncertainty Factors (UFs) 81 5.1.2. Chronic Oral RfD 82 5.1.2.1. Choice of Principal Study and Critical Effect - with Rationale and Justification 82 5.1.2.2. Methods of Analysis—Including Models (PBPK, BMD, etc.) 82 5.1.2.3. RfD Derivation—Including Application of UFs 83 5.1.3. RfD Comparison Information 84 5.1.4. Previous RfD Assessment 90 5.2. INHALATION REFERENCE CONCENTRATION (RfC) 90 5.2.1. Choice of Principal Study and Critical Effect—with Rationale and Justification 90 5.2.2. Methods of Analysis—Including Models (PBPK, BMD, etc.) 91 5.2.3. Previous RfC Assessment 92 5.3. UNCERTAINTIES IN THE INHALATION REFERENCE CONCENTRATION (RfC) AND ORAL REFERENCE DOSE (RfD) 92 5.4.1. Choice of Study/Data—with Rationale and Justification 95 5.4.2. Dose-response Data 95 5.4.3. Dose Adjustments and Extrapolation Method(s) 96 5.4.4. Oral Slope Factor and Inhalation Unit Risk 98 5.4.5. Uncertainties in Cancer Risk Values 98 5.4.6. Previous Cancer Assessment 101 iv DRAFT - DO NOT CITE OR QUOTE ------- 6. MAJOR CONCLUSIONS IN THE CHARACTERIZATION OF HAZARD AND DOSE RESPONSE 102 6.1. HUMAN HAZARD POTENTIAL 102 6.2. DOSE RESPONSE 103 6.2.1. Noncancer/Oral 103 6.2.2. Noncancer/Inhalation 109 6.2.3. Cancer/Oral and Inhalation 110 7. REFERENCES 113 APPENDIX A. SUMMARY OF EXTERNAL PEER REVIEW AND PUBLIC COMMENTS AND DISPOSITION A-l APPENDIX B. BENCHMARK DOSE MODELING RESULTS FOR THE DERIVATION OFTHERFD B-A APPENDIX C. BENCHMARK DOSE MODELING RESULTS FOR THE DERIVATION OF THE ORAL SLOPE FACTOR C-l v DRAFT - DO NOT CITE OR QUOTE ------- LIST OF TABLES 2-1. Chemical and physical properties of 1,1,2,2-tetrachloroethane . 4-1. Final body weights (g) and percent change compared to controls in F344/N rats exposed to 1,1,2,2-tetrachloroethane in feed for 14 weeks 17 4-2a. Absolute liver weights (g) and percent change compared to controls in F344/N rats exposed to 1,1,2,2-tetrachloroethane in feed for 14 weeks 17 4-2b. Relative liver weight (mg organ weight/g body weight) and percent change compared to controls in F344/N rats exposed to 1,1,2,2-tetrachloroethane in feed for 14 weeks 18 4-3. Serum chemistry and hematology changes in rats exposed to dietary 1,1,2,2-tetra- chloroethane for 14 weeks 19 4-4. Incidences of selected histopathological lesions in rats exposed to dietary 1,1,2,2-tetra- chlorethane for 14 weeks 21 4-5. Final body weights (g) and percent change compared to controls in B6C3Fi mice exposed to 1,1,2,2-tetrachloroethane in feed for 14 weeks 23 4-6a. Absolute liver weights (g) and percent change compared to controls in B6C3Fi mice exposed to 1,1,2,2-tetrachloroethane in feed for 14 weeks 24 4-6b. Relative liver weights (mg organ weight/g body weight) and percent change compared to controls in B6C3Fi mice exposed to 1,1,2,2-tetrachloroethane in feed for 14 weeks ... 24 4-7. Selected clinical chemistry changes in male mice exposed to dietary 1,1,2,2-tetra- chloroethane for 14 weeks 25 4-8. Selected clinical chemistry changes in female mice exposed to dietary 1,1,2,2-tetra- chloroethane for 14 weeks 26 4-9. Incidences of selected histopathological lesions in mice exposed to dietary 1,1,2,2-tetra- chloroethane for 14 weeks 27 4-10. Incidence of neoplasms in male Osborne-Mendel rats exposed to 1,1,2,2-tetrachloro- ethane in feed for 78 weeks 29 4-11. Incidence of neoplasms in female Osborne-Mendel rats exposed to 1,1,2,2-tetra- chloroethane in feed for 78 weeks 30 4-12. Incidence of hepatocelluar carcinomas in male and female B6C3Fi mice exposed to 1,1,2,2-tetrachloroethane in feed for 78 weeks 32 4-13. Incidence of additional neoplasms in male and female B6C3Fi mice exposed to 1,1,2,2-tetrachloroethane in feed for 78 weeks 32 vi DRAFT - DO NOT CITE OR QUOTE ------- 4-14. Effects of acute (60 minutes) 1,1,2,2-tetrachloroethane treatment on rat liver 39 4-15. Results of in vitro and in vivo genotoxicity studies of 1,1,2,2-tetrachloroethane 51 4-16. Pulmonary adenomas from 1,1,2,2-tetrachloroethane exposure in mice 55 4-17. Pulmonary adenomas from 1,1,2,2-tetrachloroethane exposure in A/St mice 55 4-18. Summary of noncancer results of major studies for oral exposure of animals to 1,1,2,2-tetrachloroethane 57 4-19. Summary of noncancer results of major human studies of inhalation exposure to 1,1,2,2-tetrachloroethane 63 4-20. Summary of noncancer results of major studies for inhalation exposure of animals to 1,1,2,2-tetrachloroethane 65 5-1. Summary of BMD model results for rats exposed to 1,1,2,2-tetrachloroethane in the diet for 14 weeks 80 5-2. Best-fitting BMD model predictions for relative liver weight in rats exposed to 1,1,2,2-tetrachloroethane in the diet for 14 weeks Error! Bookmark not defined. 5-3. Potential PODs with applied UFs and resulting subchronic RfDs 87 5-4. Incidences of hepatocellular carcinomas in B6C3Fi mice used for dose-response assessment of 1,1,2,2-tetrachloroethane 96 5-5. HEDs corresponding to duration-adjusted TWA doses in mice 97 5-6. Summary of human equivalent BMDs and BMDLs based on hepatocellular carcinoma incidence data in female B6C3Fi mice 98 5-7. Summary of uncertainty in the 1,1,2,2-tetrachloroethane cancer risk assessment 99 B-l. BMD modeling results based on incidence of hepatocytocellular vacuolization in male rats exposed to 1,1,2,2-tetrachloroethane in the diet for 14 weeks B-A B-2. BMD modeling results based on incidence of hepatocytocelluar vacuolization in female rats exposed to 1,1,2,2-tetrachloroethane in the diet for 14 weeks B-B B-3. Summary of BMD modeling results based on mean absolute liver weights in male rats administered 1,1,2,2-tetrachloroethane in the diet for 14 weeks ....B-Error! Bookmark not defined. B-4. Summary of BMD modeling results based on mean absolute liver weights in female rats administered 1,1,2,2-tetrachloroethane in the diet for 14 weeks ....B-Error! Bookmark not defined. vn DRAFT - DO NOT CITE OR QUOTE ------- B-5. Summary of BMD modeling results based on mean relative liver weights in male rats administered 1,1,2,2-tetrachloroethane in the diet for 14 weeks B-21 B-6. Summary of BMD modeling results based on mean relative liver weights in female rats administered 1,1,2,2-tetrachloroethane in the diet for 14 weeks ....B-Error! Bookmark not defined. B-7. Summary of BMD modeling results based on mean serum ALT levels in male rats administered 1,1,2,2-tetrachloroethane in the diet for 14 weeks B-27 B-8. Summary of BMD modeling results based on mean serum ALT levels in female rats administered 1,1,2,2-tetrachloroethane in the diet for 14 weeks.... B-Error! Bookmark not defined. B-9. Summary of BMD modeling results on mean serum SDH levels in male rats administered 1,1,2,2-tetrachloroethane in the diet for 14 weeks.... B-Error! Bookmark not defined. B-10. Summary of BMD modeling results on mean serum SDH levels in female rats administered 1,1,2,2-tetrachloroethane in the diet for 14 weeks.. B-Error! Bookmark not defined. B-l 1. Summary of BMD modeling results based on mean serum bile acids in male rats administered 1,1,2,2-tetrachloroethane in the diet for 14 weeks.. B-Error! Bookmark not defined. B-12. Summary of BMD modeling results based on mean serum bile acids in female rats administered 1,1,2,2-tetrachloroethane in the diet for 14 weeks.. B-Error! Bookmark not defined. B-l 3. BMD modeling results for decreases in mean weights of fetuses from rat dams exposed to 1,1,2,2-tetrachloroethane in the diet on GDs 4-20 B-Error! Bookmark not defined. C-l. Data used for dose-response assessment of hepatocellular carcinomas in B6C3Fi mice administered 1,1,2,2-tetrachloroethane via gavage for 78 weeks C-l C-2. Summary of human equivalent BMDs and BMDLs based on hepatocellular carcinoma incidence in B6C3Fi mice administered 1,1,2,2-tetrachloroethane via gavage for 78 weeks C-Error! Bookmarknot defined. C-3. BMD modeling results based on incidence of hepatocellular carcinomas in male B6C3Fi mice administered 1,1,2,2-tetrachloroethane via gavage for 78 weeks C-2 C-4. BMD modeling results based on incidence of hepatocellular carcinomas in female B6C3Fi mice administered 1,1,2,2-tetrachloroethane via gavage for 78 weeks C-Error! Bookmark not defined. vin DRAFT - DO NOT CITE OR QUOTE ------- LIST OF FIGURES 2-1. Structure of 1,1,2,2-tetrachloroethane 3 3-1. Suggested metabolic pathways of 1,1,2,2-tetrachloroethane 7 5-1. Exposure response array for subchronic and chronic oral exposure to 1,1,2,2-tetrachloroethane 86 5-2. PODs for selected endpoints (with critical effect circled) from Table 5-3 with corresponding applied UFs and derived sample subchronic inhalation reference values (RfVs) 88 5-3. PODs for selected endpoints (with critical effect circled) from Table 5-3 with corresponding applied UFs and derived sample chronic inhalation RfVs 89 6-1. PODs for selected endpoints (with critical effect circled) with corresponding applied UFs and derived sample subchronic inhalation RfVs 105 6-2. PODs for selected endpoints (with critical effect circled) from Table 5-3 with corresponding applied UFs and derived sample subchronic inhalation RfVs 108 ix DRAFT - DO NOT CITE OR QUOTE ------- LIST OF ABBREVIATIONS AND ACRONYMS ACTH adrenocorticotropic hormone AIC Akaike's Information Criterion ALP alkaline phosphatase ALT alanine aminotransferase AST aspartate aminotransferase ATP adenosine triphosphate AUC area under the curve BMD benchmark dose BMDL 95% confidence limit (lower bound) on the benchmark dose BMDS benchmark dose software BMR benchmark response CASRN Chemical Abstracts Service Registry Number CHO Chinese hamster ovary CNS central nervous system DEN diethylnitrosamine FEL frank effect level FOB functional observational battery G6Pase glucose-6-phosphatase GD gestation day GST glutathione S-transferase Hb hemoglobin HED human equivalent dose i.p. intraperitoneal IU International units LCso median lethal concentration LDso median lethal dose LOAEL lowest-observed-adverse-effect level mA milliampere NCI National Cancer Institute NOAEL no-observed-adverse-effect level NTP National Toxicology Program PBPK physiologically based pharmacokinetic PBTK physiologically based toxicokinetic PCNA proliferating cell nuclear antigen POD point of departure RBC red blood cell RfC reference concentration RfD reference dose RfV reference value SCE sister chromatid exchange SD standard deviation SDH sorbitol dehydrogenase TWA time-weighted average UDS unscheduled DNA synthesis UF uncertainty factor x DRAFT - DO NOT CITE OR QUOTE ------- U.S. EPA U.S. Environmental Protection Agency WBC white blood cell xi DRAFT - DO NOT CITE OR QUOTE ------- FOREWORD The purpose of this Toxicological Review is to provide scientific support and rationale for the hazard and dose-response assessment in IRIS pertaining to subchronic and chronic exposure to 1,1,2,2-tetrachloroethane. It is not intended to be a comprehensive treatise on the chemical or toxicological nature of 1,1,2,2-tetrachloroethane. The intent of Section 6, Major Conclusions in the Characterization of Hazard and Dose Response, is to present the major conclusions reached in the derivation of the reference dose, reference concentration, and cancer assessment, where applicable, and to characterize the overall confidence in the quantitative and qualitative aspects of hazard and dose response by addressing the quality of the data and related uncertainties. The discussion is intended to convey the limitations of the assessment and to aid and guide the risk assessor in the ensuing steps of the risk assessment process. For other general information about this assessment or other questions relating to IRIS, the reader is referred to EPA's IRIS Hotline at (202) 566-1676 (phone), (202) 566-1749 (fax), or hotline.iris@epa.gov (email address). xn DRAFT - DO NOT CITE OR QUOTE ------- AUTHORS, CONTRIBUTORS, AND REVIEWERS CHEMICAL MANAGER/AUTHOR Martin W. Gehlhaus, M.H.S. National Center for Environmental Assessment U.S. Environmental Protection Agency Washington, DC AUTHORS Ambuja Bale, Ph.D. National Center for Environmental Assessment U.S. Environmental Protection Agency Washington, DC Geoffrey W. Patton, Ph.D. National Center for Environmental Assessment U.S. Environmental Protection Agency Washington, DC Susan Rieth, M.S. National Center for Environmental Assessment U.S. Environmental Protection Agency Washington, DC TedBerner, M.S. National Center for Environmental Assessment U.S. Environmental Protection Agency Washington, DC Karen Hogan, M.S. National Center for Environmental Assessment U.S. Environmental Protection Agency Washington, DC CONTRACTING SUPPORT Mark Osier, Ph.D. Environmental Science Center Syracuse Research Corporation Syracuse, NY Stephen Bosch Environmental Science Center Syracuse Research Corporation Syracuse, NY xiii DRAFT - DO NOT CITE OR QUOTE ------- Marc Odin, M.S. Environmental Science Center Syracuse Research Corporation Syracuse, NY REVIEWERS This document has been provided for review to EPA scientists, interagency reviewers from other federal agencies and White House offices, and the public, and peer reviewed by independent scientists external to EPA. A summary and EPA's disposition of the comments received from the independent external peer reviewers and from the public is included in Appendix A. INTERNAL EPA REVIEWERS Joyce M. Donohue, Ph.D. Office of Water Office of Science and Technology (OST) Health and Ecological Criteria Division (HECD) Lynn Flowers, Ph.D., DABT National Center for Environmental Assessment Office of Research and Development U.S. Environmental Protection Agency Washington, DC Chris Cubbison National Center for Environmental Assessment Office of Research and Development U.S. Environmental Protection Agency Cincinnati, OH xiv DRAFT - DO NOT CITE OR QUOTE ------- 1 1. INTRODUCTION 2 3 4 This document presents background information and justification for the Integrated Risk 5 Information System (IRIS) Summary of the hazard and dose-response assessment of 6 1,1,2,2-tetrachloroethane. IRIS Summaries may include oral reference dose (RfD) and 7 inhalation reference concentration (RfC) values for chronic and other exposure durations, and a 8 carcinogenicity assessment. 9 The RfD and RfC, if derived, provide quantitative information for use in risk assessments 10 for health effects known or assumed to be produced through a nonlinear (presumed threshold) 11 mode of action. The RfD (expressed in units of mg/kg-day) is defined as an estimate (with 12 uncertainty spanning perhaps an order of magnitude) of a daily exposure to the human 13 population (including sensitive subgroups) that is likely to be without an appreciable risk of 14 deleterious effects during a lifetime. The inhalation RfC (expressed in units of mg/m3) is 15 analogous to the oral RfD, but provides a continuous inhalation exposure estimate. The 16 inhalation RfC considers toxic effects for both the respiratory system (portal-of-entry) and for 17 effects peripheral to the respiratory system (extrarespiratory or systemic effects). Reference 18 values are generally derived for chronic exposures (up to a lifetime), but may also be derived for 19 acute (<24 hours), short-term (>24 hours up to 30 days), and subchronic (>30 days up to 10% of 20 lifetime) exposure durations, all of which are derived based on an assumption of continuous 21 exposure throughout the duration specified. Unless specified otherwise, the RfD and RfC are 22 derived for chronic exposure duration. 23 The carcinogenicity assessment provides information on the carcinogenic hazard 24 potential of the substance in question and quantitative estimates of risk from oral and inhalation 25 exposure may be derived. The information includes a weight of evidence judgment of the 26 likelihood that the agent is a human carcinogen and the conditions under which the carcinogenic 27 effects may be expressed. Quantitative risk estimates may be derived from the application of a 28 low-dose extrapolation procedure. If derived, the oral slope factor is a plausible upper bound on 29 the estimate of risk per mg/kg-day of oral exposure. Similarly, an inhalation unit risk is a 30 plausible upper bound on the estimate of risk per ug/m3 air breathed. 31 Development of these hazard identification and dose-response assessments for 32 1,1,2,2-tetrachloroethane has followed the general guidelines for risk assessment as set forth by 33 the National Research Council (NRC, 1983). The U.S. Environmental Protection Agency (U.S. 34 EPA) guidelines and Risk Assessment Forum Technical Panel Reports that may have been used 35 in the development of this assessment include the following: Guidelines for Mutagenicity Risk 36 Assessment (U.S. EPA, 1986), Recommendations for and Documentation of Biological Values 37 for Use in Risk Assessment (U.S. EPA, 1988), Guidelines for Developmental Toxicity Risk 3 8 Assessment (U. S. EPA, 1991 a), Interim Policy for Particle Size and Limit Concentration Issues DRAFT - DO NOT CITE OR QUOTE ------- 1 in Inhalation Toxicity (U.S. EPA, 1994a), Methods for Derivation of Inhalation Reference 2 Concentrations and Application of Inhalation Dosimetry (U.S. EPA, 1994b), Use of the 3 Benchmark Dose Approach in Health Risk Assessment (U.S. EPA, 1995), Guidelines for 4 Reproductive Toxicity Risk Assessment (U.S. EPA, 1996), Guidelines for Neurotoxicity Risk 5 Assessment (U.S. EPA, 1998a), Science Policy Council Handbook. Risk Characterization (U.S. 6 EPA, 2000a), Benchmark Dose Technical Guidance Document (U. S. EPA, 2000b), 7 Supplementary Guidance for Conducting Health Risk Assessment of Chemical Mixtures (U.S. 8 EPA, 2000c), A Review of the Reference Dose and Reference Concentration Processes (U.S. 9 EPA, 2002), Guidelines for Carcinogen Risk Assessment (U.S. EPA, 2005a), Supplemental 10 Guidance for Assessing Susceptibility from Early-Life Exposure to Carcinogens (U.S. EPA, 11 2005b), Science Policy Council Handbook: Peer Review (U. S. EPA, 2006a), and A Framework 12 for Assessing Health Risks of Environmental Exposures to Children (U.S. EPA, 2006b). 13 The literature search strategy employed for this compound was based on the Chemical 14 Abstracts Service Registry Number (CASRN) and at least one common name. Any pertinent 15 scientific information submitted by the public to the IRIS Submission Desk was also considered 16 in the development of this document. The relevant literature was reviewed through May, 2009. 17 Portions of this document were developed under a Memorandum of Understanding, 18 signed November 4, 2004, with the Agency for Toxic Substances and Disease Registry 19 (ATSDR). DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 6 7 8 10 12 14 16 17 18 2. CHEMICAL AND PHYSICAL INFORMATION 1,1,2,2-Tetrachloroethane (1,1,2,2TCE; CASRN 79-34-5) is a synthetic halogenated hydrocarbon that is a colorless, nonflammable liquid at room temperature. It is highly volatile, somewhat soluble in water, and miscible with many organic solvents. The structure of 1,1,2,2-tetrachloroethane is shown below (Figure 2-1), and the chemical and physical properties are presented in Table 2-1. Cl Cl H- Cl -C H Cl Figure 2-1. Structure of l,l?2,2-tetrachloroethane. Table 2-1. Chemical and physical properties of l,l?2,2-tetrachloroethane Characteristic Chemical name Synonym(s) Chemical formula CASRN Molecular weight Color Freezing point Boiling point Density at 20 °C Odor threshold: Water Air Solubility: Water Organic solvents Information 1 , 1 ,2,2-Tetrachloroethane Acetylene tetrachloride; sym-tetrachloroethane; s-tetrachloro- ethane; tetrachlorethane; l,l-dichloro-2,2-dichloroethane C2H2C14 79-34-5 167.85 Colorless -43.8°C -36°C 145. 1°C 146.2°C 146. 5°C 1.594 1.595 0.50 ppm 1.5 ppm 3-5 ppm 2.87 g/L (20°C) 2.85 g/L (25°C) Miscible with ethanol, methanol, ether, acetone, benzene, petroleum, carbon tetrachloride, carbon disulfide, dimethyl formamide, oils Reference HSDB, 2009; CAS, 1994 CAS, 1994 CAS, 1994 HSDB, 2009; CAS, 1994; Lide, 1993;Riddicketal, 1986 Hawley, 1981 Riddicketal, 1986 Lide, 1993 Riddicketal., 1986 Lide, 1993 Merck Index, 1989 Riddicketal., 1986 Lide, 1993 HSDB, 2009; Amoore and Hautala, 1983 Amoore and Hautala, 1983 HSDB, 2009 Riddicketal., 1986 Merck Index, 1989 HSDB, 2009; Merck Index, 1989; Hawley, 1981 DRAFT - DO NOT CITE OR QUOTE ------- Table 2-1. Chemical and physical properties of l,l?2,2-tetrachloroethane Characteristic Vapor pressure Partition coefficients: log Kow logKoc Henry's law constant Flash point Conversions: ppm to mg/m3 mg/m3 to ppm Information 5.95mmHg(25°C) 9mmHg(30°C) 2.39 1.66 2.78 4.7 x 10~4 atm-m3/mol 4.55 x lO'4 atm-m3/mol 1.80 x 10"3 atm-m3/mol None - nonflammable 1 ppm = 6.87 mg/m3 1 mg/m3 = 0. 146 ppm Reference Riddicketal., 1986 HSDB, 2009; Flick, 1985 Hansch and Leo, 1 985 Chiouetal., 1979 ASTER, 1995 Mackay and Shiu, 1981 HSDB, 2009 ASTER, 1995 HSDB, 2009; Hawley, 1981 Calculated Calculated 1 2 3 4 5 9 10 11 12 13 In the past, the major use for 1,1,2,2-tetrachloroethane was in the production of trichloroethylene, tetrachloroethylene, and 1,2-dichloroethylene (Archer, 1979). With the development of new processes for manufacturing chlorinated ethylenes and the availability of less toxic solvents, the production of 1,1,2,2-tetrachloroethane as a commercial end-product in the United States and Canada has steadily declined since the late 1960s, and production ceased by the early-1990s (HSDB, 2009; Environment Canada and Health Canada, 1993). 1,1,2,2-Tetrachloroethane may still appear as a chemical intermediate in the production of a variety of other common chemicals. It was also used as a solvent, in cleaning and degreasing metals, in paint removers, varnishes, and lacquers, in photographic films, and as an extractant for oils and fats (Hawley, 1981). Although at one time it was used as an insecticide, fumigant, and weed killer (Hawley, 1981), it presently is not registered for any of these purposes. It was once used as an ingredient in an insect repellent, but registration was canceled in the late 1970s. DRAFT - DO NOT CITE OR QUOTE ------- 1 3. TOXICOKINETICS 2 3 4 1,1,2,2-Tetrachloroethane is well absorbed from the respiratory and gastrointestinal tracts 5 in both humans and laboratory animals and is extensively metabolized and excreted, chiefly as 6 metabolites, in the urine and breath. The metabolism of 1,1,2,2-tetrachloroethane in rats and 7 mice results in the production of trichloroethanol, trichloroacetic acid, and dichloroacetic acid. 8 The dichloroacetic acid is then broken down to glyoxalic acid, oxalic acid, and carbon dioxide. 9 When 1,1,2,2-tetrachloroethane undergoes reductive or oxidative metabolism, reactive radical 10 and acid chloride intermediates, respectively, are produced. 11 12 3.1. ABSORPTION 13 3.1.1. Oral Exposure 14 There are no known studies that quantify absorption following oral exposure in humans. 15 However, the health effects resulting from ingestion of large amounts of 1,1,2,2-tetrachloro- 16 ethane in humans (Section 4.1.1) indicate that 1,1,2,2-tetrachloroethane is absorbed following 17 oral exposure. 18 Observations in animals indicate that the oral absorption of 1,1,2,2-tetrachloroethane is 19 rapid and extensive. Cottalasso et al. (1998) reported hepatic effects, including increases in 20 serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT), a decrease in 21 microsomal glucose-6-phosphatase (G6Pase) activity, and an increase in triglyceride levels, only 22 15-30 minutes following a single oral exposure in rats. Following a single oral exposure of male 23 Osborne-Mendel rats and B6C3Fi mice to 150 mg/kg of radiolabeled 1,1,2,2-tetrachloroethane in 24 corn oil, only 4-6% of the activity was recovered in the feces 72 hours postexposure while >90% 25 of the administered activity was found in both species as metabolites, indicating that the 26 compound was nearly completely absorbed in both rats and mice within 72 hours (Dow 27 Chemical Company, 1988). Mitoma et al. (1985) exposed groups of male Osborne-Mendel rats 28 to 25 or 100 mg/kg and B6C3Fi mice to 50 or 200 mg/kg of 1,1,2,2-tetrachloroethane in corn oil 29 gavage 5 days/week for 4 weeks, followed by a single radiolabeled dose of the compound, and 30 evaluated the disposition of the radiolabeled 1,1,2,2-tetrachloroethane over the next 48 hours. 31 While absorption was not quantified, 79% of the dose was metabolized in rats and 68% was 32 metabolized in mice, suggesting that at least those levels of compound had been absorbed within 33 48 hours. 34 DRAFT - DO NOT CITE OR QUOTE ------- 1 3.1.2. Inhalation Exposure 2 While studies of the systemic toxicity of 1,1,2,2-tetrachloroethane following inhalation in 3 humans are indicative of some level of systemic absorption, comparatively few studies have 4 quantitatively addressed this issue. A study in volunteers was carried out in which a bulb 00 5 containing [ Cl]-labeled 1,1,2,2-tetrachloroethane was inserted into their mouths; they 6 immediately inhaled deeply, held their breaths for 20 seconds, and then exhaled through a trap 7 containing granulated charcoal. The study showed that approximately 96% of a single breath of 8 1,1,2,2-tetrachloroethane was absorbed systemically (Morgan etal., 1970). Two subjects were 9 reported to retain approximately 40-60% of inspired 1,1,2,2-tetrachloroethane after a 30-minute 10 exposure of up to 2,300 mg/m3 (Lehmann et al., 1936), but additional details were not provided. 11 The total body burden of 1,1,2,2-tetrachloroethane in male Osborne-Mendel rats and 12 B6C3Fi mice exposed to a vapor concentration of 10 ppm (68.7 mg/m3) for 6 hours (Dow 13 Chemical Company, 1988) was 38.7 (imol equivalents/kg in rats (9.50 umol equivalents and 14 using a body weight of 245 g from the study) and 127 umol equivalents/kg in mice (3.059 umol 15 equivalents and using a body weight of 24.1 g from the study), indicating that while absorption 16 occurred in both species, mice absorbed proportionally more 1,1,2,2-tetrachloroethane on a per- 17 body-weight basis. Ikeda and Ohtsuji (1972) detected metabolites, measured as total 18 trichlorocompounds, trichloroacetic acid, and trichloroethanol, in the urine of rats exposed to 200 19 ppm (1,370 mg/m3) 1,1,2,2-tetrachloroethane, indicating that absorption had occurred; however, 20 they did not provide a quantitative estimate of absorption rate or fraction. Similarly, Gargas and 21 Anderson (1989) followed the elimination of 1,1,2,2-tetrachloroethane as exhaled breath from 22 the blood after a 6-hour exposure to 350 ppm (2,400 mg/m3), but did not provide quantitative 23 estimates of absorption. 24 25 3.2. DISTRIBUTION 26 No studies measuring the distribution of 1,1,2,2-tetrachloroethane in humans following 27 inhalation or oral exposure were located. Following absorption in animals, 1,1,2,2-tetrachloro- 28 ethane appears to be distributed throughout the body, but may selectively accumulate to a degree 29 in certain cells and tissues. The human blood-air partition coefficient for 1,1,2,2-tetrachloro- 30 ethane has been reported to be in the range of 72.6-116 (Meulenberg and Vijverberg, 2000; 31 Gargas et al., 1989; Morgan et al., 1970). The tissue:air partition coefficients for 1,1,2,2-tetra- 32 chloroethane in rats have been reported to be 142 (blood), 3,767 (fat), 196 (liver), and 33 101 (muscle) (Meulenberg and Vijverberg, 2000; Gargas et al., 1989), indicating that 34 1,1,2,2-tetrachloroethane may partition into fatty tissues, consistent with its low water solubility. 35 Following a single intravenous injection of radiolabeled 1,1,2,2-tetrachloroethane, 36 Eriksson and Brittebo (1991) reported a selective uptake of nonvolatile radioactivity in the 37 mucosal tissues of olfactory and tracheobronchial regions of the respiratory tract and in the 38 mucosae of the oral cavity, tongue, nasopharynx, esophagus, and cardiac region of the 6 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 forestomach. High levels of radioactivity were also found in the liver, bile, inner zone of the adrenal cortices, and interstitium of the testes, although the levels were not quantified. 3.3. METABOLISM No studies were located that investigated the metabolism of 1,1,2,2-tetrachloroethane in humans. Information regarding 1,1,2,2-tetrachloroethane metabolism in animals is summarized below, and a suggested metabolic scheme based on in vivo and in vitro data is presented in Figure 3-1. ci 10 11 12 13 14 15 16 17 18 19 20 21 22 free radical CI CI CI CI CI \ / reductive /\ dechlorination cf CI 1,1,2,2-tetrachloroethane hydrolytic avage CI > / cr Trichloroethylene y CI CI trichloroacetaldehyde fa / CI OH 1 1 CI— 1 CI _ Non-enzymatic UAI dehydrochlorination 1 CI * \ o cr JdUUII CI / CI \ y \ CI O // o CI /' v\ Cl \ \s nH CI OH o J/ oxalic acid trichloroethanol Trichloroacetic acid XI . \ CI O \ _ cr OH / dichloroacetic acid NH2 HO Glyoxylic acid \ glycine O C02 + C OH formic acid Source: Adapted from ATSDR (1996). Figure 3-1. Suggested metabolic pathways of 1,1^2,2-tetrachloroethane. In vivo and in vitro studies indicate that the metabolism of 1,1,2,2-tetrachloroethane proceeds via multiple pathways in rodents (Mitoma et al, 1985; Casciola and Ivanetich, 1984; Halpert, 1982; Koizumi et al., 1982; Halpert and Neal, 1981; Ikeda and Ohtsuji, 1972; Yllner, 1971). The predominant pathway appears to involve production of dichloroacetic acid, formed as an initial metabolite via stagewise hydrolytic cleavage of 1,1,2,2-tetrachloroethane, yielding dichloroacetyl chloride and dichloroacetaldehyde as intermediates, or by cytochrome P450-based oxidation of 1,1,2,2-tetrachloroethane (Casciola and Ivanetich, 1984; Halpert and Neal, 1981; 7 DRAFT - DO NOT CITE OR QUOTE ------- 1 Yllner, 1971). Dichloroacetic acid was identified as the major urinary metabolite in mice treated 2 with 1,1,2,2-tetrachloroethane by intraperitoneal (i.p.) injection (Yllner et al., 1971) and in in 3 vitro systems with rat liver microsomal and nuclear cytochrome P450 (Casciola and Ivanetich, 4 1984; Halpert, 1982; Halpert and Neal, 1981). Dichloroacetic acid can be further metabolized to 5 glyoxylic acid, formic acid, and carbon dioxide (Yllner, 1971), with carbon dioxide a potential 6 major component of the end products (Yllner, 1971). Other pathways involve the formation of 7 trichloroethylene via dehydrochlorination or tetrachloroethylene via oxidation as initial 8 metabolites. Trichloroethylene and tetrachloroethylene are further metabolized to trichloro- 9 ethanol and trichloroacetic acid, and oxalic acid and trichloroacetic acid, respectively (Mitoma et 10 al., 1985; Ikeda and Ohtsuji, 1972; Yllner et al., 1971). 1,1,2,2-Tetrachloroethane may also form 11 free radicals by undergoing reductive dechlorination (ATSDR, 1996). The formation of free 12 radical intermediates during 1,1,2,2-tetrachloroethane metabolism has been demonstrated in 13 spin-trapping experiments (Paolini et al., 1992; Tomasi et al., 1984). 14 Metabolism of 1,1,2,2-tetrachloroethane is generally extensive, with 68-95% of a total 15 administered dose found as metabolites (Dow Chemical Company, 1988; Mitoma et al., 1985; 16 Yllner, 1971). Mice given a single 0.21-0.32 g/kg i.p. dose of [14C]-labeled 1,1,2,2-tetrachloro- 17 ethane eliminated 45-61% of the administered radioactivity as carbon dioxide in expired air and 18 23-34% of the radioactivity in urine in the following 3 days (Yllner et al., 1971). Dichloroacetic 19 acid, trichloroacetic acid, trichloroethanol, oxalic acid, glyoxylic acid, and urea accounted for 27, 20 4, 10, 7, 0.9, and 2% of the mean urinary radioactivity excreted by the mice in 24 hours, 21 respectively (Yllner et al., 1971). Yllner et al. (1971) also demonstrated that 20-23% of the 22 [14C]-tetrachloroethane was converted to glycine following the simultaneous i.p. injection of 23 [14C]-tetrachloroethane and sodium benzoate and the estimation of [14C]-hippuric acid in the 24 urine. In rats, trichloroethanol appeared to be present as a urinary metabolite at approximately 25 fourfold greater levels than trichloroacetic acid following a single 8-hour inhalation exposure 26 (Ikeda and Ohtsuji, 1972). Several studies have reported that metabolism of 1,1,2,2-tetrachloro- 27 ethane is greater in mice than in rats, with magnitudes of the reported difference generally in the 28 range of a 1.1-3.5-fold greater metabolic activity, on a per-kg basis, in mice (Dow Chemical 29 Company, 1988; Mitoma et al., 1985; Milman et al., 1984). 30 As indicated above, cytochrome P450-based metabolism of 1,1,2,2-tetrachloroethane to 31 dichloroacetic acid has been demonstrated in vitro. Multiple P450 isozymes are likely to be 32 involved, as demonstrated by studies reporting increased metabolism and covalent binding of 33 metabolites following pretreatment with phenobarbital (Casciola and Ivanetich, 1984; Halpert, 34 1982), xylene (Halpert, 1982), or ethanol (Sato et al., 1980). The isozymes induced by 35 phenobarbital, xylene, and ethanol include members of the CYP2A, CYP2B, CYP2E, and 36 CYP3A subfamilies (Omiecinski et al., 1999; Nebert et al., 1987). 37 1,1,2,2-Tetrachloroethane has also been reported to produce inactivation of cytochrome 38 P450. 1,1,2,2-Tetrachloroethane effectively inactivated the major phenobarbital-inducible P450 8 DRAFT - DO NOT CITE OR QUOTE ------- 1 isozyme, but not the major P450 isozyme induced by p-naphthoflavone, in rat liver in vitro 2 (Halpert et al, 1986). Rat liver nuclear cytochrome P450 levels were reduced following in vitro 3 incubation with 1,1,2,2-tetrachloroethane and a NADPH-generating system (Casciola and 4 Ivanetich, 1984). In an in vivo study, cytochrome P450 activity was evaluated in male and 5 female Swiss albino mice 24 hours after a single 0, 300, or 600 mg/kg i.p. dose of 1,1,2,2-tetra- 6 chloroethane (Paolini et al., 1992). 1,1,2,2-Tetrachloroethane treatment statistically significantly 7 (p < 0.01) reduced total cytochrome P450 activity 44 and 37% in males and females, respectively, 8 at 300 mg/kg and 85 and 74% in males and females, respectively, at 600 mg/kg. Treatment with 9 600 mg/kg statistically significantly reduced the microsomal activity of P450 isozymes 3 A, 2E1, 10 1A2, 2B1, and 1A1 in both genders, and 300 mg/kg reduced the activity of P4503A in both sexes 11 and P4502B1 in males. Heme content was reduced 13 and 33% at 300 and 600 mg/kg, 12 respectively, and may have contributed to the decrease in CYP450 levels. The 600 mg/kg dose 13 also reduced the activity of glutathione S-transferase (GST) toward l-chloro-2,4-dinitrobenzene, 14 a general GST substrate, in both genders. 15 Due to the extensive metabolism of 1,1,2,2 tetrachloroethane to products such as 16 trichloroethylene and dichloroacetic acid, the relevance of 1,1,2,2-tetrachloroethane interactions 17 with GST is important. Studies of human GST-zeta polymorphic variants show different 18 enzymatic activities toward and inhibition by dichloroacetic acid that could reasonably affect the 19 metabolism of 1,1,2,2-tetrachloroethane (Lantum et al., 2002; Blackburn et al., 2001, 2000; 20 Tzeng et al., 2000). Dichloroacetic acid may covalently bind to GST-zeta (Anderson et al., 21 1999) and inhibit its own metabolism, leading to an increase in the amount of unmetabolized 22 dichloroacetic acid as the dose and/or duration increases (U.S. EPA, 2003). 23 Data indicate that 1,1,2,2-tetrachlorethane can be metabolized to dichloroacetic acid 24 (ATSDR, 1996; Yllner, 1971), suggesting a potential role for this metabolite in some of the 25 cancer and noncancer effects observed following exposure to 1,1,2,2 tetrachloroethane. 26 Following an intravenous injection of radiolabeled 1,1,2,2-tetrachloroethane, radioactivity could 27 not be extracted from epithelium of the respiratory and upper alimentary tracts, or from the liver, 28 adrenal cortex, or testes (Eriksson and Brittebo, 1991). The presence of tissue-bound metabolites 29 in the epithelial linings in the upper respiratory tract may demonstrate a first-pass effect by the 30 respiratory tract (Eriksson and Brittebo, 1991). In addition, the presence of irreversible tissue- 31 bound metabolites demonstrates the metabolism of 1,1,2,2-tetrachloroethane to reactive 32 metabolites (Eriksson and Brittebo, 1991). However, the identities of the bound metabolites and 33 modified proteins or phospholipids were not identified. The presence of radiolabel in the 34 proteins may have been radiolabeled incorporated glycine. 35 Dow Chemical Company (1988) observed radiolabel in hepatic DNA, although the 36 presence of the radiolabel in the hepatic DNA likely represented the incorporation of single 37 [14C]-atoms via normal biosynethetic pathways. Mice were found to have approximately a 38 1.9-fold greater extent of [14C] activity irreversibly associated with hepatic macromolecules than 9 DRAFT - DO NOT CITE OR QUOTE ------- 1 rats, which the study authors noted was consistent with the greater metabolism, on a per-kg basis, 2 in mice compared to rats. After a 4-week oral exposure to unlabeled 1,1,2,2-tetrachloroethane 3 followed by a single oral dose of labeled 1,1,2,2-tetrachloroethane, Mitoma et al. (1985) also 4 reported greater levels of hepatic protein-binding in the tissue of mice compared to rats, and the 5 differences were on the order of twofold greater binding in mice, which would be consistent both 6 with the Dow Chemical Company (1988) studies and with the observed differences in 7 metabolism of the two species discussed above. This may also be related to the 3.2-3.5-fold 8 greater absorption, on a per-kg basis, of mice compared to rats following inhalation exposure 9 (Dow Chemical Company, 1988). 10 The kinetic constants of 1,1,2,2-tetrachloroethane metabolism in rats exposed to 350 ppm 11 of the chemical for 6 hours were determined in gas uptake studies performed by Gargas and 12 Anderson (1989). The rate of exhalation of 1,1,2,2-tetrachloroethane was measured and, 13 combined with previously published values for partition coefficients for blood/air, liver/blood, 14 muscle/blood, and fat/blood, allowed the estimation of the disposition of the chemical in rat 15 (Gargas et al., 1989). A Km of 4.77 uM and a Vmax of 12 mg/hour (scaled to a 1-kg rat) were 16 measured. 17 18 3.4. ELIMINATION 19 Morgan et al. (1970) reported that the urinary excretion rate of 1,1,2,2-tetrachloroethane 20 in humans was 0.015% of the absorbed dose/minute. No other studies measuring the elimination 21 of 1,1,2,2-tetrachloroethane in humans have been reported. 22 Available animal data indicate that following absorption into the body, 1,1,2,2-tetra- 23 chloroethane is eliminated mainly as metabolites in urine, as carbon dioxide, or as unchanged 24 compound in expired air (Gargas and Anderson, 1989; Dow Chemical Company, 1988; Mitoma 25 et al., 1985; Ikeda and Ohtsuji, 1972; Yllner et al., 1971). The patterns of elimination in rats and 26 mice are qualitatively similar (Dow Chemical Company, 1988; Mitoma et al., 1985), although 27 covalent binding is somewhat greater in mice than rats. Elimination is fairly rapid, with 28 significant amounts present in the urine and expired air at 48-72 hours postexposure (Dow 29 Chemical Company, 1988; Mitoma et al., 1985; Ikeda and Ohtsuji, 1972; Yllner et al., 1971). 30 Only one study quantitatively evaluated the elimination of 1,1,2,2-tetrachloroethane 31 following inhalation exposure. Dow Chemical Company (1988) followed the excretion of 32 1,1,2,2-tetrachloroethane for 72 hours following exposure of rats and mice to vapor 33 concentrations of 10 ppm (68.7 mg/m3) [14C]-l,l,2,2-tetrachloroethane for 6 hours. More than 34 90% of the absorbed dose was metabolized in both species. The percentage of recovered 35 radioactivity reported in rats was 33% in breath (25% as CC>2 and 8% as unchanged compound), 36 19% in urine, and 5% in feces. In mice, the percentage of recovered radioactivity was 34% in 37 breath (32% as CC>2 and 2% as unchanged compound), 26% in urine, and 6% in feces. 10 DRAFT - DO NOT CITE OR QUOTE ------- 1 Radioactivity in urine and feces was nonvolatile (inferred by the researchers to be product(s) of 2 metabolism), but was not otherwise characterized. 3 With regard to oral exposure, the excretion of 1,1,2,2-tetrachloroethane was followed for 4 72 hours following oral administration of 150 mg/kg doses to rats and mice (Dow Chemical 5 Company, 1988). Greater than 90% of the absorbed dose was detected as metabolites in both 6 species. In rats, 41% was excreted in breath (32% as CC>2 and 9% as unchanged compound), 7 23% in urine, and 4% in feces. In mice, 51% was excreted in breath (50% as CC>2 and 1% as 8 unchanged compound), 22% in urine, and 6% in feces. Radioactivity in urine and feces was 9 nonvolatile (inferred by the researchers to be product(s) of metabolism), but was not otherwise 10 characterized. Mitoma et al. (1985) found that mice given an oral dose of 1,1,2,2-tetrachloro- 11 ethane excreted about 10% of the dose unchanged in the breath, and the rest was metabolized 12 and excreted in the breath as carbon dioxide (10%) or in the urine and feces (30%, measured 13 together), or retained in the carcass (27%) after 48 hours. Rats showed similar patterns of 14 excretion (Mitoma etal., 1985). The most comprehensive study of the metabolism and excretion 15 of 1,1,2,2-tetrachloroethane was an i.p. study in mice using [14C]-labeled 1,1,2,2-tetrachloro- 16 ethane. Yllner (1971) showed that after 72 hours, about 4% of the radioactivity was expired 17 unchanged in the breath, 50% was expired as carbon dioxide, 28% was excreted in the urine, 1% 18 was excreted in the feces, and 16% remained in the carcass. 19 Delays in elimination may be the result of covalent binding of 1,1,2,2-tetrachloroethane 20 metabolites, as reflected in high levels of compound detected in the carcasses of animals. 21 Mitoma et al. (1985) reported a 30.75% retention in the carcass of rats and a 27.44% retention in 22 the carcass of mice 48 hours after exposure to a single labeled dose of 25 m/kg in rats and 50 23 mg/kg in mice 1,1,2,2-tetrachloroethane. Dow Chemical Company (1988) reported 30% 24 retention in the carcass in rats exposed to 10 ppm by inhalation, 25% in mice exposed to 10 ppm 25 by inhalation, 23% in rats exposed to 150 mg/kg by gavage, and 17.3% in mice exposed to 26 150 mg/kg by gavage. Colacci et al. (1987) reported covalent binding of radiolabeled 27 1,1,2,2-tetrachloroethane to DNA, RNA, and protein in the liver, kidneys, lung, and stomach of 28 rats and mice exposed to a single intravenous dose and analyzed 22 hours postexposure. In vitro 29 binding to calf thymus DNA was found to be greatest when the microsomal fraction was present, 30 and was inhibited by SKF-525A, indicating that metabolic activation was likely required for 31 DNA binding (Colacci et al., 1987). However, Collaci et al. (1987) did not distinguish between 32 covalent binding and whether the presence of radiolabel in the DNA, RNA, and protein was the 33 result of incorporated radiolabeled carbon into the biomolecules through normal biochemical 34 processes. 35 36 3.5. PHYSIOLOGICALLY BASED TOXICOKINETIC MODELS 37 No physiologically based toxicokinetic (PBTK) models for 1,1,2,2-tetrachloroethane 38 were located for humans. Muelenberg et al. (2003) used saline:air, rat brain:air, and olive oil:air 11 DRAFT - DO NOT CITE OR QUOTE ------- 1 partition coefficients to model 28 chemicals from three distinct chemical classes, including 2 alkylbenzenes, chlorinated hydrocarbons, and ketones. The saline:air, rat brain:air, and olive 3 oil:air partition coefficients derived for 1,1,2,2-tetrachloroethane were 35.6 ± 6.05, 344 ± 21.0, 4 and 10,125 ± 547, respectively. The brain partition coefficients for the 28 chemicals were 5 predicted with accuracy within a factor of 2.5 for 95% of the chemicals. While the study 6 demonstrates the ability to predict rat brain partition coefficients using a bilinear equation, the 7 utility of the information for this assessment is limited. Similarly, several physiologically based 8 pharmacokinetic (PBPK) investigations of 1,1,2,2-tetrachloroethane exposure in fish (McKim et 9 al., 1999; Nichols et al, 1993) provide little utility for this assessment. In sum, adequate 10 information for PBTK modeling of 1,1,2,2-tetrachloroethane remains a research need. 11 Chiu and White (2006) presented an analysis of steady-state solutions to a PBPK model 12 for a generic volatile organic chemical (VOC) metabolized in the liver. The only parameters 13 used to determine the system state for a given oral dose rate or inhalation exposure concentration 14 were the blood-air partition coefficient, metabolic constants, and the rates of blood flow to the 15 liver and of alveolar ventilation. At exposures where metabolism is close to linear (i.e., 16 unsaturated), it was demonstrated that only the effective first order metabolic rate constant was 17 needed. Additionally, it was found that the relationship between cumulative exposure and 18 average internal dose (e.g., areas under the curve [AUCs]) remains the same for time-varying 19 exposures. The study authors concluded that steady-state solutions can reproduce or closely 20 approximate the solutions using a full PBPK model. Section 5.2.2 addresses the applicability of 21 using this model to conduct a route-to-route extrapolation in this assessment. 12 DRAFT - DO NOT CITE OR QUOTE ------- 1 4. HAZARD IDENTIFICATION 2 3 4 4.1. STUDIES IN HUMANS—EPIDEMIOLOGY, CASE REPORTS, CLINICAL 5 CONTROLS 6 4.1.1. Oral Exposure 7 A number of case reports provide information on effects of intentional acute exposure to 8 lethal oral doses of 1,1,2,2-tetrachloroethane (Mant, 1953; Lilliman, 1949; Forbes, 1943; Elliot, 9 1933; Hepple, 1927). Subjects usually lost consciousness within approximately 1 hour and died 10 3-20 hours postingestion, depending on the amount of food in the stomach. Postmortem 11 examinations showed gross congestion in the esophagus, stomach, kidneys, spleen, and trachea, 12 gross congestion and edema in the lungs, and histological effects of congestion and cloudy 13 swelling in the lungs, liver, and/or kidneys (Mant, 1953; Hepple, 1927). Amounts of 14 1,1,2,2-tetrachloroethane recovered from the stomach and intestines of the deceased subjects 15 included 12 mL (Hepple, 1927), 25 g (Lilliman, 1949), 48.5 mL (Mant, 1953), and 425 mL 16 (Mant, 1953). Assuming a density of 1.594 g/mL and an average body weight of 70 kg, the 17 approximate minimum doses consumed in these cases are estimated to be approximately 273, 18 357, 1,100, and 9,700 mg/kg, respectively. No deaths occurred in eight patients (six men and 19 two women) who were accidentally given 3 mL of 1,1,2,2-tetrachloroethane (68 mg/kg, using 20 the above assumptions) or three patients (one young man, one young woman, and one 12-year- 21 old girl) who were accidentally given 2 or 3 mL (98-117 mg/kg, using the density and reported 22 body weights) as medicinal treatment for hookworm (Ward, 1955; Sherman, 1953). These 23 patients experienced loss of consciousness and other clinical signs of narcosis that included 24 shallow breathing, faint pulse, and pronounced lowering of blood pressure. 25 26 4.1.2. Inhalation Exposure 27 The symptoms of high-dose acute inhalation exposure to 1,1,2,2-tetrachloroethane 28 commonly include drowsiness, nausea, headache, constipation, decreased red blood cell (RBC) 29 count, weakness, and at extremely high concentrations, jaundice, unconsciousness, and 30 respiratory failure (Coyer, 1944; Hamilton, 1917). 31 An experimental study was conducted in which two volunteers self-inhaled various 32 concentrations of 1,1,2,2-tetrachloroethane for up to 30 minutes (Lehmann et al, 1936). The 33 results of this study suggest that 3 ppm (6.9 mg/m3) was the odor detection threshold; 13 ppm 34 (89 mg/m ) was tolerated without effect for 10 minutes, while 146 ppm (1,003 mg/m ) for 35 30 minutes or 336 ppm (2,308 mg/m3) for 10 minutes produced irritation of the mucous 36 membranes, pressure in the head, vertigo, and fatigue. No other relevant information was 37 reported. 13 DRAFT - DO NOT CITE OR QUOTE ------- 1 Minot and Smith (1921) reported that symptoms of industrial 1,1,2,2-tetrachloroethane 2 poisoning (concentrations not specified) included fatigue, perspiration, drowsiness, loss of 3 appetite, nausea, vomiting, constipation, headache, and jaundice. Hematological changes 4 included increased large mononuclear cells, elevated white blood cell (WBC) count, a slight but 5 progressive anemia, and a slight increase in platelet number. Similar symptoms were reported by 6 Parmenter (1921) and Wilcox et al. (1915). Horiguchi et al. (1964) reported that in 127 coating 7 workers employed in artificial pearl factories and exposed to 75-225 ppm (500-1,500 mg/m3) 8 1,1,2,2-tetrachloroethane (along with other solvents), observed effects included decreased 9 specific gravity of the whole blood, decreased RBC count, relative lymphocytosis, neurological 10 findings (not specified), and a positive urobilinogen test. 11 Lobo-Mendonca (1963) observed a number of adverse health effects in a mixed-gender 12 group of 380 workers at 23 Indian bangle manufacturing facilities (80% of workers employed at 13 these facilities were examined). In addition to the inhalation exposure, approximately 50% of 14 the examined workers had a substantial amount of dermal exposure to 1,1,2,2-tetrachloroethane. 15 Some of the workers were exposed to a mixture of equal parts acetone and 1,1,2,2-tetrachloro- 16 ethane. Air samples were collected at several work areas in seven facilities. Levels of 17 1,1,2,2-tetrachloroethane in the air ranged from 9.1 to 98 ppm (62.5-672 mg/m3). High 18 incidences of a number of effects were reported, including anemia (33.7%), loss of appetite 19 (22.6%), abdominal pain (23.7%), headaches (26.6%), vertigo (30.5%), and tremors (35%). The 20 significance of these effects cannot be determined because a control group of unexposed workers 21 was not examined and coexposure to acetone was possible. The study authors noted that the 22 incidence of tremors appeared to be directly related to 1,1,2,2-tetrachloroethane exposure 23 concentrations, as the percentage of workers handling tetrachloroethane and displaying tremors 24 increased as the air concentration of 1,1,2,2-tetrachloroethane increased. 25 Over a 3-year period, Jeney et al. (1957) examined 34-75 workers employed at a 26 penicillin production facility. 1,1,2,2-Tetrachloroethane was used as an emulsifier, and wide 27 fluctuations in atmospheric levels occurred throughout the day. The investigators noted that the 28 workers were only in the areas with high 1,1,2,2-tetrachloroethane concentrations for short 29 periods of time, and gauze masks with organic solvent filters were worn in these areas. During 30 the first year of the study, 1,1,2,2-tetrachloroethane levels ranged from 0.016 to 1.7 mg/L (16- 31 1,700 mg/m3; 2-248 ppm). In the second year of the study, ventilation in the work room was 32 improved and 1,1,2,2-tetrachloroethane levels ranged from 0.01 to 0.85 mg/L (10-850 mg/m ; 33 1-124 ppm). In the third year of the study, the workers were transferred to a newly built facility 34 and 1,1,2,2-tetrachloroethane levels in the new facility ranged from 0.01 to 0.25 mg/L (10- 35 250 mg/m3; 1-36 ppm). At 2-month intervals, the workers received general physical 36 examinations, and blood was drawn for measurement of hematological parameters, serum 37 bilirubin levels, and liver function tests; urinary hippuric acid levels were measured every 38 6 months. It appears that workers with positive signs of liver damage, including palpability of 14 DRAFT - DO NOT CITE OR QUOTE ------- 1 the liver, rise in bilirubin levels, positive liver function tests, and urobilinogenuria, were 2 transferred to other areas of the facility and were not examined further. 3 In the first year of the study, 31% of the examined workers had "general or gastro- 4 intestinal symptoms." Loss of appetite, bad taste in the mouth, epigastric pain, and a "dull 5 straining pressure feeling in the area of the liver" were reported by 66% of the workers 6 experiencing gastrointestinal symptoms. Other symptoms included headaches, general weakness, 7 and fatigue in 29%, severe weight loss in 4%, and "tormenting itching" in 1%. Enlargement of 8 the liver was observed in 38% of the screened workers. Urobilinogenuria was detected in 50% 9 of the workers, most often following more than 6 months of employment, and 31 % of the 10 workers with urobilinogenuria also had palpable livers. 11 In the second year of the study, there was a decline in the number of symptomatic 12 workers (13% of examined workers) and in workers with positive urobilinogenuria findings 13 (24%). Liver enlargement was observed in 20% of the examined workers. In the third year, the 14 number of workers reporting symptoms decreased to 2%, and positive urobilinogen findings 15 were found in 12%. The investigators noted that the increased urobilinogen levels during the 16 third year of observation may have been secondary to excessive alcohol consumption or dietary 17 excess. Enlarged livers were found in 5% of the examined workers. 18 During the course of the study, no alterations in erythrocyte or hemoglobin (Hb) levels 19 were found. Leukopenia (defined as leukocyte levels of <5,800 cells/mL) was found in 20% of 20 the workers, but no relationship between the number of cases and duration of 1,1,2,2-tetrachloro- 21 ethane exposure was found. A positive relationship between duration of exposure and frequency 22 of abnormal liver function test results was observed, as statistically significant correlations were 23 found on the thymol and Takata-Ucko liver function tests, but not the gold sol reaction test. The 24 thymol liver function test measures the direct precipitation of both lipids and abnormal lipid 25 protein complexes appearing in liver disease by the addition of a thymol solution (Kunkel and 26 Hoagland, 1947). The Takata-Ucko (or Takata-Ara) test detects an increase in the amounts of 27 the globulin components of the serum, signifying liver disease (Kunkel and Hoagland, 1947). 28 Abnormal hippuric acid levels were only detected in 1% of the examined workers during the first 29 2 years, and no abnormalities were observed during the third year. Increased serum bilirubin 30 levels (>1 mg/dL) were observed in 20, 18.7, and 7.6% of the workers during the first, second, 31 and third years, respectively. The prevalence of hepatitis was assessed using sickness benefit 32 files. In the 1-year period prior to the study, 21 cases of hepatitis were found (total number of 33 workers not reported). Three cases of hepatitis were found in the first year of the study, eight 34 cases in the second year, and four cases in the third year. The lack of a control group and poor 35 reporting of study design and results precludes using this study for quantitative dose-response 36 analysis. 37 Norman et al. (1981) examined the mortality of the employees of 39 chemical processing 38 plants used by the Army during World War II. Ten plants used 1,1,2,2-tetrachloroethane to help 15 DRAFT - DO NOT CITE OR QUOTE ------- 1 treat clothing, while the others plants used water in the same process. Estimates of exposure 2 levels were not reported, and coexposure to dry-cleaning chemicals was expected. At the time of 3 evaluation, 2,414 deaths were reported in the study cohort. No differences in standard mortality 4 ratios were seen between the tetrachloroethane and water groups for total mortality, 5 cardiovascular disease, cirrhosis of the liver, or cancer of the digestive and respiratory systems. 6 The mortality ratio for lymphatic cancers in the tetrachloroethane group was increased relative to 7 controls or the water group, although the number of deaths was small (4 cases, with an expected 8 number of 0.85). No other differences were seen between the groups. 9 10 4.2. SUBCHRONIC AND CHRONIC STUDIES AND CANCER BIOASSAYS IN 11 ANIMALS—ORAL AND INHALATION 12 4.2.1. Oral Exposure 13 4.2.1.1. Subchronic Studies 14 NTP (2004) fed groups of male and female F344 rats (10/sex/group) diets containing 0, 15 268, 589, 1,180, 2,300, or 4,600 ppm of microencapsulated 1,1,2,2-tetrachloroethane for 16 14 weeks. NTP (2004) reported that the microcapsules containing 1,1,2,2-tetrachloroethane 17 were specified to be no greater than 420 (im in diameter, and were not expected to have any 18 significant effect on the study. The reported average daily doses were 0, 20, 40, 80, 170, or 19 320 mg/kg-day, and vehicle control (feed with empty microcapsules) and untreated control 20 groups were used for both genders. Endpoints evaluated throughout the study included clinical 21 signs, body weight, and feed consumption. Hematology and clinical chemistry were assessed on 22 days 5 and 21 and at the end of the study; urinalyses were not performed. Necropsies were 23 performed on all animals, and selected organs (liver, heart, right kidney, lung, right testis, and 24 thymus) were weighed. Comprehensive histological examinations were performed on untreated 25 control, vehicle control, and high dose groups. Tissues examined in the lower dose groups were 26 limited to bone with marrow, clitoral gland, liver, ovary, prostate gland, spleen, testis with 27 epididymis and seminal vesicle, and uterus. A functional observational battery (FOB) was 28 performed on rats in the control groups and the 20, 40, and 80 mg/kg-day groups during weeks 4 29 and 13. Sperm motility, vaginal cytology, estrous cycle length, and percentage of time spent in 30 the various estrus stages were evaluated in control groups and the 40, 80, and 170 mg/kg-day 31 groups. 32 All animals survived to the end of the study, but clinical signs of thinness and pallor were 33 observed in all animals in the 170 and 320 mg/kg-day groups (NTP, 2004). Final body weights 34 (Table 4-1) were statistically significantly lower than vehicle controls in males at 80, 170, and 35 320 mg/kg-day (7, 29, and 65% lower, respectively) and females at 80, 170, and 320 mg/kg-day 36 (9, 29, and 56% lower, respectively), with both genders at 320 mg/kg-day losing weight over the 37 course of the study. However, feed consumption by the rats also decreased with increasing dose 38 level (NTP, 2004). 16 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-1. Final body weights (g) and percent change compared to controls in F344/N rats exposed to l,l?2,2-tetrachloroethane in feed for 14 weeks Dose (mg/kg-d) Vehicle control 20 40 80 170 320 n 10 10 10 10 10 10 Males 366 ±5a 354 ±9 353±6 341 ±6b 259 ± 9b 127 ± 9b - -3% -4 -7 -29 -65 n 10 10 10 10 10 10 Females 195 ±4a 192 ±4 189±2 177 ± 2b 139 ±4b 85±3b - -2% -3 -9 -29 -56 2 3 4 5 9 10 aMean ± standard error. V<0.05. Source: NTP (2004). Statistically significant increases in absolute liver weights were observed in female rats exposed to 80 mg/kg-day, and statistically significant decreases in absolute liver weight were observed at >170 mg/kg-day in males and at 320 mg/kg-day in females (Table 4-2a). Statistically significant increases in relative liver weights (Table 4-2b) were observed at >40 mg/kg-day in males and females (NTP, 2004). Significant alterations in absolute and/or relative weights were also observed in the thymus, kidney, heart, lung, and testes primarily at 170 and 320 mg/kg-day. Table 4-2a. Absolute liver weights (g) and percent change compared to controls in F344/N rats exposed to l,l?2,2-tetrachloroethane in feed for 14 weeks Dose (mg/kg-d) Vehicle control 20 40 80 170 320 n 10 10 10 10 10 10 Males 12.74 ±0.26a 12. 99 ±0.35 14. 47 ±0.44 15.54±0.39 11.60±0.44b 6.57±0.18b - 2% 14 22 -9 -48 n 10 10 10 10 10 10 Females 6.84±0.17a 7.03 ±0.12 7.14±0.16 7.80±0.08b 6.66 ±0.21 4.94±0.12b - 3% 4 14 -3 -28 aMean ± standard error. V<0.05. Source: NTP (2004). 11 17 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-2b. Relative liver weight (mg organ weight/g body weight) and percent change compared to controls in F344/N rats exposed to l,l?2,2-tetra- chloroethane in feed for 14 weeks Dose (mg/kg-d) Vehicle control 20 40 80 170 320 n 10 10 10 10 10 10 Males 34.79 ±0.42a 36.72 ±0.44 41.03 ±0.85b 45.61 ±0.52b 44.68 ±0.45b 52.23 ±1.42b - 6% 18 31 28 50 n 10 10 10 10 10 10 Females 35.07±0.56a 36.69 ±0.36 37.84±0.51b 44.20 ±0.27b 48.03 ±0.89b 58.40 ±1.42b - 5% 8 26 37 67 1 2 3 4 5 9 10 11 12 13 14 15 16 17 18 19 aMean ± standard error. V<0.05. Source: NTP (2004). Results of the FOB showed no exposure-related findings of neurotoxicity. The hematology evaluations indicated that 1,1,2,2-tetrachloroethane affected the circulating erythroid mass in both genders (Table 4-3). There was evidence of a transient erythrocytosis, as shown by increases in hematocrit values, Hb concentration, and erythrocyte counts on days 5 and 21 at >170 mg/kg-day. The erythrocytosis was not considered clinically significant and disappeared by week 14, at which time minimal to mild, dose-related anemia was evident, as shown by decreases in hematocrit and Hb at >40 mg/kg-day. For example, although males exposed to 40 mg/kg-day showed a statistically significant decrease in Hb at week 14, the magnitude of the change was small (3.8%). The anemia was characterized as microcytic based on evidence suggesting that the circulating erythrocytes were smaller than expected, including decreases in mean cell volumes, mean cell Hb values, and mean cell Hb concentration in both genders at >80 mg/kg-day at various time points. At week 14, there were no changes in reticulocyte counts, suggesting that there was no erythropoietic response to the anemia, which was in turn supported by the bone marrow atrophy observed microscopically. As discussed by NTP (2004), the erythrocytosis suggested a physiological response consistent with hemoconcentration due to dehydration, as well as compromised nutritional status due to the reduced weight gain and food consumption, both of which may have contributed to the development of the anemia. 18 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-3. Serum chemistry and hematology changes3 in rats exposed to dietary l,l?2,2-tetrachloroethane for 14 weeks Oral dose (mg/kg-d) Vehicle control 20 40 80 170 320 Males (10/group) Serum total protein (g/dL) Serum cholesterol (mg/dL) ALT (IU/L) ALP (IU/L) SDH (IU/L) Bile acids (umol/L) Hematocrit (%) (automated) Hb (g/dL) Mean cell volume (fL) Mean cell Hb (pg) Platelets (103/uL) 7.2±0.1 73 ±2 48 ±2 256 ±7 23 ±1 29. 2 ±2. 9 45.2±0.5 15.8±0.1 50.7±0.1 17.7±0.1 728.4 ±12. 3 7. 3 ±0.1 74 ±3 49 ±2 260 ±5 27±lb 27. 5 ±2.7 44. 9 ±0.4 15.6±0.1 51.8±0.3 18.1±0.1 707.0 ±5. 8 7.3 ±0.1 76 ±2 53±2 248 ±5 26 ±2 27.2 ±2.7 44.0 ±0.9 15.2±0.3b 52. 3 ±0.2 18.0±0.1 727.0 ±25. 2 7. 3 ±0.1 67 ±2 69±3b 245 ±6 31±lb 35.9±3.9 43. 3 ±0.7 14.9±0.1b 51. 3 ±0.2 17.7 ±0.2 716.3 ±9.7 6.7±0.1b 68 ±2 115±8b 353±12b 47±2b 92.0±16.6b 43.1±0.6b 14.6±0.1b 49.4 ±0.2 16.8±0.1b 692.8 ±12.6b 6.0±0.1b 65±2b 292±18b 432±24b 74±4b 332.4 ±47.4b 39.0±l.lb 13.6±0.3b 44.4±0.4b 15.5±0.2b 773.4±23.2b Females (10/group) Serum total protein (g/dL) Serum cholesterol (mg/dL) ALT (IU/L) ALP (IU/L) SDH (IU/L) Bile acids (umol/L) Hematocrit (%) (automated) Hb (g/dL) Mean cell volume (fL) Mean cell Hb (pg) Platelets (103/uL) 7.2±0.1 104 ±4 46 ±2 227 ±5 27 ±1 37.0±7.1 42.8 ±0.4 15.2±0.1 55.4±0.1 19.7±0.1 742.1 ±20.4 7. 3 ±0.0 105 ±3 42 ±1 216±4 27 ±1 46.6 ±6.5 43.2±0.4 15.3±0.1 56.1 ±0.1 19.8±0.1 725. 9 ±12.7 7.3 ±0.1 98 ±1 41±2 220 ±3 28 ±2 39.1 ±5.6 42.1 ±0.4 14.9±0.1 55.8±0.1 19.7±0.1 733. 9 ±8. 8 6.9±0.1 81±2b 49 ±2 225 ±11 25 ±1 36. 3 ±3. 9 40.1±0.5b 14.2±0.2b 53.3±0.2b 18.9±0.1b 727.4 ±14.2 6.4±0.1b 64±3b 112±7b 341 ±7b 45±3b 39.3 ±7. 9 42.8 ±0.7 14.5±0.2b 49.0±0.2b 16.6±0.2b 639.4 ±9.9b 5.6±0.1b 55±3b 339±18b 468 ± 22b 82±3b 321.5±50.6b 34.7±0.7b 12.5±0.2b 44.4±0.4b 16.0±0.2b 662.5 ±19.4b 1 2 3 4 5 "Mean ± standard error. bStatistically significantly different from control value. ALP = alkaline phosphatase; IU = international units; SDH = sorbitol dehydrogenase Source: NTP (2004). Changes in serum clinical chemistry parameters indicative of liver damage were observed in both genders, occurring at all time points (day 5, day 21, and week 14) and increasing in magnitude with increasing dose and time. At week 14 (Table 4-3), these effects included statistically significant increases in ALT and sorbitol dehydrogenase (SDH) activity in males at >80 mg/kg-day (41, 134, and 496%, and 15, 74, and 174%, respectively) and females at 19 DRAFT - DO NOT CITE OR QUOTE ------- 1 >170 mg/kg-day (167 and 707%, and 67 and 204%, respectively), increases in alkaline 2 phosphatase (ALP) activity in both genders at >170 mg/kg-day (36 and 66% in males and 58 and 3 117% in females), increases in bile acids levels in males at >170 mg/kg-day (233 and 1,110%) 4 and females at 320 mg/kg-day (590%), and decreases in serum cholesterol levels in females at 5 >80 mg/kg-day (23, 39, and 48%, respectively) and males at 320 mg/kg-day (12%). There were 6 no exposure-related changes in rat serum 5'-nucleotidase activity at week 14, although increases 7 occurred on day 5 in females at >20 mg/kg-day and on day 21 in males and females at 80, 170, 8 and/or 320 mg/kg-day. 9 A summary of histopathological alterations following 1,1,2,2-tetrachloroethane exposure 10 is presented in Table 4-4. Hepatic cytoplasmic vacuolization was noted in males exposed to 11 >20 mg/kg-day and in females exposed to >40 mg/kg-day. Although incidence of this alteration 12 was high in affected groups, severity was only minimal-to-mild and only increased with dose 13 from 20 to 40 mg/kg-day in males and 40 to 80 mg/kg-day in females. Females exposed to 14 >80 mg/kg-day showed an increase in the incidence of hepatocyte hypertrophy with an increase 15 in severity and incidence with increasing exposure level, and males showed similar results at 16 exposures >170 mg/kg-day. A statistically significant increase in the incidence of hepatocellular 17 necrosis was observed in male and female rats at 170 and 320 mg/kg-day, accompanied by an 18 increased severity with an increase in dose. At >170 mg/kg-day, additional effects in the liver in 19 both genders were hepatocyte pigmentation and mitotic alteration and mixed cell foci, with bile 20 duct hyperplasia observed in females only. Pigmentation of the spleen was statistically 21 significantly increased in male rats exposed to >80 mg/kg-day and in female rats exposed to 22 >170 mg/kg-day. Other histological effects included statistically significantly increased 23 incidences of atrophy (red pulp and lymphoid follicle) in the spleen of males at 170 and 320 24 mg/kg-day and the spleen of females at 320 mg/kg-day. A statistically significant increase in 25 atrophy of bone (metaphysis) and bone marrow, prostate gland, preputial gland, seminal vesicles, 26 testes (germinal epithelium), uterus, and clitoral gland, as well as an increase in ovarian 27 interstitial cell cytoplasmic alterations, was observed in females at >170 mg/kg-day and in males 28 at 320 mg/kg-day. 29 20 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-4. Incidences of selected histopathological lesions in rats exposed to dietary l,l?2,2-tetrachlorethane for 14 weeks Dose (mg/kg-d) Vehicle control 20 40 80 170 320 Males (10/group) Hepatocyte cytoplasmic vacuolization Hepatocyte hypertrophy Hepatocyte necrosis Hepatocyte pigmentation Hepatocyte mitotic alteration Mixed cell foci Bile duct hyperplasia Spleen pigmentation Spleen red pulp atrophy Spleen lymphoid follicle atrophy Oa 0 0 0 0 0 0 0 0 0 7b(1.3) 0 0 0 0 0 0 0 0 0 9b (2.0) 0 0 0 0 0 0 1 (1.0) 0 0 10b(1.9) 1 (1.0) 0 0 0 0 0 9b(1.0) 0 0 8b(1.4) 9b(1.3) 8b(1.0) 7b(1.0) 0 3 0 9b(1.0) 5b(1.0) 0 0 10b(3.2) 10b(1.6) 10b(1.9) 6b (2.0) 5b 10b(1.7) 9b(1.6) 9b(1.4) 5b(1.0) Females (10/group) Hepatocyte cytoplasmic vacuolization Hepatocyte hypertrophy Hepatocyte necrosis Hepatocyte pigmentation Hepatocyte mitotic alteration Mixed cell foci Bile duct hyperplasia Spleen pigmentation Spleen, red pulp atrophy Spleen lymphoid follicle atrophy oa 0 0 0 0 0 0 1 (1.0) 0 0 0 0 0 0 0 0 0 0 0 0 10b(1.7) 0 0 0 0 0 0 0 0 0 10b (2.2) 4b(1.0) 1 (1.0) 0 0 0 0 4(1.0) 0 0 4b(1.3) 10b(1.7) 7b(1.0) 10b(1.3) 3 (2.0) 8b 5b(1.0) 8b(l.l) 0 0 0 10b(2.8) 10b(l.l) 10b(2.0) 10b(1.9) 1 10b(1.9) 8b(1.3) 9b(1.6) 3(1.0) aValues represent number of animals with the lesion, with the severity score in parenthesis; severity grades are as follows: 1 = minimal, 2 = mild, 3 = moderate, 4 = severe. bSignificantly different from vehicle control group. Source: NTP (2004). 1 2 Epididymal spermatozoal motility was statistically significantly decreased at >40 mg/kg- 3 day, with statistically significant decreases in epididymis weight at >80 mg/kg-day and cauda 4 epididymis weight at 320 mg/kg-day. Exposed female rats spent more time in diestrus and less 5 time in proestrus, estrus, and metestrus than control rats (see Section 4.3.1). 6 In summary, the NTP (2004) 14-week rat study provides evidence that the liver is a 7 primary target of 1,1,2,2-tetrachloroethane toxicity. At the lowest dose tested, 20 mg/kg-day, 8 there was a significant increase in the incidence of hepatic cytoplasmic vacuolization in males. 9 At 40 mg/kg-day, significant increases in relative liver weights were observed in both males and 10 females. Hepatocellular hypertrophy and spleen pigmentation were observed at 80 mg/kg-day in 11 both males and females, although these changes were generally of minimal severity. Increases in 21 DRAFT - DO NOT CITE OR QUOTE ------- 1 serum ALT and SDH, were observed at 80 mg/kg-day in males and at 170 mg/kg-day in females. 2 Decreases in serum cholesterol levels were decreased in females at 80 mg/kg-day and at 320 3 mg/kg-day in males. A decrease in body weight (>10%) was observd at 170 mg/kg-day in both 4 males and females. Increases in serum ALP activity and bile acids levels, hepatocellular necrosis, 5 bile duct hyperplasia, hepatocellular mitotic alterations, foci of cellular alterations, and liver 6 pigmentation occurred at 170 and/or 320 mg/kg-day. A no-observed-adverse-effect level 7 (NOAEL) of 20 mg/kg-day and a lowest-observed-adverse-effect level (LOAEL) of 40 mg/kg- 8 day was identified by EPA for increased relative liver weight in male and female rats. NTP 9 (2004) identified a NOAEL of 20 mg/kg-day in rats based on survival and body weight changes 10 and increased lesion incidences. There were no clinical signs of neurotoxicity at doses as high as 11 320 mg/kg-day or exposure-related findings in the FOB at doses as high as 80 mg/kg-day 12 (highest tested dose in the FOB), indicating that the nervous system may be less sensitive than 13 the liver for subchronic dietary exposure. 14 NTP (2004) also exposed groups of male and female B6C3Fi mice (10/sex/group) to 15 diets containing 0, 589, 1,120, 2,300, 4,550, or 9,100 ppm of microencapsulated 1,1,2,2-tetra- 16 chloroethane for 14 weeks, with vehicle and untreated control groups for each gender. The 17 reported average daily doses were 0, 100, 200, 370, 700, or 1,360 mg/kg-day for males and 0, 80, 18 160, 300, 600, or 1,400 mg/kg-day for females. Endpoints evaluated throughout the study 19 included clinical signs, body weight, and feed consumption. Clinical chemistry was assessed at 20 the end of the study, but hematological evaluations and urinalyses were not performed. 21 Necropsies were conducted on all animals and selected organs (liver, heart, right kidney, lung, 22 right testis, and thymus) were weighed. Comprehensive histological examinations were 23 performed on untreated control, vehicle control, and high dose groups. Tissues examined in the 24 lower dose groups were limited to the liver, spleen, and thymus in both genders; preputial gland 25 in males; and lungs in females. An FOB (21 parameters) was performed on mice in both control 26 and 160/200, 300/370, and 600/700 mg/kg-day (1,120, 2,300, and 4,550 ppm, respectively) dose 27 groups during weeks 4 and 13. Sperm motility, vaginal cytology, estrous cycle length, and 28 percentage of time spent in the various estrus stages were evaluated in both control and 160/200, 29 600/700, and 1,360/1,400 mg/kg-day (1,120, 2,300, and 4,550 ppm, respectively) dose groups. 30 All mice survived to the end of the study (NTP, 2004). Thinness was observed clinically 31 in male mice (3/10, 9/10, 10/10) at 370, 700, and 1,400 mg/kg-day, respectively, and in female 32 mice (1/10, 2/10, 10/10) at 300, 600, and 1,360 mg/kg-day, respectively. Final body weights 33 were statistically significantly lower than vehicle controls in male mice at 370, 700, and 34 1,360 mg/kg-day (12, 16, and 23%, respectively) and female mice at 600 and 1,400 mg/kg-day 35 (11 and 12%, respectively) (Table 4-5). Feed consumption was less than controls in males at 36 >700 mg/kg-day, but similar to controls in females. 37 22 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-5. Final body weights (g) and percent change compared to controls in B6C3Fi mice exposed to 1,1^2,2-tetrachloroethane in feed for 14 weeks Dose (mg/kg-d) Vehicle control 100 200 370 700 1,360 n 10 10 10 10 10 10 Vehicle control 80 160 300 600 1,400 10 10 10 10 10 10 Males 30.1±0.6a 30.6 ±0.6 30.0 ±0.3 26.5±0.4b 25.2±0.2b 23.1±0.5b - 2% 0 -12 -16 -23 Females 24.3±0.5a 24.2 ±0.2 24. 3 ±0.6 23. 3 ±0.4 21.7±0.2b 21.5±0.6b - 0% 0 -4 -11 -12 1 2 3 4 5 6 7 8 9 10 11 "Mean ± standard error. V<0.05. Source: NTP (2004). Statistically significant increases in absolute liver weights were observed in the male mice exposed to 200 and 370 mg/kg-day (16 and 10%, respectively), but not at higher doses, and in female mice exposed to >80 mg/kg-day (11, 29, 27, 22, and 32%, respectively) (Table 4-6a). Statistically significant increases in relative liver weights were observed in male mice at >200 mg/kg-day (16, 24, 24, and 38%, respectively) and in female mice at >80 mg/kg-day (11, 28, 33, 36, and 49%, respectively) (Table 4-6b). Other organ weight changes (increased kidney weights in males at >370 mg/kg-day and decreased thymus weights in both genders at 1,360/ 1,400 mg/kg-day) were considered to be secondary to the body weight changes. Results of the FOBs showed no exposure-related neurotoxicity. 23 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-6a. Absolute liver weights (g) and percent change compared to controls in B6C3Fi mice exposed to l,l?2,2-tetrachloroethane in feed for 14 weeks Dose (mg/kg-d) Vehicle control 100 200 370 700 1,360 n 10 10 10 10 10 10 Vehicle control 80 160 300 600 1,400 10 10 10 10 10 10 Males 1.467 ±0.020 1.557±0.039 1.701±0.020b 1.607 ±0.038b 1.531 ±0.052 1.558±0.045 - 6% 16 10 4 6 Females 1.048 ±0.028 1.160±0.022b 1.356±0.058b 1.336±0.037b 1.277 ±0.030b 1.386±0.047b - 11% 29 27 22 32 "Mean ± standard error. V<0.05. Source: NTP (2004). Table 4-6b. Relative liver weights (mg organ weight/g body weight) and percent change compared to controls in B6C3Fi mice exposed to l,l?2,2-tetrachloroethane in feed for 14 weeks Dose (mg/kg-d) Vehicle control 100 200 370 700 1,360 n 10 10 10 10 10 10 Vehicle control 80 160 300 600 1,400 10 10 10 10 10 10 Males 48.84±1.17 50. 94 ±0.93 56.82±0.63b 60.63 ±1.20b 60.71±1.76b 67.43 ±1.83b - 4% 16 24 24 38 Females 43.26 ±1.05 47.90±0.85b 55.54±1.17b 57.39±0.84b 58.73 ±1.23b 64.42 ±1.14b - 11% 28 33 36 49 aMean ± standard error. V<0.05. Source: NTP (2004). 24 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 9 10 11 12 13 14 15 16 17 18 19 20 Clinical chemistry findings in the mice are summarized in Tables 4-7 and 4-8 and included statistically significant decreases in total serum protein levels in males at >200 mg/kg- day, total serum protein levels in females at >300 mg/kg-day, and serum albumin levels in females at 1,400 mg/kg-day (NTP, 2004). Decreased serum albumin levels could not fully account for the decreased total protein levels, suggesting that other factors (e.g., changes in other protein fractions, hydration status, and/or hepatic function) contributed to the hypoproteinemia (NTP, 2004). A statistically significant increase of serum SDH activity in females was observed at>80 mg/kg-day (22, 111, 444, 575, and 1,181%, respectively) and in males at>200 mg/kg-day (38, 424, 424, and 715%, respectively). A statistically significant decrease in serum cholesterol levels was observed in females at >160 mg/kg-day (22, 38, 41, and 16%, respectively), and a statistically significant increase in ALT activity was observed in females at >160 (30, 278, 294, and 602%, respectively) and in males at >370 mg/kg-day (234, 177, and 377%, respectively). Total bile acids levels increased statistically significantly in females at>160 mg/kg-day (18, 69, 97, and 290%, respectively) and in males at >370 mg/kg-day (148, 178, and 377%, respectively). A statistically significant increase in ALP activity was observed in males (67, 83, and 136%, respectively) and in females at 300 mg/kg-day (19, 28, 55%, respectively) at, and a statistically significant increase in 5'-nucleotidase was observed in males at >370 mg/kg-day (88, 131, and 288%, respectively). Table 4-7. Selected clinical chemistry changes in male mice exposed to dietary I,l92,2-tetrachloroethane for 14 weeks Dose (mg/kg-d) Serum total protein (g/dL) Serum cholesterol (mg/dL) ALT (IU/L) ALP (IU/L) SDH (IU/L) 5'-Nucleotidase (IU/L) Bile acids (umol/L) Vehicle control 5.4±0.1a 131±7 66 ±8 85 ±2 55 ±3 18±1 25. 3 ±1.2 100 5.2±0.1 125 ±4 62 ±19 78 ±2 53 ±2 16±1 22.8 ±1.5 200 5.1±0.1b 94±3b 74 ±8 89 ±2 76±3b 18±0 24.8 ±0.6 370 5.1±0.1b 110±5 207±18b 130 ±3b 288 ± 20b 30±2b 56.5 ±5. lb 700 5.1±0.1b 112±4 172±18b 143 ±7b 288±29b 37±3b 63.3±7.5b 1,360 5.1±0.1b 126 ±5 296 ± 24b 184±llb 448±25b 62±7b 108.7 ±8.1b aMean ± standard error. Statistically significantly different from control value. Source: NTP (2004). 21 25 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-8. Selected clinical chemistry changes in female mice exposed to dietary l,l?2,2-tetrachloroethane for 14 weeks Dose (mg/kg-d) Serum total protein (g/dL) Serum cholesterol (mg/dL) ALT (IU/L) ALP (IU/L) SDH (IU/L) 5'-Nucleotidase (IU/L) Bile acids (umol/L) Vehicle control 5.6±0.1a 109 ±2 34 ±5 131±5 36 ±1 59 ±3 27.2 ±1.2 80 5.6±0.1 109 ±3 50 ±15 126 ±2 44±3b 71±2 26.1 ±1.9 160 5. 5 ±0.0 85±3b 65±5b 139±5 76±4b 84±5b 30.9±l.lb 300 5.4±0.1b 68±2b 189±33b 150 ±3b 197±15b 62 ±2 44.2±3.9b 600 5.4±0.0b 64±3b 197±21b 161 ±7b 243 ± 23b 62 ±3 51.5±3.6b 1,400 5.1±0.1b 92±4b 351±35b 195 ±6b 461±59b 83±4b 101.7 ±12.0b 1 2 3 4 5 6 7 8 9 10 11 12 13 14 "Mean ± standard error. bStatistically significantly different from control value. Source: NTP (2004). The histopathological results in the B6C3Fi mice are summarized in Table 4-9. A statistically significant increased incidence of minimal to moderate hepatocyte hypertrophy was observed at >160 mg/kg-day in females and >200 mg/kg-day in males. The incidence of hepatocellular necrosis was statistically significantly increased in male mice at >370 mg/kg-day and in female mice at >300 mg/kg-day. A statistically significant increased incidence of pigmentation and bile duct hyperplasia occurred at >300 mg/kg-day in females and >370 mg/kg- day in males. Additionally, the histological findings included an increased incidence of preputial gland atrophy in males in the 100, 700, and 1,360 mg/kg-day dose groups (Table 4-9), but this effect did not appear dose-related. Based on the increase in serum SDH activity and increased absolute and relative liver weights at 80 mg/kg-day in female mice, as well as serum chemistry changes at > 160 mg/kg-day and clear evidence of histopathology at higher doses, a LOAEL of 80 mg/kg-day was identified based on liver toxicity. 26 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-9. Incidences of selected histopathological lesions in mice exposed to dietary l,l?2,2-tetrachloroethane for 14 weeks Males (10/group) Oral dose (mg/kg-d) Hepatocyte hypertrophy Hepatocyte necrosis Liver focal pigmentation Bile duct hyperplasia Preputial gland atrophy Vehicle control Oa 0 0 0 0 100 0 0 0 0 4b (2.0) 200 7b(1.0) 1 (2.0) 0 0 2(1.0) 370 10b (2.2) 8b(l.l) 10b(1.2) 7b(1.4) 0 700 10b (2.8) 8b(1.0) 10b(1.4) 9b(1.3) 4b (2.5) 1,360 10b(3.1) 9b(1.0) 8b(1.3) 10b (2.0) 5b (2.2) Females (10/group) Oral dose (mg/kg-d) Hepatocyte hypertrophy Hepatocyte necrosis Liver focal pigmentation Bile duct hyperplasia Vehicle control Oa 0 0 0 80 2(1.5) 0 0 0 160 9b(1.0) 0 2(1.0) 0 300 10b(1.9) 3 (1.0) 9b(1.0) 8b(1.0) 600 10b (2.5) 7b(1.0) 8b(1.0) 10b(1.4) 1,400 10b (3.0) 4b(1.0) 7b(l.l) 10b (2.0) aValues represent number of animals with the lesion, with the severity score in parenthesis; severity grades are as follows: 1 = minimal, 2 = mild, 3 = moderate, 4 = severe. Significantly different from vehicle control group. Source: NTP (2004). 1 2 4.2.1.2. Chronic Studies 3 Information on the chronic oral toxicity of 1,1,2,2-tetrachloroethane is available from a 4 bioassay in rats and mice. NCI (1978) exposed groups of 50 male and 50 female Osborne- 5 Mendel rats to 1,1,2,2-tetrachloroethane in corn oil via gavage 5 days/week for 78 weeks. 6 Vehicle and untreated control groups (20 animals/sex/species) were also used. The initial low 7 and high doses used for rats of both genders were 50 and 100 mg/kg-day. At week 15, the doses 8 were raised to 65 mg/kg-day for low-dose males and 130 mg/kg-day for high dose males. At 9 week 26, the doses were decreased to 40 mg/kg-day for the low-dose females and 80 mg/kg-day 10 for the high-dose females. Beginning at week 33, intubation of all high-dose rats was suspended 11 for 1 week followed by 4 weeks of dosing, and this cyclic pattern of dosing was maintained for 12 the remainder of the treatment period. Low-dose rats were not subject to this regimen. The 13 reported time-weighted average (TWA) doses were 62 and 108 mg/kg for male rats and 43 and 14 76 mg/kg for female rats. The exposure period was followed by a 32-week observation period in 15 which the rats were not exposed to 1,1,2,2-tetrachloroethane. Clinical signs, survival, body 16 weight, food consumption, gross pathology, and histology (32 major organs and tissues as well 17 as gross lesions) were evaluated. 18 There were no clear effects on survival in the male rats. In females, survival in the 19 vehicle control, low-dose, and high-dose groups at the end of the study was 70, 58, and 40%, 27 DRAFT - DO NOT CITE OR QUOTE ------- 1 respectively. Although there was a statistically significant association between increased 2 mortality and dose in the females, the increased mortality was affected by the deaths of 10 high- 3 dose females, 8 with pneumonia and 2 with no reported lesions, during the first 5 weeks of the 4 study. The study authors also stated that there was no evidence that the early deaths were tumor- 5 related. The male and female rats also demonstrated an increased incidence of endemic chronic 6 murine pneumonia. Incidences of chronic murine pneumonia in the vehicle control, low-, and 7 high-dose groups were 40, 68, and 76% in females and 55, 50, and 65% in males. Clinical 8 observations included squinted or reddened eyes in all control and treated groups of both genders, 9 but these effects occurred with greater frequency in the exposed rats. There was a low or 10 moderate incidence of labored breathing, wheezing, and/or nasal discharge in all control and 11 treated groups during the first year of the study, and near the end of the study these signs were 12 observed more frequently in the exposed animals. 13 Dose-related decreases in body weight gain were observed. However, as the study 14 approached termination (weeks 100-110), the differences in body weight across the dose groups 15 decreased. 16 Histopathological effects included a dose-related increased incidence of hepatic fatty 17 metamorphosis in high-dose males (2/20, 0/20, 2/50, and 9/49 in the untreated control, vehicle 18 control, low-dose, and high-dose groups, respectively). In addition, inflammation, focal cellular 19 changes, and angiectasis were observed in male and female rats but were not statistically 20 significant or biologically relevant. NCI (1978) stated that the inflammatory, degenerative, and 21 proliferative lesions observed in the control and dosed animals were similar in incidence and 22 type to those occurring in naturally aged rats. 23 A statistically significant increase in tumor incidence was not observed in the rats; 24 however, two hepatocellular carcinomas, which are rare tumors in male Osborne-Mendel rats 25 (NCI, 1978), as well as one neoplastic nodule, were observed in the high-dose males 26 (Table 4-10). A hepatocellular carcinoma was also observed in an untreated female control. 27 Although interpretation of this study is complicated by the chronic murine pneumonia, it is 28 unlikely to have contributed to the fatty metamorphosis observed in the liver of male rats. 29 28 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-10. Incidence of neoplasms in male Osborne-Mendel rats exposed to 1,1^2,2-tetrachloroethane in feed for 78 weeks Neoplasm Papilloma, stomach Squamous cell carcinoma, stomach Neoplastic nodule/carcinoma, liver Follicular-cell carcinoma, thyroid Hemangiosarcoma, all sites Adenocarcinoma, mammary gland Fibroadenoma, mammary gland Chromophode adenomas, pituitary Islet-cell adenomas, pancreatic islets Fibroma, subcutaneous tissue Dose (mg/kg-d) Control Vehicle control 62 108 Males 0/20 0/20 0/20 1/19 0/20 1/20 1/20 2/20 0/20 0/20 0/20 0/20 0/20 3/20 0/20 2/20 1/'20 5/14 2/20 1/20 0/50 0/50 0/50 0/49 2/50 2/50 1/50 5/48 2/49 2/50 1/48 1/48 3/49 2/48 3/49 0/49 0/49 5/48 2/49 2/49 Source: NCI (1978). 1 2 In addition, one papilloma of the stomach, one squamous-cell carcinoma of the stomach, 3 two follicular-cell carcinomas of the thyroid, and three hemangiosarcomas were each observed in 4 high-dose males (Table 4-10). In the low-dose males, two mammary gland adenocarcinomas 5 (2/20 in vehicle controls) and two hemangiosarcomas (0/20 in vehicle control) were observed. 6 Adenomas were observed as follows: pituitary chromophobe adenomas in the vehicle control 7 (5/14) and low- and high-dose males (5/48 and 5/48, respectively); pancreatic islet-cell 8 adenomas in the vehicle control (2/20) and low- and high-dose males (2/49 and 2/49, 9 respectively); mammary gland fibroadenomas in the vehicle control (1/20) and low-dose males 10 (1/50); and subcutaneous tissue fibromas in the vehicle control (1/20) and low- and high-dose 11 females (2/50 and 2/49, respectively). In male rats, the incidence of chromophobe adenomas, 12 islet-cell adenomas, and follicular-cell carcinomas in the vehicle controls was significantly 13 increased over the incidence in historical controls (NCI, 1978). 14 In the female rats (Table 4-11), one follicular-cell carcinoma was observed in both the 15 low- and high-dose groups. One mammary gland adenocarcinoma was observed in a low-dose 16 female, and two were observed in the high-dose group. One hemangiosarcoma was observed in 17 a low-dose female. Adenomas were observed as follows: pituitary chromophobe adenomas in 18 the vehicle control (3/20) and low- and high-dose females (11/49 and 6/48, respectively); one 19 pancreatic islet-cell adenoma in a low-dose female; mammary gland fibroadenomas in the 20 vehicle control (9/20) and low- and high-dose females (13/50 and 11/50, respectively); and 21 subcutaneous tissue fibromas in the vehicle control (1/20) and low- and high-dose females 22 (2/50 and 1/50, respectively). The incidence of fibroadenomas of the mammary gland in the 29 DRAFT - DO NOT CITE OR QUOTE ------- 1 vehicle control group was statistically significantly increased over the incidence in historical 2 controls (NCI, 1978). 3 Table 4-11. Incidence of neoplasms in female Osborne-Mendel rats exposed to l,l?2,2-tetrachloroethane in feed for 78 weeks Neoplasm Adenocarcinoma, mammary gland Fibroadenoma, mammary gland Hemangiosarcomas, uterus Chromophode adenomas, pituitary Islet-cell adenomas, pancreatic islets Follicular-cell carcinoma, thyroid Fibroma, subcutaneous tissue Dose (mg/kg-d) Control Vehicle control 43 76 Females 2/20 2/20 0/20 6/19 1/20 0/20 0/20 0/20 9/20 0/20 3/20 0/20 0/20 1/20 1/50 13/50 1/50 11/49 1/50 1/49 2/50 2/50 11/50 0/50 6/48 0/50 1/50 1/50 4 5 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 Source: NCI (1978). NCI (1978) also exposed groups of 50 male and 50 female B6C3Fi mice to 1,1,2,2-tetra- chloroethane in corn oil via gavage 5 days/week for 78 weeks. Initial dose levels were 100 and 200 mg/kg-day in both genders. In week 19, the doses were increased to 150 and 300 mg/kg-day, respectively. Three weeks later, the doses were increased to 200 and 400 mg/kg-day, respectively. In week 27, the doses were decreased to 150 and 300 mg/kg-day, respectively. The reported TWA doses were 142 and 284 mg/kg for male and female mice. The exposure period was followed by a 12-week observation period in which the mice were not exposed to 1,1,2,2-tetrachloroethane. Vehicle and untreated control groups (20 animals/sex) and a pooled vehicle control were also used. The pooled vehicle control group comprised the vehicle controls from the studies of 1,1,2,2-tetrachloroethane and chloropicrin. Clinical signs, survival, body weight, food consumption, gross pathology, and histology (32 major organs and tissues as well as gross lesions) were evaluated. A statistically significant association between mortality and dose was observed, as survival was markedly decreased in the high-dose male and female mice. Terminal survival data were not reported for the males, although acute toxic tubular nephrosis was determined to be the apparent cause of death in 33 high-dose males dying between weeks 69 and 70. Survival in the vehicle control, low-dose, and high-dose females at the end of the study was 75, 74, and 34%, respectively, but the cause of death in the high-dose females was not reported. The male and female mice also demonstrated an increased incidence of endemic chronic murine pneumonia. Incidences of chronic murine pneumonia in the vehicle control, low-, and high-dose groups were 11, 0, and 2% in males and 5, 13, and 18% in females. 30 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 9 10 A high incidence (approximately 95%) of pronounced abdominal distension, possibly resulting from liver tumors, was observed in the high-dose females beginning in week 60 and continuing throughout the recovery period. Nodular hyperplasia and organized thrombus were observed in male and female mice, but the incidences were not statistically significant. Nonneoplastic lesions observed included hydronephrosis (16/46) and chronic inflammation in the kidneys (5/46) in high-dose females and chronic inflammation in the low- (13/39) and high- dose (10/47) males (Table 4-12). In addition, acute toxic tubular nephrosis was observed, and was the apparent cause of death as identified by the study authors, in high-dose male mice that died during weeks 69 and 70. Table 4-12. Incidence of nonneoplastic kidney lesions observed in male and female B6C3Fi mice exposed to l,l?2,2-tetrachloroethane in feed for 78 weeks Lesion Chronic inflammation - kidney Hydro nephro sis Chronic inflammation Dose (mg/kg-d) Control Vehicle control 142 284 Males 7/19 5/18 13/39 10/47 Females 0/19 0/19 0/20 0/20 0/46 0/46 16/46 5/46 11 12 13 14 15 16 17 18 19 20 21 22 23 24 Source: NCI (1978). Statistically significant increases in the incidences of hepatocellular carcinomas occurred in both sexes and at both dose levels (Table 4-13). The incidences in the vehicle control, pooled vehicle control, 142, and 284 mg/kg-day groups were 1/18, 3/36, 13/50, and 44/49, respectively, in males and 0/20, 1/40, 30/48, and 43/47, respectively, in females. Information on the progression from preneoplastic pathology to hepatocellular carcinoma is not available due to the lack of interim sacrifices. The hepatocellular carcinomas varied in microscopic appearance, with some tumors composed of well-differentiated cells and a relatively uniform rearrangement of cords, while other tumors were composed of anaplastic cells with large hyperchromatic nuclei with eosinophilic inclusion bodies and/or vacuolated pale cytoplasm. In addition, a decrease in the time to tumor for the hepatocellular carcinomas was also evident in both genderss of mice. The spontaneous tumor rate for hepatocellular carcinoma in the historical vehicle controls at the testing laboratory was 74/612 (12%) for male B6C3Fi mice and 8/560 for female B6C3Fi mice. 31 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-13. Incidence of hepatocelluar carcinomas in male and female B6C3Fi mice exposed to l,l?2,2-tetrachloroethane in feed for 78 weeks Hepatocellular carcinoma Incidence Time to first tumor Incidence Time to first tumor Dose (mg/kg-d) Vehicle control Pooled vehicle control 142 284 Males 1/18 72 3/36 NA 13/503 84 44/49a 52 Females 0/20 NA 1/40 NA 30/483 58 43/47a 53 1 2 3 4 5 "Significantly different from control groups. Source: NCI (1978). In addition to the liver tumors, alveolar/bronchiolar adenomas in the lung were observed in the male matched vehicle controls (1/18), male and female pooled-vehicle controls (1/36 and 1/40, respectively), low-dose males and females (2/39 and 1/46, respectively), and high-dose males and females (2/47 and 1/44, respectively) (Table 4-14). Lymphomas were observed in low- and high-dose males (4/50 and 3/49, respectively), and in female pooled vehicle controls (2/40) and low- and high-dose females (7/48 and 3/47, respectively). Table 4-14. Incidence of additional neoplasms in male and female B6C3Fi mice exposed to 1,1^2,2-tetrachloroethane in feed for 78 weeks Neoplasm Alveolar/bronchiolar adenomas, lung Lymphomas, multiple organ Alveolar/bronchiolar adenomas, lung Lymphomas, multiple organ Dose (mg/kg-d) Matched control Pooled vehicle control 142 284 Males 1/18 0/18 1/36 0/36 2/39 4/50 2/47 3/49 Females 0/20 0/20 1/40 2/40 1/46 7/48 1/44 3/47 9 10 11 12 13 14 15 Source: NCI (1978). For chronic inflammation in the kidneys of male mice, a LOAEL of 142 mg/kg-day was selected. A NOAEL was not identified. For hydronephrosis and chronic inflammation in the kidneys in females, a NOAEL of 142 mg/kg-day and a LOAEL of 284 mg/kg-day were selected. 4.2.2. Inhalation Exposure 4.2.2.1. Subchronic Studies 32 DRAFT - DO NOT CITE OR QUOTE ------- 1 Truffert et al. (1977) exposed groups of female Sprague-Dawley rats (55/dose) to 2 1,1,2,2-tetrachloroethane vapor at reported calculated atmospheric concentrations of 0 or 3 560 mL/m3 5 days/week for 15 weeks (78 exposures). The daily exposure duration was 6 hours 4 for the first 8 exposures and 5 hours for the remaining 70 exposures. There is uncertainty 5 regarding the actual concentration employed due to the unusual unit of exposure (i.e., mL/m ). It 6 is assumed that mL/m3 is a volume/volume vapor concentration, so the reported concentration is 7 equivalent to 560 ppm (3,909 mg/m3). Interim sacrifices were conducted after 2, 4, 9, 19, 39, 8 and 63 exposures, although the number of animals killed at each time period was not reported. 9 This study is limited by poor reporting quality and minimal quantitative data. 10 Pronounced prostration was observed "after the first exposures to 1,1,2,2-tetrachloroethane, 11 followed by recovery". Body weight gain was decreased at the end of the study, but the 12 magnitude of the change was not reported. Increases in relative liver weights were observed 13 beginning 15 days after exposure initiation, but were not quantified. Hematological alterations 14 consisting of a decrease in hematocrit "confirmed by the joint RBC and WBC counts" were 15 observed at the end of the study, but were not quantified. A marked increase (313%) in 16 thymidine uptake in hepatic DNA was observed after four exposures, but by the ninth exposure 17 the thymidine uptake had decreased to levels similar to controls. Histological alterations were 18 observed in the liver after nine exposures and included granular appearance, cytoplasmic 19 vacuolization, and evidence of hyperplasia (increase in the number of binucleated cells and the 20 appearance of mitosis), but the alterations regressed after 19 exposures and were no longer 21 observed after 39 exposures. Incidences and severity of the liver lesions were not reported. 22 Considering the lack of incidence and severity data and other inadequately reported results, lack 23 of information on dose-response due to the use of a single exposure level, and uncertainty 24 regarding the exposure concentration, a NOAEL or LOAEL cannot be identified from this study. 25 Horiuchi et al. (1962) exposed one adult male monkey (Macaca cynomolga Linne] to 26 1,1,2,2-tetrachloroethane for 2 hours/day, 6 days/week for a total of 190 exposures in 9 months. 27 The exposure level was 2,000-4,000 ppm (13,700-27,500 mg/m3) for the first 20 exposures, 28 1,000-2,000 ppm (6,870-13,700 mg/m3) for the next 140 exposures, and 3,000^,000 ppm 29 (20,600-27,500 mg/m ) for the last 30 exposures. The TWA concentration was 1,974 ppm 30 (13,560 mg/m3). The authors noted that the monkey was weak after approximately seven 31 exposures and had diarrhea and anorexia between the 12th and 15th exposures. Beginning at the 32 15th exposure, the monkey was "almost completely unconscious falling upon his side" for 20- 33 60 minutes after each exposure. The authors noted a gradual increase in body weight during 34 months 3-5 followed by a gradual decrease until the study was terminated. Hematological 35 parameters demonstrated sporadic changes in hematocrit and RBC and WBC counts, but the 36 significance of these findings cannot be determined because there were no clear trends, only one 37 monkey was tested, and there was no control group. Histological alterations consisted of fatty 38 degeneration in the liver and splenic congestion, and no effects were observed in the heart, lung, 33 DRAFT - DO NOT CITE OR QUOTE ------- 1 kidneys, pancreas, or testes. This study cannot be used to identify a NOAEL or LOAEL for 2 subchronic exposure due to the use of a single animal without a control. 3 A 6-month inhalation study in rats was performed by the Mellon Institute of Industrial 4 Research (1947). Groups of 12 male and 12 female albino rats were exposed to 0 or 167 ppm 5 (1,150 mg/m3) of 1,1,2,2-tetrachloroethane for 7 hours/day on alternate days for the 6-month 6 study period. A statistically significant increase (15%) in kidney weight was observed in the 7 1,1,2,2-tetrachloroethane-exposed rats. The rats also appeared to develop lung lesions following 8 exposure to tetrachloroethane; however, the study authors stated that the pathology reported for 9 tetrachloroethane must be discounted due to approximately 50% of the control animals 10 demonstrating major pathology of the kidneys, liver, or lung. Meaningful interpretation of these 11 results is precluded by the observed endemic lung infection, which resulted in significant early 12 mortality in all of the rats (57 and 69% mortality in the control and tetrachloroethane-exposed 13 groups, respectively). This study also included one mongrel dog that followed the same study 14 design and evaluation as the rats. Serum phosphatase activity levels, mean of 33 units/100 mL, 15 and blood urea nitrogen levels, mean of 20.66%, were increased in the treated dog compared to 16 control values of 5.72/100 mL and 14.94%, respectively. The dog survived the 6-month 17 exposure with effects that included cloudy swelling of the liver and of the convoluted tubules of 18 the kidneys, and light congestion of the lungs. Identification of a LOAEL or NOAEL is 19 precluded by poor study reporting, high mortality in the rats, and the use of a single treated 20 animal in the dog study. 21 Kulinskaya and Verlinskaya (1972) examined effects of 1,1,2,2-tetrachloroethane on the 22 blood acetylcholine system in Chinchilla rabbits exposed to 0 or 10 mg/m3 (0 or 1.5 ppm) 23 3 hours/day, 6 days/week for 7-8.5 months. The animals were immunized twice, at 1.5-2 and 24 4 months, subcutaneously with a 1.2 and 1.5 billion microbe dose of typhoid vaccine in an 25 attempt to reveal changes in the immunological reactivity following 1,1,2,2-tetrachloroethane 26 exposures. The exposed group contained six animals, and the size of the control group was not 27 specified. In comparison with both initial and control levels, serum acetylcholine levels were 28 decreased after 1.5 months, significantly increased after 4.5 months, and significantly decreased 29 at the end of the study. The concentration of acetylcholine in the blood was increased following 30 the first immunization. No changes in serum acetylcholinesterase activity were reported, 31 although serum butyrylcholinesterase activity was reduced after 5-6 months of exposure. This is 32 a poorly reported study that did not examine any other relevant endpoints. A NOAEL or 33 LOAEL could not be identified because the changes in acetylcholine levels were inconsistent 34 across time and incompletely quantified, and the biological significance of the change is unclear. 35 36 4.2.2.2. Chronic Studies 37 In a chronic inhalation study by Schmidt et al. (1972), groups of 105 male rats were 38 exposed to 0 or 0.0133 mg/L (13.3 mg/m3) 1,1,2,2-tetrachloroethane for 4 hours daily for up to 34 DRAFT - DO NOT CITE OR QUOTE ------- 1 265 days. Subgroups of seven treated and seven control rats were killed after 110 or 265 days of 2 exposure and 60 days after exposure termination, with the remaining animals observed until 3 natural death. There were no significant alterations in survival. Weight gain in exposed rats was 4 2.1, 11.6, and 12.2% less than controls on study days 110, 260, and 324, although the only 5 statistically significant decreases in body weight gain occurred between days 90 and 170. Other 6 statistically significant changes included increased leukocyte (89%) and Pi-globulin (12%) levels 7 compared to controls after 110 days, and an increased percentage of segmented nucleated 8 neutrophils (36%), decreased percentage of lymphocytes (17%), and increased percentage of 9 liver total fat content (34%) after 265 days. There was a statistically significant decrease in 10 y-globulin levels (32%) at 60 days postexposure and a decrease in adrenal ascorbic acid content 11 (a measure of pituitary adrenocorticotropic hormone [ACTH] activity) at all three time periods 12 (64, 21, and 13%, respectively). This study is insufficient for identification of a NOAEL or 13 LOAEL for systemic toxicity because the experimental design and results were poorly reported, 14 and histological examinations were not conducted. 15 16 4.3. REPRODUCTIVE/DEVELOPMENTAL STUDIES—ORAL AND INHALATION 17 4.3.1. Oral Exposure 18 Gulati et al. (199la) exposed timed-pregnant CD Sprague-Dawley rats (8-9 animals/ 19 group) to diets containing 0, 0.045, 0.135, 0.27, 0.405, or 0.54% microencapsulated 20 1,1,2,2-tetrachloroethane from gestation days (GDs) 4 through 20. Based on body weight and 21 food consumption data, the reported estimated doses of 1,1,2,2-tetrachloroethane were 0, 34, 98, 22 180, 278, or 330 mg/kg-day. Dams were sacrificed and litters were evaluated on GD 20. 23 Evaluations included maternal body weight, feed consumption and clinical signs, uterine weight, 24 and numbers of implantations, early and late resorptions, live fetuses, and dead fetuses. 25 Necropsies were performed on the maternal animals, but fetuses were not examined for 26 malformations. 27 All dams survived to study termination on GD 20. Maternal body weight was 28 statistically significantly decreased 9, 11, 14, and 24% at 98, 108, 278, and 330 mg/kg-day, 29 respectively, compared to controls, and demonstrated a dose-dependent and time-dependent 30 decrease in all dose groups. However, an increase in maternal body weight on day 20, compared 31 to body weight on day 4, was apparent for all dose groups. Daily food consumption was 32 significantly decreased in all dose groups, and this may have contributed to the decreased body 33 weights observed in the study. Four out of nine rats in the 278 mg/kg-day dose group had 34 slightly rough fur beginning on GD 10, while rough fur was present in all animals in the 35 330 mg/kg-day dose group. No statistically significant changes were observed in the numbers of 36 live fetuses/litter, dead fetuses/litter, resorptions/litter, or implants/litter. One dam in the 37 98 mg/kg-day group and four of nine dams in the 330 mg/kg-day group completely resorbed 38 their litters. At scheduled sacrifice, average fetal weights were statistically significantly 3 5 DRAFT - DO NOT CITE OR QUOTE ------- 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 decreased 3.9, 12.7, 10.5, and 20.6% in the 98, 108, 278, and 330 mg/kg-day dose groups, respectively (Table 4-15). Gravid uterine weight was statistically significantly reduced only in the 330 mg/kg-day animals. Small, but statistically significant, decreases were seen in maternal body weight and average fetal weight at >98 mg/kg-day. Using statistical significance and a 10% change as the criterion for an adverse change in maternal body weight, a NOAEL of 34 mg/kg-day and LOAEL of 98 mg/kg-day were selected for changes in maternal body weight. A NOAEL of 34 mg/kg-day and LOAEL of 98 mg/kg-day were selected for developmental toxicity based on the lowest dose that produced a statistically significant decrease in fetal body weight. Table 4-15. Fetal body weight in CD Sprague-Dawley rats exposed to microencapsulated l,!92,2-tetrachloroethane on gestation days (CDs) 4-20 Dose (mg/kd-day) 0 34 98 180 278 330 N 9 8 8 9 9 5 Mean 2.28 2.17 2.19 1.99 2.04 1.81 SD 0.12 0.11 0.08 0.15 0.42 0.26 % change 4.8 3.9 12.7 10.5 20.6 Source: Gulati et al. (1991) Gulati et al. (1991b) exposed timed-pregnant Swiss CD-I mice (n = 5-11) to diets containing 0, 0.5, 1, 1.5, 2, or 3% microencapsulated 1,1,2,2-tetrachloroethane from GDs 4 through 17. Based on body weight and food consumption data, the reported estimated doses of 1,1,2,2-tetrachloroethane were 0, 987, 2,120, 2,216, or 4,575 mg/kg-day; an average dose could not be calculated for the 3% group due to early mortality. Dams were sacrificed and litters were evaluated on GD 17. Evaluations included maternal body weight, feed consumption and clinical signs, uterine weight, and numbers of implantations, early and late resorptions, live fetuses, and dead fetuses. Necropsies were performed on the maternal animals, but fetuses were not examined for malformations. All animals (9/9) in the 3% group died prior to the end of the study. Mortality was 0/11, 0/9, 2/10, 4/5, and 5/7 in the 0, 987, 2,120, 2,216, or 4,575 mg/kg-day groups, respectively, and the mortality in the higher dose groups affected the statistical power of the study for those groups. Maternal body weights were statistically significantly decreased compared to controls at >2,120 mg/kg-day beginning on study day 9, although the day 17 data were not statistically significantly different from controls for any treatment group. Average daily feed consumption was statistically significantly decreased in all treated groups except in the 987 mg/kg-day animals. Gross hepatic effects were reported in dams from all groups except the 987 mg/kg-day 36 DRAFT - DO NOT CITE OR QUOTE ------- 1 group and included pale or grey and/or enlarged livers and a prominent lobulated pattern. 2 Complete litter resorption occurred in 1/11, 0/9, 2/8, 1/1, and 1/2 dams in the 0, 987, 2,120, 3 2,216, and 4,575 mg/kg-day groups, respectively. No changes in developmental endpoints were 4 noted in the 987 or 2,120 mg/kg-day groups. The 2,120 and 4,575 mg/kg-day groups had too 5 few litters, due to maternal toxicity, to permit statistical analysis of the findings. The high 6 mortality in the exposed mice precluded the identification of a NOAEL or LOAEL for this study. 7 NTP (2004) conducted a 14-week study in which groups of 10 male and 10 female 8 F344 rats were fed diets containing microencapsulated 1,1,2,2-tetrachloroethane at reported 9 average daily doses of 0, 20, 40, 80, 170, or 320 mg/kg-day. The main part of this study is 10 summarized in Section 4.2.1.1. Reproductive function (fertility) was not evaluated. Endpoints 11 relevant to reproductive toxicity included histology (testis with epididymis and seminal vesicle, 12 preputial gland, prostate gland, clitoral gland, ovary, and uterus) and weights (left cauda 13 epididymis, left epididymis, and left testis) of selected reproductive tissues in all control and 14 treated groups. Sperm evaluations and vaginal cytology evaluations were performed in animals 15 in the 0, 40, 80, and 170 mg/kg-day dose groups. The sperm evaluations consisted of spermatid 16 heads per testis and per gram testis, spermatid counts, and epididymal spermatozoal motility and 17 concentration. The vaginal cytology evaluations consisted of measures of estrous cycle length. 18 Sperm motility was 17.1, 14.9, and 24.0% lower than in vehicle controls at 40, 80, and 19 170 mg/kg-day, respectively. Other statistically significant effects in the males included 20 reductions in absolute epididymis weight at >80 mg/kg-day and absolute left cauda epididymis 21 weight at 170 mg/kg-day, and statistically significant increases in the incidences (90-100%) of 22 minimal to moderate atrophy of the preputial and prostate gland, seminal vesicle, and testicular 23 germinal epithelium at 320 mg/kg-day. Effects in the females included statistically significant 24 increases in incidences of minimal to mild uterine atrophy (70-90%) at >170 mg/kg-day and 25 clitoral gland atrophy (70%) and ovarian interstitial cell cytoplasmic alterations (100%) at 26 320 mg/kg-day. The vaginal cytology evaluations indicated that the females in the 170 mg/kg- 27 day group spent more time in diestrus and less time in proestrus, estrus, and metestrus than did 28 the vehicle controls. Body weight loss and reduced body weight gain at the lower dose levels 29 may have contributed to the atrophy and other effects observed in both genders (NTP, 2004). 30 NTP (2004) also tested groups of 10 male and 10 female B6C3Fi mice that were 31 similarly exposed to 1,1,2,2-tetrachloroethane for 14 weeks at reported average daily dietary 32 doses of 0, 100, 200, 370, 700, or 1,360 mg/kg-day (males) or 0, 80, 160, 300, 600, or 33 1,400 mg/kg-day (females). The main part of this study is summarized in Section 4.2.1.1. 34 Reproductive function (fertility) was not evaluated, and toxicity endpoints in reproductive organs 35 are the same as those evaluated in the rat part of the study summarized above. The sperm and 36 vaginal cytology evaluations were performed in the 0, 1,120, 4,550, or 9,100 mg/kg-day dose 37 groups. 37 DRAFT - DO NOT CITE OR QUOTE ------- 1 Effects observed in the male mice included statistically significant increases in the 2 incidence of preputial gland atrophy at 100, 700, and 1,360 mg/kg-day (incidences in the control 3 to high dose groups were 0/10, 4/10, 2/10, 0/10, 4/10, and 5/10, respectively), decreased absolute 4 testis weight at >700 mg/kg-day and absolute epididymis and cauda epididymis weights at 5 1,360 mg/kg-day, and decreased epididymal spermatozoal motility at 1,360 mg/kg-day (3.1% 6 less than vehicle controls). In female mice, the length of the estrous cycle was significantly 7 increased at 9,100 pm (1,400 mg/kg-day) (8.7% longer than vehicle controls). The pronounced 8 decreases in body weight gain or body weight loss were similar to those observed in rats. 9 10 4.3.2. Inhalation Exposure 11 Male rats were exposed to 0 or 15 mg/m3 (2.2 ppm) 1,1,2,2-tetrachloroethane 4 hours/day 12 for up to 8 days in a 10-day period (Gohlke and Schmidt, 1972; Schmidt et al, 1972). 13 Reproductive function was not tested, but evaluations included histological examinations of the 14 testes in groups of seven control and seven treated males following the second, fourth, and eighth 15 exposures, as detailed in Schmidt et al. (1972) in Section 4.2.2.2. This study is limited by 16 imprecise and incomplete reporting of results. It was noted that testicular histopathology, 17 described as atrophy of the seminal tubules with strongly restricted or absent spermatogenesis, 18 was observed in five exposed animals following the fourth exposure; data for the other time 19 periods and the control group were not reported. 20 The Schmidt et al. (1972) chronic inhalation study, summarized in Section 4.2.2.2, 21 included a limited reproductive function/developmental toxicity assessment. Male rats were 22 exposed to 0 or 13.3 mg/m3 (1.9 ppm) 1,1,2,2-tetrachloroethane 4 hours/day for 265 days, as well 23 as during the mating period. One week before the end of the exposure period, seven control and 24 seven exposed males were each mated with five unexposed virgin females. Dams were 25 permitted to deliver and the offspring were observed for 84 days and were examined 26 macroscopically for malformations. The percentage of mated females having offspring, littering 27 interval, time to 50% littered, total number of pups, pups/litter, average birth weight, postnatal 28 survival on days 1, 2, 7, 14, 21, and 84, sex ratio, and average body weight on postnatal day 84 29 were also measured. No macroscopic malformations or significant group differences in the other 30 indices were found, indicating that 13.3 mg/m3 was a NOAEL for male reproductive toxicity. 31 No effects attributable to 1,1,2,2-tetrachloroethane were reported in rats exposed to 5 or 32 50 ppm (34.3 or 343 mg/m3, respectively) 7 hours/day for 5 days in a dominant lethal test 33 (McGregor, 1980). A viral infection may have resulted in increased numbers of early deaths in 34 all groups, including the control group, possibly affecting study sensitivity. The frequency of 35 sperm with hook abnormalities was statistically significantly increased in the 343 mg/m3 group, 36 but not at 34.3 mg/m3. 37 38 4.4. OTHER DURATION-OR ENDPOINT-SPECIFIC STUDIES 3 8 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 4.4.1. Acute Studies (Oral and Inhalation) 4.4.1.1. Oral Studies Oral (single-dose gavage) median lethal dose (LDso) values of 250-800 mg/kg have been reported in rats (NTP, 2004; Schmidt et al., 1980b; Gohlke et al, 1977; Smyth et al, 1969). Cottalasso et al. (1998) described a series of experiments evaluating the effect of a single gavage dose of 1,1,2,2-tetrachloroethane on the liver of exposed rats. In the first experiment, male Sprague-Dawley rats (5/group) were given a single gavage dose of 0, 143.5, 287, 574, or 1,148 mg/kg in mineral oil and five animals from each group were sacrificed 5, 15, 30, or 60 minutes later. Sixty minutes after treatment, statistically significant, dose-related increases in serum activity levels of AST (66, 129, and 201%, respectively) and ALT (54, 88, and 146%, respectively) were observed at >287 mg/kg. The increase in rat serum activities of AST and ALT were also increased in a time-dependent manner. Serum AST increased 13-130% from 5 to 60 minutes in rats at 574 mg/kg-day and serum ALT increased 8-88% from 5 to 60 minutes. A statistically significant decrease in hepatic microsomal G6Pase activity (19, 36, and 47%, respectively) was observed at >287 mg/kg. A statistically significant decrease in levels of dolichol, a polyisoprenoid compound believed to be important in protein glycosylation reactions, in the liver (41 and 56%, respectively) and a statistically significant increase in triglyceride levels in liver homogenate (60 and 83%, respectively) were observed at >574 mg/kg. A statistically significant increase in the trigylceride levels in liver microsomes (46, 65, and 97%, respectively) was observed at >287 mg/kg. See Table 4-16 for a summary of these acute liver toxicity results. A time-dependent effect was observed in the decrease in G6Pase, in the increase in triglyceride levels, and in the decrease in levels of dolichol in the liver at 574 mg/kg-day from 5 to 60 minutes. Table 4-16. Liver function and other effects observed following acute (60 minutes) exposure to 1,1^2,2-tetrachloroethane Dose (mg/kg) 0 143.5 287 574 1,148 Serum AST (IU/L) 62 ±9 80 ±10 103±21a 143±13a 187±24a Serum ALT (IU/L) 26 ±4 32 ±6 40±7a 49±6a 64±9a Microsomal G6Pase (nmol/min/mg protein) 361 ±29 342 ±43 291±39a 230±18a 191±31a Homogenate triglycerides (mg/g liver) 14. 5 ±2.0 15.9±2.3 19.7 ±3.2 23.2±2.8a 26.5±3.4a Microsomal triglycerides (mg/g liver) 1.61 ±0.12 1.95 ±0.21 2.35±0.30a 2.65±0.35a 3.17±0.42a Homogenate total dolichol levels (ng/mg protein) 335 ±0.28 302 ±53 268 ± 45 197±25a 147±21a 25 26 27 Significantly different from control. Source: Cottalasso et al. (1998). Schmidt et al. (1980b) administered 0 or 100 mg/kg doses of 1,1,2,2-tetrachloroethane in corn oil by gavage to groups of 10 male Wistar rats, followed immediately by increased 39 DRAFT - DO NOT CITE OR QUOTE ------- 1 environmental temperatures, and evaluated hepatic effects 20-22 hours post administration. 2 Statistically significant increases in serum leucine aminopeptidase activity, hepatic ascorbic acid, 3 and hepatic triglyceride levels (10.5, 22.3, and 125% greater than control levels, respectively) 4 were observed, but changes in body weight, liver weight, hepatic N-demethylation of 5 aminopyrine, and serum ALT activity were not observed. The report includes a general 6 statement that all chemicals tested in this study led to necrosis and fatty degeneration, which 7 suggests that 100 mg/kg was a hepatotoxic dose of 1,1,2,2-tetrachloroethane. However, the 8 significance of the histology results cannot be assessed due to a lack of incidence and severity 9 measures. No other 1,1,2,2-tetrachloroethane-related histological data were reported in this 10 study. 11 Wolff (1978) exposed 8- to 10-week-old, female Wistar rats in groups of 8-10 animals, 12 to a single gavage dose of 0, 25, or 50 mg/kg of 1,1,2,2-tetrachloroethane 30 minutes prior to 13 testing for passive avoidance (shock level of 0.4 milliamperes [mA]). Passive avoidance was 14 measured by allowing the test rats to explore the test apparatus, which consisted of a larger, lit 15 box and a smaller, dark box. After 180 seconds, the darkened box received an electrical shock 16 through the grid floor. During the 180 seconds, the rats remained in the darkened box 17 approximately 80% of the time. The test was repeated 24 hours later. No differences in 18 avoidance were observed between the control and 25 mg/kg groups, but decreased passive 19 avoidance behavior was reported following exposure to 50 mg/kg. In the second test series, the 20 shock level was increased to 0.8 mA and the 1,1,2,2-tetrachloroethane dose was increased to 21 50 mg/kg. The 1,1,2,2-tetrachloroethane doses were then increased to 80 mg/kg and then to 22 100 mg/kg. Increasing the shock level to 0.8 mA resulted in no significant differences in 23 avoidance between the controls and the 50 mg/kg-day dose group (n = 10). Passive avoidance 24 was altered at 80 mg/kg (n = 10), and at 100 mg/kg, the animals (n = 10) were ataxic and did not 25 learn to avoid the shock. The authors stated that the treatment with 1,1,2,2-tetrachloroethane 26 may have affected the threshold of perception of the shock, rather than memory (Wolff, 1978). 27 This conclusion would be consistent with the high-dose anesthetic effects characteristic of 28 volatile organic compounds in general. 29 30 4.4.1.2. Inhalation Studies 31 Schmidt et al. (1980a) established a 24-hour median lethal concentration (LCso) of 32 8,600 mg/m3 (1,256 ppm) for 1,1,2,2-tetrachloroethane in rats for a single 4-hour exposure. 33 Carpenter et al. (1949) found that a 4-hour exposure to 1,000 ppm 1,1,2,2-tetrachloroethane 34 (6,870 mg/m ) was lethal in Sherman rats, with mortality in "2/6, 3/6, or 4/6" animals. 35 Price et al. (1978) exposed rats and guinea pigs to 576, 5,050, and 6,310 ppm 36 1,1,2,2-tetrachloroethane for 30 minutes. Rats exposed to 576 ppm (3,950 mg/m3) for 37 30 minutes showed a slight reduction in activity and alertness, while increasing the concentration 38 to 5,050 or 6,310 ppm (34,700 or 43,350 mg/m3) caused lacrimation, ataxia, narcosis, labored 40 DRAFT - DO NOT CITE OR QUOTE ------- 1 respiration, and 30-50% mortality (Price et al., 1978). Eye closure, squinting, lacrimation, and 2 decreased activity were observed in guinea pigs exposed to 576 ppm for 30 minutes; exposure to 3 5,050 ppm resulted in tremors, narcosis, and labored breathing, and exposure to 6,310 ppm 4 produced 30% mortality (Price et al., 1978). Organ weight measurements and gross pathology 5 and histology evaluations performed 14 days following the 30-minute exposures did not result in 6 chemical-related effects in the lungs, liver, kidneys, heart, brain, adrenals, testes, epididymides, 7 ovaries, or uterus in either species. 8 Pantelitsch (1933) exposed groups of three mice to 1,1,2,2-tetrachloroethane concent- 9 rations of 7,000, 8,000-10,000, 17,000, 29,000, or 34,000 mg/m3 (1,022, 1,168-1,460, 3,060, 10 5,220, or 6,120 ppm, respectively) for approximately 1.5-2 hours and examined changes in 11 clinical status of the animals. All concentrations resulted in disturbed equilibrium, prostration, 12 and loss of reflexes, with deaths occurring at >8,000-10,000 mg/m3; increasing the concentration 13 resulted in a more rapid onset of symptoms. 14 Horvath and Frantik (1973) determined that effective concentrations of 1,1,2,2-tetra- 15 chloroethane following a single 6-hour exposure in rats were 360 ppm (2,470 mg/m3) for a 50% 16 decrease in spontaneous motor activity and 200 ppm (1,370 mg/m3) for a 50% increase in 17 pentobarbital sleep time. No additional relevant information was reported. 18 Schmidt et al. (1980a) exposed groups of 10 male Wistar rats to 0, 410, 700, 1,030, 2,100, 19 or 4,200 mg/m3 (0, 60, 102, 150, 307, or 613 ppm, respectively) 1,1,2,2-tetrachloroethane (mean 20 concentrations) for 4 hours and evaluated the animals immediately (within 15-100 minutes), at 21 24 hours, or at 120 hours following exposure. The purpose of this study was to determine a 22 threshold concentration for effects on the liver following inhalation exposure. Evaluation of this 23 study is complicated by imprecise and incomplete reporting of results, exposure levels, and 24 observation durations. For example, results for endpoints other than liver histology, ascorbic 25 acid content, and histochemistry were not reported for the lowest concentration (410 mg/m3), and 26 liver ascorbic acid content and serum and liver triglyceride levels were the only results reported 27 quantitatively. Histological effects included diffuse fine droplet fatty degeneration in the liver at 28 410 and 700 mg/m3 (24 hours postexposure), nonspecific inflammation and Councilman bodies 29 (eosinophilic globules derived from necrosis of single hepatocytes) in the liver at 4,200 mg/m3 30 (24 hours postexposure), and interstitial nephritis in the kidneys at 700 mg/m3 (120 hours 31 postexposure). Additional information on these findings, including incidences and results for 32 other exposure concentrations, was not reported. 33 Hepatic ascorbic acid levels were statistically significantly increased in groups exposed 34 to >700 mg/m immediately after exposure (2, 64, 29, 167, and 182% higher than controls at 410, 35 700, 1,030, 2,100, and 4,200 mg/m3, respectively), but returned to control levels within 24 hours. 36 Serum triglyceride concentrations were statistically significantly decreased at >700 mg/m3 after 37 24 hours (35, 23, 29, and 56% at 700, 1,030, 2,100, and 4,200 mg/m3, respectively) and at 38 2,100 and 4,200 mg/m3 (39 and 42%, respectively) after 120 hours. Hepatic triglyceride levels 41 DRAFT - DO NOT CITE OR QUOTE ------- 1 were significantly increased at 2,100 and 4,200 mg/m3 (92 and 76%, respectively) at 24 hours 2 postexposure. Hexobarbital sleep time was increased at 2,100 and 4,200 mg/m3 (not quantified). 3 Assessing the biological significance and adversity of the effects in this study is complicated by 4 factors that include the lack of liver lesion incidence data, the paucity of other quantitative data, 5 and other reporting insufficiencies. The authors concluded that the threshold for effects on the 6 liver was between 410 and 700 mg/m3 because the fine droplet fatty degeneration was not 7 considered to be biologically significant in the absence of accompanying serum and liver 8 biochemical changes. 9 Hepatic effects were also reported by Tomokuni (1969), who administered a single 10 3-hour exposure of 600 ppm (4,120 mg/m3) 1,1,2,2-tetrachloroethane to female Cb mice. Total 11 hepatic lipids and triglycerides were statistically significantly increased following exposure and 12 continued to increase for 8 hours postexposure. Hepatic triglyceride levels increased more than 13 total lipid levels for 8 hours postexposure. Total hepatic adenosine triphosphate (ATP) levels 14 were decreased immediately following exposure and continued to decrease over the next 8 hours. 15 A later study by the same investigator (Tomokuni, 1970) evaluated female Cb mice (5-8/group) 16 exposed to 800 ppm (5,490 mg/m3) 1,1,2,2-tetrachloroethane for 3 hours and then followed the 17 time-course of the changes in hepatic lipids and phospholipids over the next 90 hours. Increased 18 tricglyceride and decreased phospholipid levels were seen for the first 30-45 hours postexposure, 19 but the effects generally resolved by 90 hours postexposure, demonstrating that hepatic effects 20 resolved after exposure was terminated. 21 Horiuchi et al. (1962) exposed 10 male mice for a single 3-hour period to an atmosphere 22 containing 5,900 ppm (-40,500 mg/m3) or 6,600 ppm (-45,300 mg/m3) 1,1,2,2-tetrachloroethane 23 and then observed the animals for 1 week following exposure. Tissues were obtained for 24 histologic evaluation from animals at sacrifice or when discovered dead. Three mice exposed to 25 5,900 ppm and four mice exposed to 6,600 ppm died prior to the end of the study. The 26 histological results reported by Horiuchi et al. (1962) are similar to the repeated vapor exposure 27 study in mice, described in Section 4.4.2.2, with slight to moderatie congestion and fatty 28 degeneration of the liver and congestion of the other mail tissues. 29 Deguchi (1972) administered a single 6-hour exposure of 0, 10, 100, or 1,000 ppm (0, 69, 30 690, or 6,900 mg/m3, respectively) of 1,1,2,2-tetrachloroethane to male rats and evaluated serum 31 AST activity and ALT activity levels up to 72 hours postexposure. This study was reported in 32 Japanese and included an English translation of the abstract. Based on information in the 33 English abstract and data graphs in this Japanese study, there was a minimal increase in serum 34 AST at all exposure concentrations 72 hours postexposure. 35 36 4.4.2. Short-term Studies (Oral and Inhalation) 37 4.4.2.1. Oral Studies 42 DRAFT - DO NOT CITE OR QUOTE ------- 1 Dow Chemical Company (1988) exposed groups of male Osborne-Mendel rats (n = 5) to 2 daily gavage doses of 0, 25, 75, 150, or 300 mg/kg-day 1,1,2,2-tetrachloroethane every 24 hours 3 for 4 days, followed by an injection of [3H]-thymidine, for DNA incorporation studies, 24 hours 4 following the last 1,1,2,2-tetrachloroethane dose. The fourth dose was not administered to the 5 300 mg/kg-day group due to signs of central nervous system (CNS) depression and debilitation, 6 and one animal in this group died before [ H]-thymidine injection. Terminal body weights of the 7 300 mg/kg-day animals were statistically significantly decreased 17% compared to controls. 8 Absolute liver weights at the highest dose were decreased and relative liver weights were 9 statistically significantly increased 14% in the 150 mg/kg-day dose group. 10 Histological examinations of the livers showed increased numbers of hepatocytes in 11 mitosis in the 75, 150, and 300 mg/kg-day groups, although this response was variable in high- 12 dose rats due, possibly, to the increased toxicity observed in this group (Dow Chemical 13 Company, 1988). Increased numbers of reticuloendothelial cells were seen at 300 mg/kg-day. 14 Increased hepatic glycogen content was found in hepatocytes of 75 and 150 mg/kg-day animals, 15 although this could be an outcome of altered feeding patterns resulting from sedative effects of 16 dosing (Dow Chemical Company, 1988). 17 Hepatic DNA synthesis ([3H]-thymidine incorporation) was increased 2.8-, 4.8-, and 18 2.5-fold at 75, 150, and 300 mg/kg-day, respectively; the decline at 300 mg/kg-day may have 19 been due to the poor clinical status of the rats in this group (Dow Chemical Company, 1988). 20 Total hepatic DNA content was not increased. Other endpoints were not evaluated. The 300 21 mg/kg-day dose is a frank effect level (FEL) based on the CNS depression and mortality. The 75 22 mg/kg dose may represent a NOAEL for increased relative liver weight in rats. However, the 23 increase in DNA synthesis and mitosis are not necessarily indicative of hepatotoxicity, and the 24 histological examinations showed no accompanying degeneration or other adverse liver lesions. 25 Dow Chemical Company (1988) similarly exposed groups of male B6C3Fi mice (n = 5) 26 to daily gavage doses of 0, 25, 75, 150, or 300 mg/kg-day 1,1,2,2-tetrachloroethane for 4 days, 27 followed by [3H]-thymidine injection for the DNA incorporation studies. All animals survived 28 treatment, and changes in body weight were not observed at any dose level. Absolute and 29 relative liver weights were increased 13 and 11%, respectively, at 150 mg/kg-day and 19 and 30 72%, respectively, at 300 mg/kg-day, although only the increase in relative liver weight at 300 31 mg/kg-day was statistically significantly. 32 Histopathologic examination of the liver revealed centrilobular swelling, with a 33 corresponding decrease in hepatocyte size in the periportal region due to decreased glycogen 34 content, in mice at >75 mg/kg-day. Increased hepatocyte mitosis was also observed in mice at 35 300 mg/kg-day. Hepatic DNA synthesis was increased 1.7-fold at 150 mg/kg-day and 4.4-fold at 36 300 mg/kg-day, although total hepatic DNA content was not increased. Other endpoints were 37 not evaluated. 43 DRAFT - DO NOT CITE OR QUOTE ------- 1 TSI Mason Laboratories (1993a, unpublished) administered 1,1,2,2-tetrachloroethane in 2 corn oil to groups of male and female (n = 5) F344/N rats at 0, 135, 270, or 540 mg/kg for 3 12 days over a 16-day period. Rats were weighed prior to dosing, after 7 days, and prior to 4 euthanasia, and all surviving rats were euthanized and subject to necropsy. Study endpoints 5 included clinical observations, body weight, necropsy, selected organ weights (liver, kidneys, 6 thymus, lung, heart, and testes), and histology of gross lesions. All of the high-dose rats died by 7 day 5 of the study. Male rats exposed to 270 mg/kg displayed an increase in body weight from 8 day 1 through day 17 of 37%, compared to an increase of 64% in controls. Female rats exposed 9 to 270 mg/kg displayed a decrease in body weight from day 1 through day 17 of 3%, compared 10 with an increase of 30% in controls. The automatic watering system for the low- and high-dose 11 males failed prior to the administration of 1,1,2,2-tetrachloroethane, and the low and high doses 12 of the study were repeated in a subsequent study by TSI Mason Laboratories (1993b, 13 unpublished). 14 Clinical signs were absent in the 135 mg/kg animals, but animals exposed to 270 or 15 540 mg/kg were lethargic following treatment. Absolute liver weights were statistically 16 significantly increased (19%) in the 135 mg/kg-day female rats, while relative liver weights were 17 statistically significantly increased at both 135 and 270 mg/kg-day (16 and 34%, respectively). 18 No changes in absolute or relative liver weights were seen in exposed male rats. Absolute right 19 kidney weight was significantly increased 9 and 37% in females at 135 and 270 mg/kg-day, 20 respectively. Absolute thymus weight was statistically significantly decreased in the mid-dose 21 group of male rats (33% at 270 mg/kg-day) while absolute (45%) and relative (32%) thymus 22 weights were statistically significantly decreased in only the mid-dose females. Relative right 23 testis weight was statistically significantly increased (10% at 270 mg/kg-day) in male rats. 24 Absolute, but not relative, lung weights were statistically significantly decreased in 270 mg/kg- 25 day females (17%), while relative heart weights were statistically significantly increased (14%) 26 in females. 27 Gross and microscopic lesions were observed in the liver (i.e., hepatodiaphragmatic 28 nodules) of one control, one mid-dose, and one high-dose rat, but these were common 29 spontaneous lesions. 30 In another study, TSI Mason Laboratories (1993b, unpublished) exposed groups of male 31 F344/N rats (n = 5) to 0, 135, 270, or 540 mg/kg-day 1,1,2,2-tetrachloroethane by gavage in corn 32 oil on 12 days in a 16-day period. Study endpoints included clinical observations, body weight, 33 necropsy, selected organ weights (liver, kidneys, thymus, lung, heart, and testes), and histology 34 of gross lesions. All animals exposed to 540 mg/kg-day died by day 3 of the study. Rats in the 35 270 and 540 mg/kg-day groups were extremely lethargic following administration of the test 36 article, with recovery observed only in the 270 mg/kg-day rats. 37 The weight gain observed in the low- and mid-dose rats was 55.2 and 28%, respectively. 38 At 135 mg/kg, statistically significant increases of 17 and 13% in absolute and relative liver 44 DRAFT - DO NOT CITE OR QUOTE ------- 1 weights, respectively, were observed compared to controls. In the mid-dose group, statistically 2 significant decreases in absolute testes weight (7%), absolute kidney weight (9%), absolute and 3 relative heart weight (10 and 6%, respectively), and absolute and relative thymus weight (33 and 4 21%, respectively) were observed. Statistically significant increases in relative thymus (10%), 5 liver (16%), and kidney weights (7%) were observed at 270 mg/kg compared to controls. 6 Gross and microscopic lesions were observed in the liver of one 270 mg/kg-day male and 7 in the glandular stomach of one 540 mg/kg-day male, but these were diagnosed as spontaneous 8 lesions commonly observed in F344/N rats. The lesion observed in the liver was a dark nodule 9 on the median lobe and corresponded histomorphologically to a hepatodiaphragmatic nodule, 10 and the lesion observed in the glandular stomach was a pale foci. 11 TSI Mason Laboratories (1993c, unpublished) exposed groups of five male and five 12 female B6C3Fi mice to 0, 337.5, 675, or 1,350 mg/kg-day 1,1,2,2-tetrachloroethane by gavage in 13 corn oil on 12 days during a 16-day period. Study endpoints included clinical observations, body 14 weight, necropsy, selected organ weights (liver, kidneys, thymus, lung, heart, and testes), and 15 histology of gross lesions. All mice of both genders in the 1,350 mg/kg-day groups were found 16 dead or euthanized by day 3 of the study. Additionally, one 675 mg/kg-day female died and one 17 337.5 mg/kg-day female was euthanized prior to the end of the study. 18 No significant changes in body weight were reported in treated groups. Animals in the 19 675 and 1,350 mg/kg-day groups appeared lethargic within 15 minutes of dosing, and the 20 1,350 mg/kg-day mice failed to recover after the third treatment. Lethargy also occurred in the 21 337.5 mg/kg-day female that was sacrificed, but not in other animals in that exposure group. In 22 male mice, relative liver weight was statistically significantly increased 9% at 337.5 mg/kg, and 23 absolute and relative liver weights were statistically significantly increased 28 and 37%, 24 respectively, at 675 mg/kg-day. In female mice, absolute and relative liver weights were 25 statistically significantly increased by 50 and 42%, respectively, at 675 mg/kg. 26 Gross hepatic changes, described as pale livers, were noted in one male and three females 27 at 337.5 mg/kg-day and in four males and three females at 675 mg/kg-day. Histological 28 examination of the gross lesions showed that they correlated with centrilobular hepatocellular 29 degeneration characterized by hepatocellular swelling, cytoplasmic rarefaction, and 30 hepatocellular necrosis in the 675 and 1,350 mg/kg-day males and the 337.5, 675, and 31 1,350 mg/kg-day females. Hepatocellular necrosis was the most common lesion observed at 32 675 mg/kg-day. 33 In a study examining the potential renal toxicity of orally administered halogenated 34 ethanes, groups of five male F344/N rats received 0, 0.62, or 1.24 mmol/kg-day 1,1,2,2-tetra- 35 chloroethane by gavage in corn oil (0, 104, or 208 mg/kg-day, respectively) for 21 consecutive 36 days (NTP, 1996). All rats in the high-dose group died or were killed moribund on days 13-14 37 and were not evaluated further. Evaluations of the 0 and 104 mg/kg-day animals included 38 weekly body weights, end-of-study urinalysis (volume, specific gravity, creatinine, glucose, total 45 DRAFT - DO NOT CITE OR QUOTE ------- 1 protein, AST, y-glutamyl transpeptidase, and N-acetyl-p-D-glucosaminidase), gross necropsy, 2 selected organ weights (right kidney, liver, and right testis), selected histopathology (right kidney, 3 left liver lobe, and gross lesions), and kidney cell proliferation analysis (proliferating cell nuclear 4 antigen [PCNA] labeling index for proximal and distal tubule epithelial cells in S phase). 5 Clinical signs in the high-dose animals included thinness and lethargy (5/5 rats), diarrhea, 6 abnormal breathing, and ruffled fur (3/5 rats). In the low-dose group, no effects on survival, 7 body weight gain, urinalysis parameters, absolute or relative kidney weights, renal or testicular 8 histopathology, or kidney cell PCNA labeling index were observed. 9 Hepatic effects in the low-dose group included increased absolute and relative liver 10 weights (24 and 29% greater than controls, respectively) and cytoplasmic vacuolization of 11 hepatocytes. The vacuolation occurred in hepatocytes of all low-dose rats and consisted of 12 multifocal areas with clear droplets within the cytoplasm. Changes in the kidneys of the male 13 rats were not observed. 14 In a range-finding study, the NTP (NTP, 2004; TSI Mason Laboratories, 1993d) exposed 15 male and female F344/N rats (5/sex/group) to 0, 3,325, 6,650, 13,300, 26,600, or 53,200 ppm 16 1,1,2,2-tetrachloroethane in the diet (microcapsules) for 15 days. Unexposed and vehicle control 17 groups were also evaluated, with the latter being given feed with empty microcapsules. Study 18 endpoints included clinical observations, body weight, food consumption, necropsy, selected 19 organ weights (liver, kidneys, thymus, lung, heart, and testes), and histology of gross lesions; 20 histology was not evaluated in animals without gross lesions. The study authors reported that 21 average daily doses for the three lowest concentrations were 300, 400, or 500 mg/kg-day for both 22 genders. All rats exposed to 26,600 or 53,200 ppm were killed moribund on day 11. The 23 average daily doses for these groups were not reported. 24 Female rats exposed to 400 mg/kg-day and both genders exposed to 500 mg/kg-day were 25 thin and displayed ruffled fur. Body weight at study termination was statistically significantly 26 lower than controls in both genders of all treated groups. Male rats exposed to 300 mg/kg-day 27 showed decreased weight gain compared to controls and those exposed to higher doses lost 28 weight, with final body weights in male rats 28, 46, and 53% less than vehicle controls at 300, 29 400, and 500 mg/kg-day, respectively. Females lost weight at doses of >300 mg/kg-day, with 30 final body weights in female rats 25, 38, and 47% less than vehicle controls at 300, 400, and 31 500 mg/kg-day, respectively. Decreased feed consumption likely contributed to the decreased 32 weight gains because consumption was reduced in a dose-related manner in both genders of all 33 treated groups (NTP, 1996). 34 Absolute thymus weights were decreased 24, 69, and 84% in male rats and 37, 61, and 35 81% in female rats at doses of >300 mg/kg-day and relative thymus weights were decreased 36 42 and 65% in male rats and 38 and 65% in female rats at >400 mg/kg-day (NTP, 2004; TSI 37 Mason Laboratories, 1993d). In male rats, absolute liver weights were decreased 22, 49, and 38 60% compared to controls at 300, 400, and 500 mg/kg-day, respectively. Relative liver weight 46 DRAFT - DO NOT CITE OR QUOTE ------- 1 was increased 7% compared to controls at 300 mg/kg-day and decreased 14% compared to 2 controls at 500 mg/kg-day. In female rats, absolute liver weight was decreased 25 and 34% 3 compared to controls at 400 and 500 mg/kg-day, respectively, and relative liver weight was 4 increased 34 and 23% compared to controls at 300 and 500 mg/kg-day, respectively. Relative 5 kidney weights were increased 14, 26, and 18% in male rats at 300, 400, and 500 mg/kg-day, 6 respectively, and 17 and 36% in female rats at 400 and 500 mg/kg-day, respectively. Absolute 7 kidney weights were decreased 17, 32, and 45% in males and 16, 27, and 27% in females at 300, 8 400, and 500 mg/kg-day, respectively. Other organ weight decreases were considered a 9 reflection of the decreased body weights. 10 Focal areas of alopecia occurred on the skin of four female rats in the 500 mg/kg-day 11 group, and these lesions correlated with minimal to moderate acanthosis, which is an abnormal 12 benign increase in the thickness of the stratum spinosum, a layer of cells that is capable of 13 undergoing mitotic cell division, of the epidermis. In the liver, mild or moderate centrilobular 14 degeneration was observed microscopically in the exposed male and female rats. 15 Groups of five male and five female B6C3Fi mice were exposed to 0, 3,325, 6,650, 16 13,300, 26,600, or 53,200 ppm of encapsulated 1,1,2,2-tetrachloroethane in the diet for 15 days 17 (NTP, 2004; TSI Mason Laboratories, 1993d). Organ weights, gross necropsy, and histology of 18 gross lesions were evaluated in surviving mice at the termination of the study. Average daily 19 doses were not determined by the study authors because feed consumption could not be 20 measured accurately due to excessive scattering of feed. All male and female mice exposed to 21 53,200 ppm, all males exposed to 26,600 ppm, and two males exposed to 13,300 ppm were 22 sacrificed in extremis before the end of the study. Final body weights were decreased 16, 24, 23 and 22%, in comparison to vehicle controls, in males at 3,325, 6,650, and 13,300 ppm, 24 respectively. In females, final body weights were decreased 9, 20, 31, and 34% at 3,325, 6,650, 25 13,300, and 26,600 ppm, respectively. 26 Clinical findings included hyperactivity in males and females exposed to 3,325, 6,650, or 27 13,300 ppm and in females in the 26,600 ppm group. Males in the 26,600 and 53,200 ppm 28 groups were lethargic. Males exposed to >6,650 ppm and females exposed to 26,600 and 29 53,200 ppm were thin and had ruffled fur. A statistically significant decrease in absolute (31, 47, 30 82, and 81%, respectively) and relative (22, 33, 74, and 72%, respectively) thymus weights 31 compared to controls was observed in all exposed female mice. Relative liver weights were 32 statistically significantly increased 22, 31, and 34% in male mice at 3,325, 6,650, and 33 13,300 ppm, respectively. Absolute liver weights were statistically significantly decreased 11,9, 34 and 5% in female mice at 6,650, 13,300, and 26,600 ppm, respectively, and relative liver weight 35 increased 30 and 44% at 13,300 and 26,600 ppm, respectively. Other organ weight changes 36 were associated with changes in body weight. Pale or mottled livers were noted in all exposed 37 groups of male and female mice and correlated microscopically with hepatocellular degeneration, 38 which was characterized by hepatocellular swelling, cytoplasmic rarefaction, single paranuclear 47 DRAFT - DO NOT CITE OR QUOTE ------- 1 vacuoles, hepatocellular necrosis, and infrequent mononuclear infiltrates. The severity of the 2 hepatic changes increased with increasing exposure concentration. 3 The histological examinations in the surviving mice showed hepatocellular degeneration 4 in 3/3, 4/4, 4/4, 1/1, and 1/1 males, and 4/4, 4/4, 3/3, 3/3, and 3/3 females, at 3,325, 6,650, 5 13,300, 26,600, and 53,200 ppm, respectively (TSI Mason Laboratories, 1993d). For both 6 genders, the lesions tended to be minimal to mild at 3,325 and 6,650 ppm, with more moderate to 7 marked severity observed at the higher doses. 8 The National Cancer Institute (NCI, 1978) conducted a range-finding study in rats and 9 mice in order to estimate the maximum tolerated dose for administration in the chronic bioassay. 10 In this study, Osborne-Mendel rats (5/sex/group) received gavage doses of 0 (vehicle control 11 group), 56, 100, 178, 316, or 562 mg/kg 1,1,2,2-tetrachloroethane in corn oil 5 days/week for 12 6 weeks, followed by a 2-week observation period. B6C3Fi mice (5/sex/group) were similarly 13 exposed to 0, 32, 56, 100, 178, or 316 mg/kg 1,1,2,2-tetrachloroethane. It appears that mortality 14 and body weight gain were the only endpoints used to assess toxicity and determine the high- 15 dose levels for the NCI (1978) chronic bioassays in rats and mice. In the rats, one male exposed 16 to 100 mg/kg and all five females exposed to 316 mg/kg died (mortality rates in the 562 mg/kg 17 groups were not reported). Body weight gain was reduced 3, 9, and 38% in male rats and 9, 24, 18 and 41% in female rats at 56, 100, and 178 mg/kg-day, respectively. No deaths or significant 19 alterations in body weight gain were observed in the mice. In male rats, 100 and 178 mg/kg-day, 20 were selected as the NOAEL and LOAEL, respectively, for the observed decrease in body 21 weight, while in female rats the NOAEL and LOAEL were 56 and 100 mg/kg-day, respectively, 22 for the same endpoint. The highest dose in mice, 316 mg/kg-day, was selected as the NOAEL 23 for body weight changes and mortality. 24 25 4.4.2.2. Short-term Inhalation Studies 26 Rats (n = 84) were exposed to 0 or 15 mg/m3 (2.2 ppm) 1,1,2,2-tetrachloroethane 27 4 hours/day for up to 8 days in a 10-day period (Gohlke and Schmidt, 1972; Schmidt et al., 1972). 28 Following the first, third, and seventh exposures, seven control and exposed rats were given an 29 unknown amount of ethanol. Evaluations were performed on seven males from the control and 30 treated groups, with and without ethanol, following the second, fourth, and eighth exposures. 31 Statistically significant changes included increased serum total protein and decreased 32 serum ar and (X2-globulin fractions compared to controls after the eighth exposure (day 10), 33 although the difference was not quantified (Schmidt et al., 1972). Histological effects included a 34 fine to medium droplet fatty degeneration of the liver that involved increasing numbers of 35 animals with increasing duration of exposure, although the incidences and severity were not 36 reported (Gohlke and Schmidt, 1972). The results of the serum and histochemical evaluations 37 were illegible in the best copy of the translated reference available. Testicular atrophy in the 38 seminal tubules was observed in five treated animals following the fourth exposure (Gohlke and 48 DRAFT - DO NOT CITE OR QUOTE ------- 1 Schmidt, 1972). This study is limited by imprecise and incomplete reporting of results. 2 Assessment of the adversity of liver and other effects in this study is complicated by the 3 reporting insufficiencies, particularly the paucity of incidence and other quantitative data, as well 4 as effects that were not consistently observed in the three time periods and a lack of information 5 on dose-response due to the use of a single exposure level. 6 Horiuchi et al. (1962) exposed nine male mice to an average concentration of 7 approximately 7,000 ppm (48,000 mg/m3) 1,1,2,2-tetrachloroethane for 2 hours once/week for a 8 total of five exposures over 29 days. All animals died during the study with none of the deaths 9 occurring during exposure, and most (5/9) of the mice died within 5 days of the first exposure. 10 The only other reported findings in the exposed animals were slight to moderate congestion and 11 fatty degeneration of the liver and congestion of "other main tissues." 12 Horiuchi et al. (1962) exposed six male rats to an average concentration of 9,000 ppm 13 (62,000 mg/m ) 1,1,2,2-tetrachloroethane 2 hours/day, 2-3 times a week for 11 exposures in 14 29 days. All rats died during the study. No changes in body weight were reported. Exposed 15 animals generally showed hypermotility within the first few minutes of exposure, followed by 16 atactic gait within approximately 20 minutes and eventual near-complete loss of consciousness 17 1-1.5 hours after the onset of exposure. Hematology was assessed in three rats that survived 18 beyond 2 weeks, and two of these animals showed a decrease in RBC count and Hb content. 19 Exposed animals generally showed moderate congestion and fatty degeneration of the liver and 20 congestion of "other main tissues." 21 As discussed in Section 4.2.2.1, one monkey was exposed to varying concentrations 22 (2,000-4,000 ppm for the first 20 exposures, 1,000-2,000 ppm for the 20th-160th exposure, and 23 3,000^,000 ppm for the remaining exposures) of 1,1,2,2-tetrachloroethane for 2 hours/day, 24 6 days/week for 9 months (Horiuchi et al., 1962). Effects of short-term exposure included 25 weakness after seven exposures, diarrhea and anorexia between the 12th and 15th exposures, and 26 beginning at the 15th exposure, near-complete unconsciousness for 20-60 minutes after each 27 exposure. 28 29 4.4.3. Acute Injection Studies 30 Paolini et al. (1992) exposed groups of male and female Swiss Albino mice to a single i.p. 31 dose of 0, 300, or 600 mg/kg 1,1,2,2-tetrachloroethane and sacrificed the animals 24 hours after 32 dosing to assess hepatotoxicity. An LD50 of 1,476 mg/kg for 1,1,2,2-tetrachloroethane was 33 calculated using six animals/dose and eight dose groups. At 600 mg/kg, absolute and relative 34 liver weights were statistically significantly decreased 16 and 37%, respectively, in female mice. 35 No changes in total microsomal protein were noted. Statistically significant decreases (37-74%) 36 in hepatic cytochrome P450 enzymes of numerous classes were reported at both dose levels in 37 male and female mice (see Section 3.3). Other hepatic enzymes with statistically significantly 38 decreased activity included NADPH-cytochrome c-reductase, 5-aminolevulinic acid-synthetase, 49 DRAFT - DO NOT CITE OR QUOTE ------- 1 ethoxyresorufin-O-deethylase, pentoxyresorufin O-depentylase, GST (600 mg/kg only), and 2 epoxide hydrolase. Total hepatic heme was reduced at both doses, and heme oxygenase activity 3 was increased in a dose-related manner, but was statistically significant only in high-dose males 4 and females. 5 Wolff (1978) exposed groups of female Wistar rats to a single i.p. dose of 0, 20, or 6 50 mg/kg 30 minutes prior to testing for passive avoidance of a 0.4 mA electric shock. No 7 differences between the control and 25 mg/kg groups were reported, but doses of 50 mg/kg 8 resulted in decreased passive avoidance behavior. Similarly, no differences were seen in the 9 open-field test at any dose level. In male ICR-mice, a single i.p. dose of 20 mg/kg resulted in a 10 significant reduction in spontaneous locomotor activity, and 50-60 mg/kg resulted in a 50% 11 reduction (Wolff, 1978). 12 In an abstract, Andrews et al. (2002) described the exposure of a rat whole embryo 13 culture system to 1,1,2,2-tetrachloroethane. Gestational day 9 embryos were exposed to 14 concentrations between 0.5 and 2.9 mM 1,1,2,2-tetrachloroethane for 48 hours and then 15 evaluated for morphological changes. At concentrations >1.4 mM, 1,1,2,2-tetrachloroethane 16 resulted in rotational defects and anomalies of the heart and eye. Embryo lethality was observed 17 at>2.4mM. 18 19 4.4.4. Immunotoxicological Studies 20 Shmuter (1977) exposed groups of 12 Chinchilla rabbits to 0, 2, 10, or 100 mg/m3 (0, 0.3, 21 1.5, or 14.6 ppm, respectively) 1,1,2,2-tetrachloroethane 3 hours/day, 6 days/week for 8- 22 10 months. Animals were vaccinated with 1 mL of a 1.5 x 10 suspension of heated typhoid 23 vaccine 1.5, 4.5-5, and 7.5-8 months after the initiation of 1,1,2,2-tetrachloroethane exposure. 24 Significant increases and decreases in total antibody levels were observed in the 2 and 25 100 mg/m3 groups, respectively. No significant changes in 7S-typhoid antibody levels were 26 observed. Significant alterations in the levels of "normal" hemolysins to the Forsman's antigen 27 of sheep erythrocytes were observed in the 10 and 100 mg/m3 groups, as levels were increased in 28 the 10 mg/m group after 1.5, 2, and 2.5 months of exposure, decreased after 4 months, and 29 absent at 5 months of exposure. Levels of these hemolysins were decreased in the 100 mg/m 30 group during the first 6 months of exposure. Increases in the electrophoretic mobility of specific 31 antibodies following 1,1,2,2-tetrachloroethane were also reported. Exposure to 100 mg/m3 32 1,1,2,2-tetrachloroethane resulted in a decrease in the relative content of antibodies in the 33 y-globulin fraction and an increase in the T and (3 fractions. This is a poorly reported study that 34 provides inadequate quantitative data. The inconsistent dose-response patterns preclude 35 assessing biological significance and identification of a NOAEL or LOAEL. 36 37 4.5. MECHANISTIC DATA AND OTHER STUDIES IN SUPPORT OF THE MODE OF 38 ACTION 50 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 4.5.1. Genotoxicity As discussed in Section 3.4, radiolabeled 1,1,2,2-tetrachloroethane may covalently bind to DNA and RNA (Colacci et al, 1987), suggesting the potential for mutagenicity. A summary of the results of genotoxicity studies of 1,1,2,2-tetrachloroethane is presented in Table 4-17. Table 4-17. Results of in vitro and in vivo genotoxicity studies of l,l?2,2-tetrachloroethane In vitro gene mutation assays Test system Endpoint Cells/strain Concentrations Results -S9 +S9 Reference (a) Bacterial assays Salmonella typhimurium (Ames test) Escherichia coli Saccharomyces cerevisisae Aspergillus nidulans Reverse mutation Forward mutation DNA damage Gene conversion Gene reversion Gene recombina- tion Mitotic crossover TA100, 1535, 1537, 1538,98 TA1530, 1535, 1538 TA1535, 1537, 98 TA1535 TA97, 98, 100, 1535, 1537 TA98, 100, 1535, 1537 TA98, 100, 1535, 1537 TA100 BA13 pol A+/pol Af WP2S(X) D7 D7 D7 PI NA 10 uL/plate 10 uL/plate NA 10-3,333 uL/plate NA 5-l,OOOuL/plate NA 0.06-2,979 nmol/ plate 10 uL/plate 15-23 6 mM 3. 1-7.3 mM NA 3. 1-7.3 mM NA 3. 1-7.3 mM 0.01-0.04%v:v - NP - - - - - - - NP + NP NP NP NP NP NP - + - - - - - - - + - + - + - + + Nestmann et al., 1980 Rosenkranz, 1977; Bremetal, 1974 Mitomaetal, 1984 Onoetal, 1996 NTP, 2004 Milmanetal, 1988 Haworthetal., 1983 Warner et al., 1988 Roldan-Arjona et al., 1991 Rosenkranz, 1977; Bremetal., 1974 DeMarini and Brooks, 1992 Callenetal, 1980 Nestmann and Lee, 1 983 Callenetal., 1980 Nestmann and Lee, 1 983 Callenetal., 1980 Crebellietal, 1988 (b) Mammalian cell assays Mouse Lymphoma Hepatocytes (primary) Gene mutation DNA repair L5178Y Osborne Mendel rats B6C3F] mice 25-500 nL/mL NA NA - NP NP - - - NTP, 2004 Milmanetal., 1988; Williams, 1983 51 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-17. Results of in vitro and in vivo genotoxicity studies of 1,1^2,2-tetrachloroethane In vitro chromosomal damage assays Test system Cells/organs Concentrations Results Reference Mammalian Cells Chromosomal Aberrations Sister chromatid exchanges (SCE) UDS CHO cells CHO cells BALB/C-3T3 cells Human embryonic intestinal fibroblasts 453-804 ug/mL 16.8-558 ug/mL 500-1,000 ug/mL <15,869 ug/mL - + + - - + + NP NTP, 2004; Galloway et al, 1987 NTP, 2004; Galloway et al., 1987 Colaccietal, 1992 McGregor (1980) Other in vitro assays: Cell transformation (initiation) Cell transformation (promotion) BALB/C-3T3 cells 1-250 ug/mL 1-250 ug/mL 125-1,000 ug/mL NA 0. 1-1,000 ng/mL NP NP + - NP - - + - - Arthur Little, Inc., 1983 Tuetal, 1985 Colaccietal., 1990 Milmanetal, 1988 Colaccietal., 1996 In vivo bioassays Test system Cells/organs Doses Results Reference Chromosomal damage: mammalian Chromosomal aberrations Micronucleus UDS DNA alkylation Rat bone marrow cells, male Rat bone marrow cells, female Mouse peripheral blood erythrocytes Mouse hepatocytes Mouse hepatocytes, male Mouse hepatocytes, female Mouse hepatocytes 50 ppm 50 ppm 589-9, 100 ppm 200 mg/kg 50-1, 000 (mg/kg) 50-1,000 mg/kg 1 50 mg/kg - + + + - - + McGregor, 1980 NTP, 2004 Miyagawa et al., 1995 Mirsalis et al., 1989 Dow Chemical Co., 1988 Other in vivo assays S-phase DNA synthesis Mitotic recombination Recessive lethal mutation Mouse hepatocytes, male Mouse hepatocytes, female Drosophila melanogaster D. melanogaster 200-700 mg/kg 200-700 mg/kg 500-1, 000 ppm 800 ppm (injected) 1,500 (feed) - +/- - - Mirsalis et al., 1989 Vogel and Nivard, 1993 Woodruff etal, 1985 1 2 3 4 5 + = positive; - = negative/no change; CHO = Chinese hamster ovary; NA = not available; NP = assay not performed; UDS = unscheduled DNA synthesis 1,1,2,2-Tetrachloroethane has been shown to be predominantly inactive in reverse mutation assays in Salmonella typhimurium (strains TA97, TA98, TA100, TA1530, TA1535, TA1537, and TA1538), either with or without the addition of S9 metabolic activating mixture, even at concentrations that lead to cytotoxicity (NTP, 2004; Ono et al., 1996; Milman et al., 52 DRAFT - DO NOT CITE OR QUOTE ------- 1 1988; Warner et al., 1988; Mitoma et al, 1984; Haworth et al, 1983; Nestmann et al, 1980). 2 Two studies reported reverse mutation activity in S. typhimurium (Rosenkranz, 1977; Brem et al., 3 1974). Results of studies employing methods to prevent volatilization were not notably different 4 from those that did not use those methods. 1,1,2,2-Tetrachloroethane also did not induce 5 forward mutations (L-arabinose resistance) in S. typhimurium strain BA13 (Roldan-Arjona et al., 6 1991). Assays with Escherichia coli indicated that 1,1,2,2-tetrachloroethane induced DNA 7 damage, as shown by growth inhibition in DNA polymerase deficient E. coli (Rosenkranz, 1977; 8 Brem et al., 1974) and induction of prophage lambda (DeMarini and Brooks, 1992). In 9 Saccharomyces cerevisiae, 1,1,2,2-tetrachloroethane induced gene conversion, reversion, and 10 recombination in one study (Callen et al., 1980), whereas another study found no conversion or 11 reversion (Nestmann and Lee, 1983). In Aspergillus nidulans, 1,1,2,2-tetrachloroethane induced 12 aneuploidy, but no crossing over (Crebelli et al., 1988). 13 1,1,2,2-Tetrachloroethane did not induce trifluorothymidine resistance in L5178Y mouse 14 lymphoma cells, with or without S9, at concentrations up to those producing lethality (NTP, 15 2004). Primary hepatocytes from rats and mice exposed in vitro to 1,1,2,2-tetrachloroethane did 16 not show altered DNA repair at concentrations that were not cytotoxic (Milman et al., 1988; 17 Williams, 1983). McGregor (1980) reported no increase in unscheduled DNA synthesis (UDS) 18 in human embryonic intestinal fibroblasts exposed to 1,1,2,2-tetrachloroethane. Treatment of 19 Chinese hamster ovary (CHO) cells with up to 653 ug/mL (which was cytotoxic) did not result in 20 increased induction of chromosomal aberrations (NTP, 2004; Galloway et al., 1987) but did 21 produce an increased frequency of sister chromatid exchanges (SCEs) at concentrations of 22 >55.8 ug/mL (NTP, 2004; Galloway et al., 1987). SCEs were also induced in BALB/C-3T3 cells 23 treated in vitro with high concentrations (>500 ug/mL) of 1,1,2,2-tetrachloroethane, either with 24 or without S9 activating mixture (Colacci et al., 1992). 25 In BALB/C-3T3 cells, 1,1,2,2-tetrachloroethane exposure of up to 250 ug/mL in the 26 absence of exogenous metabolic activation did not result in increased numbers of transformed 27 cells (Colacci et al., 1992; Milman et al., 1988; Tu et al., 1985; Arthur Little, Inc., 1983); 28 survival was generally >70%. Higher concentrations (>500 ug/mL) were capable of 29 transforming the cells, but also showed higher levels of cytotoxicity (Colacci et al., 1990). 30 However, even relatively low levels (31.25 ug/mL) of 1,1,2,2-tetrachloroethane used as an 31 initiating agent, followed by promotion with 12-O-tetradecanoylphorbol-13 -acetate, resulted in 32 increased numbers of transformed cells (Colacci et al., 1992). 1,1,2,2-Tetrachloroethane did not 33 act as a promoter in BALB/C-3T3 cells in vitro without metabolic activation (Colacci et al., 34 1996). 35 1,1,2,2-Tetrachloroethane tested negative for sex-linked recessive lethal mutations and 36 mitotic recombination inD. melanogaster (NTP, 2004; Vogel and Nivard, 1993; Woodruff et al., 37 1985; McGregor, 1980). Replicative DNA synthesis was increased in hepatocytes isolated from 38 male B6C3Fi mice exposed to a single gavage dose of 200 mg/kg (24 and 48 hours 53 DRAFT - DO NOT CITE OR QUOTE ------- 1 postexposure) or 400 mg/kg (24, 39, and 48 hours postexposure) relative to hepatocytes from 2 unexposed mice (Miyagawa et al., 1995). Hepatocytes isolated from mice following a single 3 gavage dose of up to 1,000 mg/kg did not show an increase in UDS or S-phase DNA synthesis 4 (Mirsalis et al., 1989). Hepatocytes isolated from B6C3Fi mice 6 hours after a single gavage 5 dose of 150 mg/kg in corn oil demonstrated irreversible alkylation of hepatic DNA (Dow 6 Chemical Co., 1988). Inhalation exposure to 5 or 50 ppm (34.3 or 343 mg/m3) for 7 hours/day, 7 5 days/week did not result in increased frequency of chromosomal aberrations in bone marrow 8 cells isolated from male rats (McGregor, 1980); female rats exposed to 50 ppm (343 mg/m ), but 9 not to 5 ppm (34.3 mg/m3), showed an increase in bone marrow cell aberrations other than gaps 10 (McGregor, 1980). 11 In summary, genotoxicity studies provide limited evidence of a mutagenic mode of action. 12 1,1,2,2-Tetrachloroethane has some genotoxic activity, but in vitro genotoxicity tests generally 13 reported non-positive results. Similarly, in vivo studies had mostly non-positive results with the 14 exception of chromosomal aberrations in female rat bone marrow cells and micronucleus 15 formation in mouse bone marrow peripheral erythrocytes. The results of rat liver preneoplastic 16 foci and mouse BALB/C-3T3 cell neoplastic transformation assays suggest that 1,1,2,2-tetra- 17 chloroethane may have initiating and promoting activity. Overall, results of genotoxicity studies 18 for 1,1,2,2-tetrachloroethane are mixed and insufficient for establishing a mutagenic mode of 19 action. 20 21 4.5.2. Short-Term Tests of Carcinogenicity 22 Treatment of partially hepatectomized male Osborne-Mendel rats with a single 23 100 mg/kg gavage dose of 1,1,2,2-tetrachloroethane, followed by 7 weeks of promotion with 24 phenobarbital in the diet, did not result in increased numbers of preneoplastic (GGT-positive) 25 foci in the liver (Milman et al., 1988; Story et al., 1986). Exposure of partially hepatectomized 26 male Osborne-Mendel rats to a single i.p. dose of diethylnitrosamine (DEN) as an initiating agent 27 followed by promotion with 100 mg/kg-day of 1,1,2,2-tetrachloroethane by gavage 5 days/week 28 for 7 weeks produced a significantly increased number of GGT-positive foci in the liver (Milman 29 et al., 1988; Story et al., 1986). 1,1,2,2-Tetrachloroethane also significantly increased the 30 number of GGT-positive foci in rats administered the promotion protocol in the absence of the 31 DEN initiator. The study authors concluded that 1,1,2,2-tetrachloroethane induces 32 hepatocarcinogenesis primarily through a promoting mechanism (Story et al., 1986). 33 Using a mouse strain that had been shown to be susceptible to pulmonary adenomas 34 when exposed to organic chemicals, Theiss et al. (1977) administered i.p. injections of 80, 200, 35 or 400 mg/kg 1,1,2,2-tetrachloroethane in Tricaprylin 5-18 times to groups of 20 male A/St mice 36 for 8 weeks. There was a dose-related increase in number of lung tumors/mouse (Table 4-18), 37 and the dose-response was nearly statistically significant (Theiss et al., 1977). 38 54 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-18. Pulmonary adenomas from l,l?2,2-tetrachloroethane exposure in mice Dose/injection (mg/kg) Number of i.p. injections Total dose (mg/kg) Number of surviving animals Number of lung tumors/mouse 0 24 0 15/20 0.27±0.15 80 5 400 10/20 0.30±0.21 200 18 3,600 15/20 0.50 ±0.14 400 16 6,400 5/20 1.00 ±0.45 Source: Thiess et al. (1977). 1 2 Maronpot et al. (1986) tested 65 chemicals at three doses in 6- to 8-week-old male and 3 female strain A/St or A/J mice housed 10/cage. Doses were set based on the highest dose 4 exhibiting a lack of overt toxicity from a preliminary dose-setting study, with the mid and low 5 dose as half the higher dose. Mice were injected i.p. 3 times/week for 8 weeks. Lungs were 6 examined histologically. The data for 1,1,2,2-tetrachloroethane-exposed male and female strain 7 A/St are presented in Table 4-19. 8 Table 4-19. Pulmonary adenomas from l,l?2,2-tetrachloroethane exposure in A/St mice Compound Dose/injection (mg/kg) Vehicle Untreated control — - Saline vehicle control — - Tricaprylin vehicle control — - Urethan positive control 1,000 - 1,1,2,2-Tetrachloroethane 62.5 Tricaprylin 99 Tricaprylin 187.5 Tricaprylin Male A/St mice Number of surviving animals3 Percent survivors with tumors Tumors per mouseb 119/120 2 0.017 45/50 9 0.089 54/60 13 0.167 47/50 96 11.9 10/10 10 0.1 8/10 0 0 5/10 0 0 Female A/St mice Number of surviving animals3 Percent survivors with tumors Tumors per mouse 79/80 8 0.076 44/50 14 0.186 54/60 11 0.11 47/50 96 10.3 9/10 0 0 5/10 20 0.2 3/10 0 0 3Numerator is number of mice alive at study termination; denominator is number of mice started on study. Based on all surviving mice at study termination. Source: Maronpot et al. (1986). 55 DRAFT - DO NOT CITE OR QUOTE ------- 1 4.6. SYNTHESIS OF MAJOR NONCANCER EFFECTS 2 4.6.1. Oral 3 4.6.1.1. Human Data 4 Information on the acute oral toxicity of 1,1,2,2-tetrachloroethane in humans is available 5 from several case reports. Based on amounts of 1,1,2,2-tetrachloroethane recovered from the 6 gastrointestinal tract of deceased subjects following intentional ingestion (Mant, 1953; Sherman, 7 1953; Lilliman, 1949; Forbes, 1943; Elliot, 1933; Hepple, 1927), estimated lethal doses ranged 8 from 273 to 9,700 mg/kg. Patients who accidentally consumed a known volume of 1,1,2,2-tetra- 9 chloroethane, corresponding to single doses ranging from 68 to 117 mg/kg, as medicinal 10 treatment for hookworm experienced loss of consciousness and other clinical signs of narcosis 11 (Ward, 1955; Sherman, 1953). Chronic oral effects of 1,1,2,2-tetrachloroethane in humans have 12 not been reported in the literature. 13 14 4.6.1.2. Animal Data 15 Few studies have evaluated acute oral toxicity in animals, and the endpoints assessed 16 consist of data on lethality and neurological and liver effects (Table 4-20). Oral LDso values 17 ranged from 250 to 800 mg/kg in rats (NTP, 2004; Schmidt et al, 1980a; Gohlke et al, 1977; 18 Smyth et al., 1969). Neurological effects of acute, oral 1,1,2,2-tetrachloroethane administration 19 revealed ataxic effects and decreased passive avoidance behavior (Wolff, 1978). Hepatic 20 changes were noted in two separate acute oral toxicity studies. Male Sprague-Dawley rats 21 administered between 287 and 1,148 mg/kg 1,1,2,2-tetrachloroethane had dose-dependent 22 increases in the serum activity levels of AST and ALT as well as a decrease in hepatic 23 microsomal G6Pase activity (Cottalasso et al., 1998). Male Wistar rats were administered 100 24 mg/kg 1,1,2,2-tetrachloroethane and had increases in hepatic ascorbic acid levels and serum 25 leucine aminopeptidase activity, but no changes in serum ALT activity (Schmidt et al., 1980a, b). 26 Both studies noted increases in triglyceride levels in the liver. 56 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-20. Summary of noncancer results of major studies for oral exposure of animals to l,l?2,2-tetrachloroethane Species Sex Average daily dose (mg/kg-d) Exposure duration NOAEL (mg/kg-d) LOAEL (mg/kg-d) Response Comments Reference Acute exposure Rat (Wistar) Rat (Sprague- Dawley) Rat (Wistar) F M M 0, 25, 50, 80, 100 (gavage) 0, 143.5,287, 574, or 1,148 (gavage) 0 or 100 Single dose Single dose Single dose 25 143.5 100 50 287 ND Increased electric shock perception threshold. Increased serum AST activity and ALT activity, increased liver triglycerides levies; decreased liver dolichol levels. Increased hepatic ascorbic acid levels and serum leucine aminopeptidase activity Results suggestive of a subtle anesthetic effect. Ataxia observed at 100 mg/kg. Evaluations performed 1 hr postexposure. Approximately twofold increases in AST and ALT at >574 mg/kg. Liver histology and neurotoxicity not assessed. No changes in serium ALT Wolff, 1978 Cottalasso et al., 1998 Schmidt et al., 1980a,b Short-term exposure Rat (Osborne- Mendel) Mouse (B6C3FO Rat (F344/N) Rat (F344/N) Mouse (B6C3FO Rat (F344/N) M M M,F M M,F M 0,25,75, 150, or 300 (gavage) 0,25,75, 150, or 300 (gavage) 0, 135,270, or 540 (gavage) 0, 135,270, or 540 (gavage) 0,337.5, 675, or 1,350 (gavage) 0, 104, or 208 (gavage) 3-4 d 4d 12 doses in 16 d 12 doses in 16 d 12 doses in 16 d 13-21 d 150 300 135 135 ND ND 300 (PEL) ND 270 270 337.5 104 (PEL) CNS depression and mortality. No histopathological changes in liver. Decreased body weight in females, plus lethargy and increased organ weights. Lethargy, decreased body weight gain. Hepatocellular degeneration (females). Hepatic cytoplasmic vacuolization at low dose, mortality at high dose. Increased hepatocellular DNA synthesis and mitosis at >75 mg/kg-d; increased liver weight at > 150 mg/kg-d. No nonhepatic endpoints evaluated. Centrilobular swelling at >75 mg/kg-d and increased hepatocellular DNA synthesis and mitosis at > 150 mg/kg-d. No nonhepatic endpoints evaluated. The highest dose caused 100% mortality. Limited histology3. Mortality at 540 mg/kg-d. Limited histology3. Lethargy, increased liver weight, and mortality at higher doses. Limited histology3. No changes in body weight, kidney weights, kidney histology, or urinalysis. Dow Chemical Company, 1988 Dow Chemical Company, 1988 TSI Mason Laboratories, 1993a, unpubl. TSI Mason Laboratories, 1993b, unpubl. TSI Mason Laboratories, 1993c, unpubl. NTP, 1996; 57 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-20. Summary of noncancer results of major studies for oral exposure of animals to l,l?2,2-tetrachloroethane Species Rat (F344/N) Mouse (B6C3FO Sex M,F M F Average daily dose (mg/kg-d) 0, 300, 400, or 500 (diet) 3,325,6,650, 13,300,26,600, or 53,200 ppm Exposure duration 15 d 15 d NOAEL (mg/kg-d) ND ND LOAEL (mg/kg-d) 300 ND Response Decreased body weight gain. Decreased body weight, hyperactivity, decreased absolute and relative thymus weight, increased relative liver weight, pale or mottled livers, hepatocellular degeneration Comments Changes in liver and kidney weights and clinical signs at higher doses. Limited histology3. feed consumption could not be measured accurately Reference NTP, 2004 NTP, 2004; TSI Mason Laboratories, 1993d Subchronic exposure Rat (F344) Mouse (B6C3FO M,F M F 0, 20, 40, 80, 170, or 320 (diet) 0, 100, 200, 370, 700, or 1,360 (male); 0, 80, 160, 300, 600, or 1,400 (female) (diet) 14wks 14wks 20 40 80 40 80 160 Increased liver weight, as well as decreased sperm motility. Increased serum ALT activity, SDH activity, and cholesterol levels, reduced epididymis weight. Increased liver weight, increased ALT activity, ALP activity, SDH activity, and bile acids levels. Comprehensive study. More serious hepatic effects, including hepatocyte necrosis and bile duct hyperplasia, as well as effects on other organs, at >170 mg/kg-d. Comprehensive study. Wide array of endpoints evaluated, including histopathology. More serious hepatic effects, including hepatocyte necrosis and bile duct hyperplasia, as well as effects on other organs, at >300 mg/kg-d. NTP, 2004 NTP, 2004 Chronic exposure Rat (Osborne- Mendel) Mouse (B6C3FO M,F M,F 0, 62, or 108 (male) 0,43, or 76 (female) (gavage) 0, 142, or 284 (gavage) 78wks 78wks 62 (M) 76 (F)? ND 142 108 (M) ND(F) 142 (M) 284 (F) Fatty changes in liver. Reduced survival. Acute toxic tubular nephrosis, hydronephrosis, and chronic inflammation in the kidneys. Study is confounded by endemic chronic murine pneumonia, but this is unlikely to have contributed to the liver pathology. High incidences of hepatocellular tumors in all dose groups precluded evaluation of noncancer effects in the liver. NCI, 1978 NCI, 1978 58 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-20. Summary of noncancer results of major studies for oral exposure of animals to l,l?2,2-tetrachloroethane Species Sex Average daily dose (mg/kg-d) Exposure duration NOAEL (mg/kg-d) LOAEL (mg/kg-d) Response Comments Reference Developmental exposure Rat (Sprague- Dawley) Mouse (CD-I) F F 0, 34, 98, 180, 278, or 330 (diet) 0,987,2,120, 2,216, or 4,575 (diet) GDs 4-20 GDs 4-17 34 ND 98 ND Decreased maternal and fetal body weights. Maternal mortality and litter resorptions. Effects were more pronounced at higher doses. high mortality in the exposed mice precluded the identification of a NOAEL or LOAEL. Gulati et al., 1991a Gulati et al., 1991b aHistology only evaluated in animals with gross lesions. 59 DRAFT - DO NOT CITE OR QUOTE ------- 1 Short-term oral exposure (Table 4-18) to 1,1,2,2-tetrachloroethane produced clinical 2 signs of neurotoxicity and mortality at doses as low as 208-300 mg/kg-day by gavage in rats 3 (NTP, 1996; TSI Mason Laboratories, 1993a, b, unpublished; Dow Chemical Company, 1988). 4 Body weight gain was decreased at similar dose levels in rats exposed by gavage or diet (NTP, 5 2004; TSI Mason Laboratories, 1993a, b, unpublished; Dow Chemical Company, 1988; NCI, 6 1978). Hepatic effects consisted of increased DNA synthesis and centrilobular swelling in mice 7 exposed to 75 mg/kg-day in the diet (Dow Chemical Company, 1988) and hepatocellular 8 cytoplasmic vacuolation in rats exposed to 104 mg/kg-day (NTP, 1996). At higher doses (337.5 9 mg/kg-day), hepatocellular degeneration was observed in mice (TSI Mason Laboratories, 1993c, 10 unpublished). 11 Subchronic and chronic oral administration studies (Table 4-18) with 1,1,2,2-tetrachloro- 12 ethane in animals indicated that the liver is the most sensitive organ for toxicity. Oral toxicity 13 studies in F344 and Osborne-Mendel rats and B6C3Fi mice were evaluated (NTP, 2004, NCI, 14 1978). The 14-week subchronic study by the National Toxicology Program (NTP, 2004) in both 15 F344 rats and B6C3Fi mice was the most comprehensive evaluation of 1,1,2,2-tetrachloroethane- 16 mediated toxicity through an orally administered route. NCI (1978) conducted a chronic study 17 on Osborne Mendel rats and B6C3Fi mice in which dosing regimens were modified during the 18 course of the study. 19 In F344 rats, an increased incidence of hepatocellular cytoplasmic vacuolization was 20 observed at 20 mg/kg-day in males and 40 mg/kg-day in females, increased relative liver weights 21 were observed at 40 mg/kg-day, and hepatocellular hypertrophy was observed at 80 mg/kg-day 22 in the subchronic NTP (2004) study. Additional hepatic effects included increases in serum ALT 23 and SDH activity at 80 mg/kg-day, decreases in serum cholesterol levels at 80 mg/kg-day, and 24 increases in serum ALP activity and bile acids levels, hepatocellular necrosis, bile duct 25 hyperplasia, hepatocellular mitotic alterations, foci of cellular alterations, and hepatocyte 26 pigmentation at 170 and 320 mg/kg-day. A NOAEL of 20 mg/kg-day and a LOAEL of 40 27 mg/kg-day was selected based on the increase in relative liver weight; however, it should be 28 noted that an increased incidence of hepatocellular cytoplasmic vacuolization was observed at 20 29 and 40 mg/kg-day in male and female rats, respectively. In the Osborne-Mendel rats, significant 30 increases in hepatic fatty metamorphosis were observed in male rats following a chronic 31 exposure to 108 mg/kg-day (TWA, based on changes in dosing regimen) (NCI, 1978). Mortality 32 was significantly increased in female rats dosed at a TWA dose of 43 and 76 mg/kg-day; 33 however, the increased mortality was affected by the deaths of 10 high-dose females, 8 with 34 pneumonia and 2 with no reported lesions, during the first 5 weeks of the study. A NOAEL of 35 62 mg/kg-day and a LOAEL of 108 mg/kg-day were identified in male rats based on an 36 increased incidence of hepatic fatty metamorphosis (NCI, 1978). 37 Mice appear to be less sensitive than rats to noncancer effects mediated by orally 38 administered 1,1,2,2-tetrachloroethane. Relative liver weight was statistically significantly 60 DRAFT - DO NOT CITE OR QUOTE ------- 1 increased in female and male B6C3Fi mice at 80 and 200 mg/kg-day, respectively. Effects in the 2 mice also included minimal hepatocellular hypertrophy, increased serum SDH activity, ALT 3 activity, and bile acids levels, and decreased serum cholesterol levels at 160-200 mg/kg-day, and 4 increased serum ALP and 5'-nucleotidase activities, necrosis, pigmentation, and bile duct 5 hyperplasia at 300-370 mg/kg-day. Based on the increase in relative liver weight observed in 6 the NTP (2004) study, a NOAEL of 100 mg/kg-day and a LOAEL of 200 mg/kg-day in male 7 mice and a LOAEL of 80 mg/kg-day in female mice was identified . In addition, male and 8 female B6C3Fi mice were evaluated for chronic oral toxicity by NCI (1978). For this study, a 9 LOAEL of 142 mg/kg-day was selected for chronic inflammation in the kidneys of male mice, 10 while a NOAEL of 142 mg/kg-day and a LOAEL of 284 mg/kg-day were selected for 11 hydronephrosis and chronic inflammation in the kidneys of female mice. 12 Comprehensive neurobehavioral testing showed no evidence of neurotoxicity in either 13 species at doses equal to or higher than the LOAELs based on liver effects (NTP, 2004), 14 indicating that the liver is more sensitive than the nervous system to subchronic dietary exposure 15 to 1,1,2,2-tetrachloroethane. 16 Developmental parameters were significantly affected by oral administration of 17 1,1,2,2-tetrachloroethane in rats and mice. Significant decreases in rat maternal and fetal body 18 weights were noted at doses of >98 mg/kg-day (Gulati et al, 1991a). Using statistical 19 significance and a 10% change as the criteria for establishing an adverse effect in maternal body 20 weight, a NOAEL of 34 mg/kg-day and LOAEL of 98 mg/kg-day were selected. A NOAEL of 21 34 mg/kg-day and LOAEL of 98 mg/kg-day were selected for developmental toxicity based on 22 the lowest dose that produced a statistically significant decrease in fetal body weight. In mice, 23 the FEL based on maternal toxicity and resorption of litters is 2,120 mg/kg-day (Gulati et al., 24 1991 b). The high mortality in the exposed mice precluded the identification of a NOAEL or 25 LOAEL from this study. 26 Toxicity to reproductive tissues following 1,1,2,2-tetrachloroethane exposure to adult rats 27 and mice was observed at dose levels as low as 40 mg/kg-day (NTP, 2004). In male rats, sperm 28 motility was decreased at >40 mg/kg-day. Higher doses resulted in decreased epididymal 29 absolute weight and increased atrophy of the preputial and prostate gland, seminal vesicle, and 30 testicular germinal epithelium. In female rats, minimal to mild uterine atrophy was increased at 31 >170 mg/kg-day and clitoral gland atrophy and ovarian interstitial cell cytoplasmic alterations 32 were increased at 320 mg/kg-day. Female F344 rats in the 170 mg/kg-day group spent more 33 time in diestrus than did the vehicle controls. 34 Male B6C3Fi mice had increased incidences of preputial gland atrophy at >100 mg/kg- 35 day. Less sensitive effects included decreases in absolute testis weight (>700 mg/kg-day) and 36 absolute epididymis and cauda epididymis weights (1,360 mg/kg-day) and a decrease in 37 epididymal spermatozoal motility (1,360 mg/kg-day). The only noted reproductive toxicity 61 DRAFT - DO NOT CITE OR QUOTE ------- 1 parameter in female mice affected was a significant increase in the length of the estrous cycle at 2 a dose of 1,400 mg/kg-day (NTP, 2004). 3 4 4.6.2. Inhalation 5 4.6.2.1. Human Data 6 Limited information is available on the acute inhalation toxicity of 1,1,2,2-tetrachloro- 7 ethane in humans (Table 4-21). The results of an early, poorly reported experimental study with 8 two volunteers suggest that 3 ppm (6.9 mg/m3) was the odor detection threshold. Irritation of the 9 mucous membranes, pressure in the head, vertigo, and fatigue were observed at 146 ppm (1,003 10 mg/m3) for 30 minutes or 336 ppm (2,308 mg/m3) for 10 minutes. Common reported symptoms 11 of high-level acute inhalation exposure to 1,1,2,2-tetrachloroethane in humans include 12 drowsiness, nausea, headache, and weakness, and at extremely high concentrations, jaundice, 13 unconsciousness, and respiratory failure (Coyer, 1944; Hamilton, 1917). 62 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-21. Summary of noncancer results of major human studies of inhalation exposure to 1,1^2,2-tetrachloroethane Study population Sex Exposure level (mg/m3) Exposure NOAEL duration (mg/m3) LOAEL (mg/m3) Response Comments Reference Acute exposure Two volunteers NS 6.9-2,308 30 min ND ND Irritation, vertigo, head pressure, fatigue. Effect levels could not be determined due to limited analysis. Lehmann et al., 1936 Occupational exposure 127 coating workers Workers from 3 9 chemical processing plants 380 workers from 23 factories 34-75 workers in penicillin production NS NS M,F NS 500-1,500 NS 62.5-672 10-1,700 NS NS Generally <1 yr Up to 3 yrs ND ND ND ND ND ND ND ND Decreased whole blood specific gravity, decreased RBC count, lymphocytosis, unspecified neurological findings. Increased mortality for lymphatic cancers. Anemia, loss of appetite, abdominal pain, headache, vertigo, and tremors. Loss of appetite, epigastric pain, hepatic enlargement, urobilinogenuria, weakness, fatigue, weight loss, and itching. Effect levels could not be determined due to limited analysis. Mortality from cardiovascular disease, cirrhosis of the liver, and digestive or respiratory cancers was not elevated. Effect levels could not be determined due to a lack of a control population and possible coexposure. Effect levels could not be determined due to a lack of a control population, limited reporting, and possible coexposure. Horiguchi et al., 1964 Norman etal, 1981 Lobo-Mendonca, 1963 Jeney et al., 1957 ND = not determined; NS = not stated 1 63 DRAFT - DO NOT CITE OR QUOTE ------- 1 Chronic toxicity of inhaled 1,1,2,2-tetrachloroethane in humans (Table 4-19) resulted in 2 neurological symptoms including headache, weakness, fatigue, and hematological changes such 3 as anemia and elevated WBC count (Norman et al, 1981; Lobo-Mendonca, 1963; Jeney et al, 4 1957; Minot and Smith, 1921). Most occupational exposure studies failed to evaluate hepatic 5 endpoints, other than an urobilinogen test. Jeney et al. (1957) reported a positive relationship 6 between duration of exposure and frequency of abnormal liver function test results, loss of 7 appetite, bad taste in the mouth, epigastric pain, and a "dull straining pressure feeling in the area 8 of the liver". 9 10 4.6.2.2. Animal Data 11 Acute inhalation exposures in animals (Table 4-22) resulted in near-lethal or lethal effects 12 at levels >1,000 ppm (Schmidt et al., 1980a; Price et al., 1978; Horiuchi et al., 1962; Carpenter et 13 al., 1949; Pantelitsch, 1933). Death was typically preceded by signs of CNS toxicity (e.g., 14 incoordination, loss of reflexes, labored respiration, prostration, and loss of consciousness) and 15 was often accompanied by congestion and fatty degeneration of the liver. Nonlethal exposures 16 increased lipid and triglyceride levels in the liver in mice following exposure to 600-800 ppm 17 (4,120-5,490 mg/m3)for 3 hours (Tomokuni, 1970, 1969). Nonlethal exposures also reduced 18 motor activity in rats following exposure to 576 ppm (3,950 mg/m3) for 30 minutes (Price et al., 19 1978) and 360 ppm (2,470 mg/m ) for 6 hours (Horvath and Frantik, 1973) and in guinea pigs 20 following exposure to 576 ppm (3,950 mg/m3) (Price et al., 1978). 64 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-22. Summary of noncancer results of major studies for inhalation exposure of animals to l,l?2,2-tetrachloroethane. Species Sex Exposure level (mg/m3) Exposure duration NOAEL (mg/m3) LOAEL (mg/m3) Response Comments Reference Acute exposure Rat Rat (Wistar) Rat (Sherman) Rat Guinea pig Rat (NR) Mouse (Cb) Mouse (Cb) Mouse Mouse Rat NR M NR NR NR NR F F NS M M NR 0,410,700, 1,030,2,100, or 4,200 6870 3,950, 34,700, or 43,350 3,950, 34,700, or 43,350 1,370 or 2,470 4,120 5,490 7,000, 8,000- 10,000, 17,000, 29,000, or 34,000 40,500 or 45,300 0, 69, 690, or 6,900 4Hrs 4Hrs 4Hrs 30 mins 30 mins 6Hrs 3Hrs 3Hrs 1.5-2Hrs 3Hrs 6Hrs NR ND ND ND ND ND ND ND ND ND ND 8,600 ND ND 3,950 3,950 2,470 4,120 ND 7,000 ND 69 LC50 24-Hr observation. Hepatic effects included histological alterations and increases in serum enzymes and liver triglycerides. Identification of a NOAEL or LOAEL precluded by reporting inadequacies. Mortality slight reduction in activity and alertness; lacrimation, ataxia, narcosis, labored respiration, and 30-50% mortality when concentration increased Eye closure, squinting, lacrimation, and decreased activity; tremors, narcosis, and labored breathing and mortality when concentration increased Effective concentration for a 50% decrease in spontaneous motor activity. Increased hepatic lipid and triglyceride levels, decreased hepatic ATP. Increased tricglyceride and decreased phospholipid levels Disturbed equilibrium, prostration, and loss of reflexes. Effective concentration for a 50% increase in pentobarbital sleep time was 1,370 mg/m3. A limited number of endpoints were evaluated. effects generally resolved by 90 hours postexposure Limited number of endpoints and poor reporting. Mortality at >8,000 mg/m3. Mortality: 3/10 and 4/10, respectively minimal increase in serum AST at all exposure concentrations 72 hours postexposure Schmidt et al, 1980a Schmidt et al, 1980a Carpenter etal, 1949 Price etal, 1978 Price etal., 1978 Horvath and Frantik, 1973 Tomokuni, 1969 Tomokuni, 1970 Pantelitsch, 1933 Horiuchietal, 1962 Deguchi, 1970 65 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-22. Summary of noncancer results of major studies for inhalation exposure of animals to l,l?2,2-tetrachloroethane. Species Sex Exposure level (mg/m3) Exposure duration NOAEL (mg/m3) LOAEL (mg/m3) Response Short-term exposure Rat Rat Mouse M M M Oor 15 62,000 48,000 4 Hrs/d for up to eight exposures inlOd 2 Hrs/d, 2-3 times a week for 1 1 exposures in 29 d 2 Hrs/d for 5 exposures in 29 d ND ND ND ND ND ND Comments Reference Increases in serum proteins and histological alterations in the liver. Identification of a NOAEL or LOAEL precluded by reporting inadequacies. All rats died during the study. No changes in body weight were reported. Exposed animals generally showed moderate congestion and fatty degeneration of the liver Moderate congestion and fatty degeneration of the liver Subchronic exposure Rat (Osborne- Mendel) Mouse (B6C3F1) Rat (Sprague- Dawley) Monkey (Macaca sp.) Rats Mongrel dog Rabbits M, F M, F F M M,F M NS 0, 56, 100, 178,316,or 562 0,32,56, 100, 178, or 316 0 or 3,909 13,560 Oor 1,150 Oor 1,150 Oor 10 5 d/wk for 6 wks 5 d/wk for 6 wks 5-6 Hrs/d, 5 d/wk for 15 wks 2 hrs/d, 6 d/wk for total of 190 exposures in 9 mo 7 hrs/d for 6 mo 7 hrs/d for 6 mo 3 hrs/d, 6 d/wk for 7-8.5 mo 100 (male) 56 (female) 316 ND ND ND ND ND 178 (male) 100 (female) ND ND ND ND ND ND Decreased body weight gain Body weight changes and mortality Most (5/9) of the mice died within 5 days of the first exposure Gohlke and Schmidt, 1972; Schmidt etal, 1972 Horiuchi et al, 1962 Horiuchi et al, 1962 Mortality and body weight gain were the only endpoints used to assess toxicity Mortality and body weight gain were the only endpoints used to assess toxicity Increased liver weight, transient liver cytoplasmic vacuolization. Identification of a NOAEL or LOAEL precluded by reporting inadequacies. Fatty degeneration and splenic congestion. Identification of a LOAEL or NOAEL is precluded by the use of a single animal and lack of control. Pathological effects in the liver, kidney, and lung, precluded by an endemic lung infection. Increased serum phosphatase and blood urea nitrogen levels, cloudy swelling of the liver and convoluted tubule of the kidney, and light congestion of the lungs. A NOAEL or LOAEL was not identified due to single treated dog Altered serum acetylcholine levels. A NOAEL or LOAEL can not be identified due to incomplete NCI, 1978 NCI, 1978 Truffertetal, 1977 Horiuchi etal., 1962 Mellon Institute of Industrial Research, 1947 Mellon Institute of Industrial Research, 1947 Kulinskaya and Verlinskaya, 1972 66 DRAFT - DO NOT CITE OR QUOTE ------- Table 4-22. Summary of noncancer results of major studies for inhalation exposure of animals to l,l?2,2-tetrachloroethane. Species Rabbits Sex NS Exposure level (mg/m3) 0,2, 10, or 100 Exposure duration 3 hrs/d, 6 d/wk for 8- 10 mo NOAEL (mg/m3) ND LOAEL (mg/m3) ND Response quantitation. Comments Increase and decrease in total antibody levels, increase in the mobility of specific antibodies, decrease in the relative content of y-globulin antibodies and an increase in the T and |3 fractions. Poorly reported study that provides inadequate quantitative data. Reference Shmuter, 1977 Chronic exposure Rats M 0 or 13.3 4 hrs/d, 110 or 265 d ND ND Increased leukocyte and Pi-globulin levels, increased percentage of segmented nucleated neutrophils, decreased percentage of lymphocytes, increased liver total fat content. Experimental design and results were poorly reported and histological examinations do not appear to have been conducted. Schmidt et al, 1972 ND = not determined 67 DRAFT - DO NOT CITE OR QUOTE ------- 1 Acute and short-term inhalation exposure (Table 4-22) to high concentrations (>7,000 2 ppm) of 1,1,2,2-tetrachloroethane produced mortality and neurological and liver effects in 3 animals. Mortality occurred in mice exposed to 7,000 ppm (48,000 mg/m ) for 2 hours 4 once/week for 4 exposures in 29 days and in rats exposed to 9,000 ppm (62,000 mg/m3) for 2 5 hours/day, 2-3 times/week for 11 exposures in 29 days. Congestion and fatty degeneration in 6 the liver (mice and rats), as well as a biphasic change in neurological motor activity 7 (hyperactivity followed by ataxia, rats only), were also reported (Horiuchi et al., 1962). At the 8 lowest inhalation exposure of 2.2 ppm (15 mg/m ) for 4 hours/day (8-10 days), rats had fine 9 droplet fatty degeneration in the liver and changes in levels of serum proteins, but no 10 neurological changes were reported (Gohlke and Schmidt, 1972; Schmidt et al., 1972). 11 There are a few subchronic inhalation exposure studies and one chronic exposure study 12 with 1,1,2,2-tetrachloroethane (Table 4-20). Overall these studies either had poor study designs, 13 one exposure concentration, low number of animals, or a combination of the above. The 14 available subchronic and chronic inhalation studies indicate that the liver was the most sensitive 15 organ to 1,1,2,2-tetrachloroethane exposure. Increased relative liver weights were reported at 16 exposures of 560 ppm (3,909 mg/m ) for 15 weeks (Truffert et al., 1977). Other transient hepatic 17 changes (e.g., histological alterations and cytoplasmic vacuolation) were observed, but these 18 effects did not persist (Truffert et al., 1977). In the chronic exposure study, rats exposed to 13.3 19 mg/m3 (1.9 ppm) 1,1,2,2-tetrachloroethane 4 hours/day for 265 days exhibited increased liver fat 20 content (Schmidt et al., 1972). In the third rat study (Mellon Institute of Industrial Research, 21 1947), none of the effects noted from 1,1,2,2-tetrachloroethane exposure could be evaluated 22 since the control animals experienced a high degree of pathological effects in the kidneys, liver, 23 and lung. Hepatic effects from long-term exposure to 1,1,2,2-tetrachloroethane were also 24 reported in a study with one mongrel dog with cloudy swelling of the liver at 167 ppm (1,150 25 mg/m3) for 6 months (Mellon Institute of Industrial Research, 1947) and one male monkey with 26 fatty degeneration of the liver at 1,974 ppm (13,560 mg/m3) for 9 months (Horiuchi et al., 1962). 27 Other endpoints that were observed following subchronic and chronic inhalation 28 exposure are described below. Hematological alterations, including increased leukocyte and 29 Pi-globulin levels, increased percentage of segmented nucleated neutrophils and decreased 30 percentage of lymphocytes, decreased y-globulin, and decreased adrenal ascorbic acid levels, 31 were observed in rats exposed to 1.9 ppm (13.3 mg/m3) for 265 days (Schmidt et al., 1972), and 32 splenic congestion was noted in a study of a single monkey (Horiuchi et al., 1962). In the 33 mongrel dog study noted above, cloudy swelling of the convoluted tubules of the kidneys and 34 light congestion of the lungs were observed (Mellon Institute of Industrial Research, 1947). 35 Kulinskaya and Verlinskaya (1972) observed alterations in serum acetylcholine levels in rabbits 36 exposed to 10 mg/m3 (1.5 ppm) 3 hours/day, 6 days/week for 7-8.5 months. Shmuter (1977) 37 observed immunological alterations (changes in antibody levels) in rabbits exposed to 2-100 38 mg/m3 (0.3-14.6 ppm) 3 hours/day, 6 days/week for 8-10 months. 68 DRAFT - DO NOT CITE OR QUOTE ------- 1 A reproductive toxicity assessment was conducted on seven male rats exposed to 2 13.3 mg/m3 1,1,2,2-tetrachloroethane for 258 days. No significant changes in reproductive 3 parameters were observed, indicating that 13.3 mg/m3 (1.9 ppm) was a NOAEL for male 4 reproductive effects in the rat (Schmidt et al, 1972). 5 6 4.6.3. Mode of action Information 7 1,1,2,2-Tetrachloroethane is rapidly and extensively absorbed following both oral and 8 inhalation exposures, with absorption of 70-100% following oral exposure in animals (Dow 9 Chemical Company, 1988; Mitoma et al., 1985) and 40-97% following inhalation exposures in 10 humans (Morgan et al., 1970; Lehmann et al., 1936). Following absorption, the chemical is 11 distributed throughout the body, although the high tissue:air partition coefficient for fat (Gargas 12 et al., 1989) suggests that it may accumulate more in lipid-rich tissues. Metabolism is extensive, 13 with >68% of a total administered dose generally found as metabolites (Dow Chemical Company, 14 1988; Mitoma et al., 1985; Yllner, 1971), and is believed to occur mostly in the liver. Urinary 15 elimination occurs mainly as metabolites, including dichloroacetic acid, glyoxalic acid, formic 16 acid, trichloroethanol, and trichloroacetic acid, while a fraction of an absorbed dose may be 17 eliminated in expired air as parent compound or carbon dioxide. 18 Metabolism of 1,1,2,2-tetrachloroethane to reactive products is likely to play a key role in 19 its toxicity. Both nuclear and microsomal cytochrome P450 enzymes have been implicated in 20 the metabolism of the compound, possibly forming a number of biologically active compounds 21 including aldehydes, alkenes, acids, and free radicals (see Figure 3-1 in Section 3.3), which may 22 react with biological tissues. Evidence for metabolism to reactive compounds comes from 23 studies of radiolabel incorporation following single doses of radiolabeled 1,1,2,2-tetrachloro- 24 ethane in which incorporated radiolabel was enhanced by pretreatment with phenobarbital, 25 xylene, or ethanol, and the variety of inducers capable of influencing this effect suggest that 26 multiple P450 isozymes may be involved (Casciola and Ivanetich, 1984; Halpert, 1982; Sato et 27 al., 1980), including members of the CYP2A, CYP2B, CYP2E, and CYP3A subfamilies 28 (Omiecinski et al., 1999; Nebert et al., 1987). Additionally, mice are known to metabolize 29 1,1,2,2-tetrachloroethylene at a 1.1 -3.5 -fold greater rate than rats and have been demonstrated to 30 have approximately a twofold greater binding of radiolabel to tissues, further implicating 31 metabolic activation as a possible step in the mode of action. However, there is uncertainty as to 32 whether the presence of radiolabel in proteins, DNA, and RNA may be radiolabeled carbon that 33 has been incorporated into biomolecules through normal biochemical processes. Studies 34 describing the mechanism of 1,1,2,2-tetrachloroethane-induced noncancer toxicological effects 35 are not available. 36 69 DRAFT - DO NOT CITE OR QUOTE ------- 1 4.7. EVALUATION OF CARCINOGENICITY 2 4.7.1. Summary of Overall Weight of Evidence 3 Under the Guidelines for Carcinogen Risk Assessment (U.S. EPA, 2005a) 1,1,2,2-tetra- 4 chloroethane is "likely to be carcinogenic to humans" based on data from an oral cancer bioassay 5 in male and female Osborne-Mendel rats and B6C3Fi mice (NCI, 1978). In B6C3Fi mice, a 6 statistically significant increase in the incidence of hepatocellular carcinomas in both genders 7 was observed at doses of 142 and 284 mg/kg-day. A decrease in the time to tumor in both 8 genders of mice was also observed. In this same bioassay, male Osborne-Mendel rats exhibited 9 an increased incidence of hepatocellular carcinomas, a rare tumor in this strain (NCI, 1978), at 10 the high dose only, although this increased incidence was not statistically significant. An 11 untreated female control rat also developed a hepatocellular carcinoma. Limitations in the study 12 included increased mortality in male and female mice and the variable doses given to the mice 13 over the course of the 78-week exposure period. In the high-dose male mice, acute toxic tubular 14 nephrosis was characterized as the cause of death in the mice that died prior to study termination, 15 although hepatocellular carcinomas were observed in most of these mice. 16 The predominant proposed metabolic pathway for 1,1,2,2-tetrachloroethane involves 17 production of dichloroacetic acid (Casciola and Ivanetich, 1984; Halpert and Neal, 1981; Yllner, 18 1971). Dichloroacetic acid was identified as the major urinary metabolite in mice treated with 19 1,1,2,2-tetrachloroethane by i.p. injection (Yllner et al, 1971) and in in vitro systems with rat 20 liver microsomal and nuclear cytochrome P450 (Casciola and Ivanetich, 1984; Halpert, 1982; 21 Halpert and Neal, 1981). Other pathways involve the formation of trichloroethylene, via 22 dehydrochlorination, or tetrachloroethylene, via oxidation, as initial metabolites (Mitoma et al., 23 1985; Ikeda and Ohtsuji, 1972; Yllner et al., 1971). 1,1,2,2-Tetrachloroethane may also form 24 free radicals by undergoing reductive dechlorination (ATSDR, 1996). 25 Dichloroacetic acid induces hepatocellular carcinomas in both genders of F344 rats and 26 B6C3Fi mice (DeAngelo et al., 1999; DeAngelo et al., 1996; Pereira, 1996; Pereira and Phelps, 27 1996; Ferreira-Gonzalez et al., 1995; Richmond et al., 1995; Daniel et al., 1992; DeAngelo et al., 28 1991; U.S. EPA, 1991b; Bull et al., 1990; Herren-Freund et al., 1987). Trichloroethylene, also a 29 metabolite of 1,1,2,2-tetrachloroethane, has been shown to produce hepatocellular carcinomas 30 and hepatocellular adenomas in male and female B6C3Fi mice, respectively, but did not 31 demonstrate carcinogenicity in Osborne-Mendel or Sprague-Dawley rats (NTP, 1990; NCI, 32 1976). Tetrachloroethylene, another metabolite of 1,1,2,2-tetrachloroethane, was characterized 33 by NCI (1977) as a liver carcinogen in B6C3Fi mice, but an evaluation of carcinogenicity in 34 Osborne-Mendel rats was inadequate due to early mortality. In a study by NTP (1986), 35 tetrachloroethylene demonstrated evidence of carcinogenicity in F344 rats, as shown by 36 increased incidences of mononuclear cell leukemia, and in B6C3Fi mice, as shown by increased 37 incidences of hepatocellular adenomas and carcinomas in males and carcinomas in females. 70 DRAFT - DO NOT CITE OR QUOTE ------- 1 Additional information on the carcinogenic potential comes from studies on the tumor 2 initiating and promoting activity in mammalian cells (Colacci et al, 1996, 1992). The results of 3 the in vivo and in vitro genotoxicity studies for 1,1,2,2-tetrachloroethane, which were generally 4 non-positive, provide limited evidence of a mutagenic mode of action and are insufficient for 5 establishing a mutagenic mode of action. 6 No animal cancer bioassay data following inhalation exposure to 1,1,2,2-tetrachloro- 7 ethane are available. However, U.S. EPA's Guidelines for Carcinogen Risk Assessment (2005a) 8 indicates that for tumors occurring at a site other than the initial point of contact the cancer 9 descriptor generally applies to all routes of exposure that have not been adequately studied unless 10 there is convincing information to indicate otherwise. No additional information is available for 11 1,1,2,2-tetrachloroethane. Thus, 1,1,2,2-tetrachloroethane is considered "likely to be 12 carcinogenic to humans" by any route of exposure. 13 The weight of evidence for the carcinogenicity of 1,1,2,2-tetrachloroethane could be 14 strengthened by additional cancer bioassays demonstrating tumor development. Currently, the 15 NCI (1978) bioassay is the only study available demonstrating 1,1,2,2-tetrachloroethane 16 tumorgenicity. The NCI (1978) study was a 78-week study, compared to a 104-week bioassay, 17 and the limitations of the study included increased mortality in male and female mice, the 18 variable doses given to the mice over the course of the 78-week exposure period, and the acute 19 toxic tubular nephrosis, characterized as the cause of death, in the high-dose male mice that died 20 prior to study termination (although hepatocellular carcinomas were observed in most of these 21 mice). 22 23 4.7.2. Synthesis of Human, Animal, and Other Supporting Evidence 24 Only one study in humans evaluated the possible carcinogenic effects of 1,1,2,2-tetra- 25 chloroethane. Norman et al. (1981) evaluated groups of clothing-treatment workers employed 26 during World War II in which some workers used 1,1,2,2-tetrachloroethane and some used water. 27 Inhalation exposure concentrations and durations were not reported and dermal exposures were 28 likely. In addition, coexposures to dry-cleaning chemicals occurred. No differences in standard 29 mortality ratios were seen between the 1,1,2,2-tetrachloroethane and water groups for total 30 mortality, cardiovascular disease, cirrhosis of the liver, or cancer of the digestive and respiratory 31 systems. The mortality ratio for lymphatic cancers in the 1,1,2,2-tetrachloroethane group was 32 increased relative to controls and the water group, although the number of deaths was small 33 (4 cases observed compared to 0.85 cases expected). No other information was located 34 regarding the carcinogenicity of 1,1,2,2-tetrachloroethane in humans. 35 The only comprehensive animal study that evaluated the carcinogenicity of 1,1,2,2-tetra- 36 chloroethane was performed by the NCI (1978). Male and female Osborne-Mendel rats were 37 exposed to TWA doses of 0, 62, or 108 mg/kg-day (males) or 0, 43, or 76 mg/kg-day (females) 38 5 days/week for 78 weeks, followed by a 32-week observation period during which the rats were 71 DRAFT - DO NOT CITE OR QUOTE ------- 1 not exposed. No statistically significant increases in tumor incidences were observed in rats. 2 However, two hepatocellular carcinomas, which were characterized by NCI (1978) as rare in 3 Osbourne-Mendel rats, and one neoplastic nodule were observed in the high-dose male rats. A 4 hepatocellular carcinoma was also observed in a female rat in the control group. NCI (1978) 5 characterized the carcinogenic results in male rats as "equivocal." Male and female B6C3Fi 6 mice were exposed to TWA doses of 0, 142, or 284 mg/kg-day 5 days/week for 78 weeks, 7 followed by a 12-week observation period during which the mice were not exposed. Statistically 8 significant, dose-related increases in the incidence of hepatocellular carcinoma were observed in 9 males (3/36, 13/50, and 44/49 in the control, low-, and high-dose groups, respectively) and 10 females (1/40, 30/48, and 43/47, respectively). In addition, a decrease in the time to tumor for 11 the hepatocellular carcinomas was also evident in both genders of mice. Lymphomas were also 12 seen in the male and female mice, but the incidences were not found to be statistically significant. 13 The only other available study observed pulmonary adenomas in female Strain A/St mice given 14 99 mg/kg injections i.p. 3 times/week for 8 weeks (Maronpot et al., 1986). 15 In vitro studies of the genotoxicity of 1,1,2,2-tetrachloroethane have yielded mixed, 16 though mainly nonpositive, results. Mutagenicity studies in S. typhimurium were predominantly 17 negative, with only 2 of 10 available studies reporting activity (NTP, 2004; Ono et al., 1996; 18 Roldan-Arjona et al., 1991; Milman et al., 1988; Warner et al., 1988; Mitoma et al., 1984; 19 Haworth et al., 1983; Nestmann et al., 1980; Rosenkranz, 1977; Brem et al., 1974). Mixed 20 results were reported for gene conversion, reversion, and recombination in S. cerevisiae 21 (Nestmann and Lee, 1983; Callen et al., 1980), and aneuploidy, but not mitotic cross over, was 22 induced in A. nidulans (Crebelli et al., 1988). Tests for DNA damage inE. coli were positive 23 (DeMarini and Brooks, 1992; Rosenkranz, 1977; Brem et al., 1974). 1,1,2,2-Tetrachloroethane 24 was not mutagenic in mouse L5178Y lymphoma cells (NTP, 2004) and was negative in tests for 25 DNA damage in other mammalian cells, including induction of DNA repair in primary rat or 26 mouse hepatocytes (Milman et al., 1988; Williams, 1983), induction of chromosomal aberrations 27 in CHO cells (NTP, 2004; Galloway et al., 1987), and induction of cell transformation in 28 BALB/C-3T3 cells (Colacci et al., 1992; Milman et al., 1988; Tu et al., 1985; Arthur Little, Inc., 29 1983). 1,1,2,2-Tetrachloroethane was positive for induction of SCEs in both BALB/C-3T3 30 (Colacci et al., 1992) and CHO cells (NTP, 2004; Galloway et al., 1987) and for induction of cell 31 transformation in BALB/C-3T3 cells at high (cytotoxic) doses (Colacci et al., 1990). 32 1,1,2,2-Tetrachloroethane also had mixed results for genotoxicity following in vivo 33 exposure. Tests for sex-linked recessive lethal mutations and mitotic recombination in 34 Drosophila were negative (NTP, 2004; Vogel and Nivard, 1993; Woodruff et al., 1985; 35 McGregor, 1980). Both positive (Miyagawa et al., 1995) and negative results (Mirsalis et al., 36 1989) have been reported in mouse hepatocytes tested for UDS, and tests for S-phase DNA 37 induction in hepatocytes were negative in male mice and equivocal in female mice (Mirsalis et 72 DRAFT - DO NOT CITE OR QUOTE ------- 1 al., 1989). Rat bone marrow cells were negative for chromosomal aberrations in male rats, but 2 positive in female rats (McGregor, 1980). 3 1,1,2,2-Tetrachloroethane showed promoting activity, but limited initiating activity, in rat 4 liver preneoplastic (GGT-positive) foci assays (Milman et al., 1988; Story et al., 1986). 5 1,1,2,2-Tetrachloroethane initiated, but did not promote, neoplastic transformation in mouse 6 BALB/c-3t3 cells (Colacci et al., 1996, 1992). 7 8 4.7.3. Mode of action Information 9 The mode of action of the carcinogenic effects of 1,1,2,2-tetrachloroethane is unknown. 10 Colacci et al. (1987) reported possible covalent binding of radiolabeled 1,1,2,2-tetrachloroethane 11 to DNA, RNA, and protein in the liver, kidneys, lung, and stomach of rats and mice exposed to a 12 single intravenous dose and analyzed 22 hours postexposure. However, the conclusion of 13 covalent binding may be influenced by the presence of radiolabel in the DNA, RNA, and protein 14 that was the result of incorporated radiolabeled carbon into the biomolecules through normal 15 biochemical processes. 16 The mutagenicity data for 1,1,2,2-tetrachloroethane are inconclusive, with in vitro 17 genotoxicity tests generally reporting negative results except for assays of SCE and cell 18 transformation, and in vivo tests of genotoxicity showing a similar pattern. Several studies have 19 reported increases in the number of hepatocytes in mitosis, but the possible role these effects 20 may have on the carcinogenicity of 1,1,2,2-tetrachloroethane has not been evaluated. The results 21 of rat liver preneoplastic foci and mouse BALB/C-3T3 cell neoplastic transformation assays 22 suggest that 1,1,2,2-tetrachloroethane may have initiating and promoting activity (Colacci, 1996, 23 1992; Milman et al., 1988; Story et al., 1986), but tumor initiation and promotion studies have 24 not been conducted. 25 Tumor formation by 1,1,2,2-tetrachloroethane may involve metabolism to one or more 26 active compounds, with the predominant pathway leading to the production of dichloroacetic 27 acid (Casciola and Ivanetich, 1984; Halpert and Neal, 1981; Yllner, 1971). 1,1,2,2-Tetrachloro- 28 ethane is metabolized extensively following absorption, at least in part, by cytochrome P450 29 enzymes from the members of the CYP2A, CYP2B, CYP2E, and CYP3A subfamilies (see 30 Section 3.3). Mice are known to metabolize 1,1,2,2-tetrachloroethane to a greater extent than 31 rats, which may, in part, account for the fact that liver tumors occurred in mice at statistically 32 significant levels, but not in rats, following chronic oral exposure. 33 Dichloroacetic acid, which appears to be the main metabolite of 1,1,2,2-tetrachloroethane, 34 induces hepatocellular carcinomas in both genders of F344 rats and B6C3Fi mice (DeAngelo et 35 al., 1999; DeAngelo et al., 1996; Pereira, 1996; Pereira and Phelps, 1996; Ferreira-Gonzalez et al., 36 1995; Richmond et al., 1995; Daniel et al., 1992; DeAngelo et al., 1991; U.S. EPA, 1991b; Bull et al., 37 1990; Herren-Freund et al., 1987). Dichloroacetic acid is recognized as hepatocarcinogenic in 38 both genders of two rodent species 73 DRAFT - DO NOT CITE OR QUOTE ------- 1 1,1,2,2-tetrachloroethane may be metabolized to form free radicals, which may, in turn, 2 covalently bind to macromolecules, including DNA. Formation of free radicals during 3 1,1,2,2-tetrachloroethane metabolism has been demonstrated in spin-trapping experiments 4 (Tomasi et al, 1984). Both nuclear and microsomal forms of cytochrome P450 enzymes have 5 been implicated in this process, as increased metabolism and covalent binding of metabolites 6 following pretreatment with phenobarbital (Casciola and Ivanetich, 1984; Halpert, 1982), xylene 7 (Halpert, 1982), or ethanol (Sato et al., 1980) have been reported. The presence of covalently 8 bound label has been reported following inhalation (Dow Chemical Company, 1988), oral 9 (Mitoma et al., 1985), and intravenous (Eriksson and Brittebo, 1991) administration of 10 radiolabeled 1,1,2,2-tetrachloroethane. 11 In summary, only limited data are available regarding the possible mode(s) of action of 12 1,1,2,2-tetrachloroethane carcinogenicity. Metabolism to one or more active compounds may 13 play a role in tumor development. Results of genotoxicity studies of 1,1,2,2-tetrachloroethane 14 are mixed and provide inconclusive evidence for establishing a mutagenic mode of action. 15 There is some evidence to indicate that the mode of carcinogenic action may involve 16 tumor promotion. Milman et al. (1988) and Story et al., (1986) concluded that 1,1,2,2-tetra- 17 chloroethane induces hepatocarcinogenesis primarily through a promoting mechanism following 18 treatment of partially hepatectomized male Osborne-Mendel rats with a single 100 mg/kg gavage 19 dose of 1,1,2,2-tetrachloroethane, followed by 7 weeks of promotion with phenobarbital in the 20 diet. This regimen failed to result in increased numbers of preneoplastic (GGT-positive) foci in 21 the liver; whereas an exposure of partially hepatectomized male Osborne-Mendel rats to a single 22 i.p. dose of diethylnitrosamine (DEN) as an initiating agent followed by promotion with 100 23 mg/kg-day of 1,1,2,2-tetrachloroethane by gavage 5 days/week for 7 weeks produced a 24 significantly increased number of GGT-positive foci in the liver.. 25 26 4.8. SUSCEPTIBLE POPULATIONS AND LIFE STAGES 27 4.8.1. Possible Childhood Susceptibility 28 Studies in humans and laboratory animals have not thoroughly examined the effect of 29 1,1,2,2-tetrachloroethane exposure on the immature organism. The Gulati rat study (Gulati et al., 30 1991b) demonstrated that fetuses exposed in utero can be adversely affected. At scheduled 31 sacrifice, average fetal weights were statistically significantly decreased in all dose groups 32 except the 34 mg/kg-day group. In the Gulati mouse study (Gulati et al., 199la), complete litter 33 resorption occurred in mice in 1/11, 0/9, 2/8, 1/1, and 1/2 dams in the 0, 987, 2,120, 2,216, and 34 4,575 mg/kg-day dose groups, respectively. The limited data evaluating the effect of 35 1,1,2,2-tetrachloroethane on the developing organism have not indicated effects on the offspring 36 at levels that did not also produce maternal effects. 37 38 4.8.2. Possible Gender Differences 74 DRAFT - DO NOT CITE OR QUOTE ------- 1 Studies evaluating the differences in potency of 1,1,2,2-tetrachloroethane in male and 2 female rodents are not available. Some toxicity studies which evaluated both genders in the 3 same study showed close concordance between genders with often no more than one dose 4 distinguishing between response levels for a given effect. Men normally have a smaller volume 5 of body fat than women, even accounting for average size differences, contributing to differential 6 disposition of organic solvents between genders (Sato and Nakajima, 1987). Rats have 7 pronounced sex-specific differences in CYPs, primarily involving the CYP2C family which is 8 not found in humans, but humans have not demonstrated sex-specific isoforms of CYP450 9 (Mugford and Kedderis, 1998). Humans have differences in CYP 3A4 activity related to 10 estrogen and progesterone, but these differences are regulated by the hormones at the level of 11 gene expression (Harris et al, 1995). Other differences may occur at the Phase 2 level 12 attributable to conjugation. Overall, no consistent differences have been reported between 13 women and men in the handling of xenobiotics such as 1,1,2,2-tetrachloroethane by CYP 14 isoforms (Shimada et al., 1994). These distinctions make it difficult to predict from the animal 15 data gender-relevant differences for human exposure to 1,1,2,2-tetrachloroethane. 16 17 4.8.3. Other Susceptible Populations 18 As metabolism is believed to play an important role in the toxicity of 1,1,2,2-tetrachloro- 19 ethane, particularly in the liver, individuals with elevated levels of cytochrome P450 enzymes 20 may have an increased susceptibility to the compound. Halpert (1982) reported an increase in in 21 vitro metabolite formation and in covalently bound metabolites following pretreatment with 22 xylene or phenobarbital, both of which increased cytochrome P450 activity. Sato et al. (1980) 23 similarly reported an increased metabolism of 1,1,2,2-tetrachloroethane in rats following ethanol 24 pretreatment. Since 1,1,2,2-tetrachloroethane has been demonstrated to inhibit cytochrome P450 25 enzymes (Paolini et al., 1992; Halpert, 1982), presumably through a suicide inhibition 26 mechanism, it is also possible that people coexposed to chemicals that are inactivated by 27 cytochrome P450 enzymes will be more susceptible to those compounds. 28 In addition, studies of human GST-zeta polymorphic variants show different enzymatic 29 activities toward and inhibition by dichloroacetic acid that could affect the metabolism of 30 1,1,2,2-tetrachloroethane (Lantum et al., 2002; Blackburn et al., 2001, 2000; Tzeng et al., 2000). 31 Dichloroacetic acid may covalently bind to GST-zeta (Anderson et al., 1999), irreversibly 32 inhibiting one of two stereochemically different conjugates, thus inhibiting its own metabolism 33 and leading to an increase in unmetabolized dichloroacetic acid as the dose and duration of 34 exposure increases (U.S. EPA, 2003). GST zeta is a hepatic enzyme that also functions in the 35 pathway for tyrosine catabolism. Populations, or single individuals, may be more sensitive to 36 1,1,2,2-tetrachloroethane toxicity depending on which GST-zeta variant they possess. 37 75 DRAFT - DO NOT CITE OR QUOTE ------- 1 5. DOSE-RESPONSE ASSESSMENTS 2 3 4 5.1. ORAL REFERENCE DOSE (RfD) 5 5.1.1. Subchronic Oral RfD 6 5.1.1.1. Choice of Principal Study and Critical Effect 1 The data available on subchronic oral exposure to 1,1,2,2-tetrachloroethane are limited to 8 experimental studies in animals. Though a number of case reports provide information on 9 effects of intentional acute oral exposure to lethal oral doses of 1,1,2,2-tetrachloroethane (Mant, 10 1953; Lilliman, 1949; Forbes, 1943; Elliot, 1933; Hepple, 1927), no subchronic studies of oral 11 exposure to 1,1,2,2-tetrachloroethane in humans exist. A single, well-designed 14-week 12 subchronic study in rats and mice that tested multiple dose levels and examined an array of 13 endpoints and tissues in rats is available (NTP, 2004). Furthermore, a developmental toxicity 14 study in rats and mice exists (Gulati et al, 1991a, b). These studies in laboratory animals 15 provide evidence suggesting that the liver and the developing fetus may be targets of toxicity 16 following subchronic oral exposure to 1,1,2,2-tetrachloroethane. 17 NTP reported multiple effects on the livers of both male and female rats and mice 18 following subchronic oral exposure to 1,1,2,2-tetrachloroethane. Specifically, NTP (2004) 19 exposed F344 rats (10/sex/group) to 0, 20, 40, 80, 170, or 320 mg/kg-day (both males and 20 females) and B6C3Fi mice (10/sex/group) to 0, 100, 200, 370, 700, or 1,360 mg/kg-day for 21 males and 0, 80, 160, 300, 600, or 1,400 mg/kg-day for females in the diet for 14 weeks. A 22 statistically significant decrease in body weight gain (<10%) in both male and female rats at 23 >80 mg/kg-day was observed. Low dose effects observed in the liver included statistically 24 significantly increased relative liver weights in both male and female rats at >40 mg/kg-day. In 25 addition, hepatocyte vacuolization was observed at >20 mg/kg-day in male rats and >40 mg/kg- 26 day in female rats. The severity of vacuolization was reported to be minimal to mild. Serum 27 enzyme activity levels of both male and female rats were also affected. For example, increases 28 in serum ALT and SDH activity were observed at >80 mg/kg-day in male rats and >170 mg/kg- 29 day in female rats. In addition, increased cholesterol levels and ALP activity were observed in 30 female rats at >80 and 170 mg/kg-day, respectively. Additional histopathology observed in the 31 liver included a statistically significantly increased incidence of minimal to moderate hepatocyte 32 hypertrophy at >170 mg/kg-day in females and >200 mg/kg-day in males. Also, increased 33 incidence of necrosis and pigmentation were observed at >80 mg/kg-day and hepatocellular 34 mitotic alterations and foci of cellular alterations were observed at >80 and >170 mg/kg-day in 35 male rats, respectively. In females, increased incidence of hepatocellular hypertrophy was 36 observed at >80 mg/kg-day and necrosis, pigmentation, and foci of cellular alterations were 37 reported at >170 mg/kg-day. Bile duct hyperplasia, increased bile acids, spleen pigmentation, 38 and spleen atrophy were also observed in both male and female rats at the two highest doses. 76 DRAFT - DO NOT CITE OR QUOTE ------- 1 Evidence of liver effects were also observed in mice by NTP (2004). A statistically 2 significant increase in relative liver weights was observed in both male and female mice at 3 >200 and 80 mg/kg-day, respectively. Increases in serum ALT and ALP activity, bile acids 4 levels, and hepatic 5'-nucleotidase activity (males only) were observed in males and females at 5 >370 and 160 mg/kg-day, respectively. The study authors also reported an increase in SDH 6 activity at >200 and 80 mg/kg-day in male and female mice, respectively. Serum cholesterol 7 levels were statistically significantly increased in female mice at >160 mg/kg-day. The 8 incidence of hepatocellular necrosis was statistically significantly increased in male mice at >370 9 mg/kg-day and in female mice at >700 mg/kg-day. Hepatocellular hypertrophy was also 10 reported in both genders at >160-200 mg/kg-day. A statistically significant increase in incidence 11 of liver pigmentation and bile duct hyperplasia occurred at >300 mg/kg-day in females and 12 >370 mg/kg-day in males. 13 In addition to effects on the liver, NTP (2004) also observed effects associated with 14 reproduction in adult rats and mice following subchronic exposure to 1,1,2,2-tetrachloroethane at 15 dose levels as low as 40 mg/kg-day. In male rats, sperm motility was decreased at >40 mg/kg- 16 day, and higher doses resulted in decreased epididymis weight and increased atrophy of the 17 preputial and prostate gland, seminal vesicle, and testicular germinal epithelium. In female rats, 18 minimal to mild uterine atrophy was increased at > 170 mg/kg-day and clitoral gland atrophy and 19 ovarian interstitial cell cytoplasmic alterations were increased at 320 mg/kg-day. Female F344 20 rats in the 170 mg/kg-day group also spent more time in diestrus compared to controls. Male 21 mice had increased incidences of preputial gland atrophy at >100 mg/kg-day. Less sensitive 22 effects included decreases in absolute testes weight (>700 mg/kg-day), absolute epididymis, and 23 cauda epididymis weights (1,360 mg/kg-day), and a decrease in epididymal spermatozoal 24 motility (1,360 mg/kg-day). The only noted reproductive toxicity parameter in female mice 25 affected was a significant increase in the length of the estrous cycle at a dose of 1,400 mg/kg-day. 26 A developmental toxicity study by Gulati et al. (1991 a) demonstrated that the developing 27 fetus may be sensitive to 1,1,2,2-tetrachloroethane exposure. Gulati et al. (1991 a) exposed 28 pregnant CD Sprague-Dawley rats to 0, 34, 98, 180, 278, or 330 mg/kg-day 29 1,1,2,2-tetrachloroethane from GDs 4 through 20. Small, but statistically significant, decreases 30 were observed in maternal body weight and average fetal weight at >98 mg/kg-day. No other 31 maternal or fetal effects were reported by the study authors. In a second study, Gulati et al. 32 (1991b) exposed pregnant Swiss CD-I mice to 0, 987, 2,120, 2,216, or 4,575 mg/kg-day 33 1,1,2,2-tetrachloroethane from GDs 4 through 17. All animals (9/9) in the high-dose group died 34 prior to the end of the study, precluding calculation of the average dose in this exposure group. 35 Maternal body weights were statistically significantly decreased compared to controls at 36 >2,120 mg/kg-day beginning on study day 9. Gross hepatic effects such as pale or grey and/or 37 enlarged livers and a prominent lobulated pattern were also reported in dams from all groups 38 except at the low dose. Complete litter resorption occurred in 1/11, 0/9, 2/8, 1/1, and 1/2 dams in 77 DRAFT - DO NOT CITE OR QUOTE ------- 1 the 0, 987, 2,120, 2,216, and 4,575 mg/kg-day groups, respectively. No other developmental 2 effects were reported. Gulati et al. (1991a, b) suggested that the developing fetus may be a target 3 of 1,1,2,2-tetrachloroethane-induced toxicity. However, these developmental studies were 4 conducted at doses higher than the subchronic NTP (2004) study, which demonstrated liver 5 effects at lower doses. Therefore, Gulati et al. (1991a, b) was not selected as the principal study 6 and the observed reproductive effects were not selected as the critical effect following 7 subchronic exposure to 1,1,2,2-tetrachloroethane. Nevertheless, potential points of departure 8 (PODs) based on the observed developmental effects from Gulati et al. (1991a) were provided 9 for comparison (see Section 5.1.2 and Appendix B). 10 In consideration of the available studies reporting effects of subchronic oral exposure to 11 1,1,2,2-tetrachloroethane in animals, NTP (2004) was chosen as the principal study for the 12 derivation of the subchronic RfD. This study was conducted in both genders of two species, 13 used five dose levels and a concurrent control group, measured a wide-range of endpoints and 14 tissues, and provides data that were transparently and completely reported. NTP (2004) 15 identified the liver as the most sensitive target organ of 1,1,2,2-tetrachloroethane-induced 16 toxicity. Specifically, NTP (2004) identified effects on the liver, including increased liver 17 weight and increased incidence of hepatocellular vacuolization, at low dose levels. Other liver 18 effects observed in rats and mice at higher doses included increased liver weight, increased ALT, 19 ALP, and SDH serum activity levels, increased bile acid levels, and an increased incidence of 20 hepatocellular vacuolization and necrosis. 21 Based on the available data from the NTP (2004) study, the liver appears to be the most 22 sensitive target organ for 1,1,2,2-tetrachloroethane-induced toxicity. Thus, the observed effects 23 in the liver were considered in the selection of the critical effect for the derivation of the 24 subchronic RfD. Specifically, liver effects including increased liver weight, increased ALT, 25 ALP, and SDH serum levels, increased bile acid levels, and an increased incidence of 26 hepatocellular vacuolization were taken into consideration and modeled for the determination of 27 the critical effect and POD (Section 5.1.1.2 and Appendix B). EPA selected increased liver 28 weight as the critical effect because this effect may represent a sensitive endpoint that occurs 29 early in the process leading to hepatocellular necrosis associated with subchronic oral exposure 30 to 1,1,2,2-tetrachloroethane. The increase in relative liver weight was selected as the basis for 31 the selection of the POD because this analysis takes into account the substantive, dose-dependent 32 decreases in body weight that were observed in both genders of rats. Rats were selected as the 33 representative species because they appeared to be more sensitive than mice to the hepatotoxic 34 effects of 1,1,2,2-tetrachloroethane. EPA recognizes that the POD for the increased incidence of 35 hepatocellular vacuolization is approximately an order of magnitude lower than the POD for 36 increased relative liver weight, and would result in a lower RfD than that derived for increased 37 relative liver weight (See Sections 5.1.1.2 and 5.1.3 for more information). However, the 78 DRAFT - DO NOT CITE OR QUOTE ------- 1 biological significance of this effect following 1,1,2,2-tetrachloroethane exposure is unclear 2 based on the following considerations. 3 Vacuoles are defined as cavities bound by a single membrane that serve several 4 functions; usually providing storage areas for fat, glycogen, secretion precursors, liquid, or debris 5 (Osol, 1972). Vacuolization is defined as the process of accumulating vacuoles in a cell or the 6 state of accumulated vacuoles (Grasso, 2002). This process can be classified as either a normal 7 physiological response or may reflect an early toxicological process. As a normal physiological 8 response, vacuolization is associated with the sequestration of materials and fluids taken up by 9 cells, and also with secretion and digestion of cellular products (Henics and Wheatley, 1999). In 10 addition, Robbins et al. (1976) characterized vacuolization (i.e., intracellular autophagy) as a 11 normal cellular functional, homeostatic, and adaptive response. 12 Vacuolization is not only a normal physiological response. Vacuolization has been 13 identified as one of four principal types of chemical-induced injury (the other three being cloudy 14 swelling, hydropic change, and fatty change) (Grasso, 2002). It is one of the most common 15 responses of the liver following a chemical exposure, typically in the accumulation of fat in 16 parenchymal cells, most often in the periportal zone (Plaa and Hewitt, 1998). The ability to 17 detect subtle ultrastructural defects, such as vacuolization, early in the course of toxicity often 18 permits identification of the initial site of the lesion and thus can provide clues to possible 19 biochemical mechanisms involved in the pathogenesis of liver injury (Hayes, 2001). 20 The hepatocellular vacuolization reported by NTP (2004) was not observed consistently 21 across species (i.e., reported only in male and female rats); whereas the other observed liver 22 effects were reported in both sexes of both species. In addition, NTP (2004) did not characterize 23 the vacuole content following exposure to 1,1,2,2-tetrachloroethane. The study authors indicated 24 that the severity of the hepatocellular vacuolization was minimal to mild and was concentration 25 independent, but NTP (2004) did not report the localization of the vacuolization in the liver. The 26 observed vacuolization in the liver at low doses appeared to diminish as dose increased. 27 Specifically, hepatocellular vacuolization increased in a dose dependant manner from 20 to 28 80 mg/kg-day in male rats. At 80 mg/kg-day, 100% of male rats were affected, and at doses of 29 >80 mg/kg-day, the incidence of vacuolization began to decrease. Concurrent with this decrease 30 in incidence of vacuolization, an increased incidence of hepatocyte hypertrophy, necrosis, and 31 pigmentation were observed. In female rats, the incidence of vacuolization was 100% at 40 and 32 80 mg/kg-day followed by a diminished response at the two highest doses. Necrosis and 33 pigmentation were observed in the females at the two high doses. Thus, the qualitative and 34 quantitative biological relationship between the observed hepatocellular toxicity (i.e., hepato- 35 cellular necrosis) and the increased incidence of hepatocellular cytoplasmic vacuolization in 36 NTP (2004) is unknown. 37 38 5.1.1.2. Methods of Analysis—Including Models (PBPK, BMD, etc.) 79 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 9 10 11 12 13 14 15 Benchmark dose (BMD) modeling was conducted using the EPA's benchmark dose software (BMDS, version 2.1.1.) to analyze the hepatotoxic effects associated with subchronic exposure to 1,1,2,2-tetrachloroethane (see Appendix B for modeling details). The software was used to calculate potential PODs for deriving the subchronic RfD by estimating the effective dose at a specified level of response (BMDX) and its 95% lower bound (BMDLX). For all continuous endpoints, a BMR of 1SD of the control mean was considered appropriate for derivation of the RfD under the assumption that it represents a minimally biologically significant response level. A BMR of 1 standard deviation (SD) of the control mean was also included for comparative purposes. For the dichotomous data, i.e., the incidence of hepatocellular cytoplasmic vacuolization, a BMR of 10% extra risk was considered appropriate for derivation of the RfD under the assumption that it represents a minimally biologically significant response level. The effects modeled include liver weight changes, serum ALT and SDH, bile acids, hepatocellular cytoplasmic vacuolization, and rat fetal body weights. Table 5-1 summarizes the BMD modeling results for the selected toxicological endpoints. Table 5-1. Summary of BMD model results for rats exposed to 1,1^2,2-tetra- chloroethane Endpoint Model BMR BMD (mg/kg-d) BMDL (mg/kg-d) Males Cytoplasmic vacuol. Relative liver weight Absolute live weight ALT SDH Bile acids Polynomial None Polynomial Polynomial None Power 10% extra risk NA 1 SD 1 SD NA 1 SD 13 NA 30 41 NA 72 11 NA 23 26 NA 57 Females Cytoplasmic vacuol. Relative liver weight Absolute liver weight ALT SDH Bile acids Weibull Polynomial Polynomial Hill Power Polynomial 10% extra risk 1 SD 1 SD 1 SD 1 SD 1 SD 31 22 36 82 157 188 19 15 26 69 113 170 Developmental Rat fetal weight Linear 1 SD 83 60 16 17 18 19 20 21 Changes in hepatocellular cytoplasmic vacuolization, ALT, SDH, ALP, and bile acids serum levels from NTP (2004), as well as mean rat fetal weights from Gulati et al. (1991a), were modeled for comparison in identifying a POD. A BMD of 31 mg/kg-day and BMDL of 19 mg/kg-day were derived from the multistage model for the increased incidence of hepatocellular cytoplasmic vacuolization in female rats. For serum ALT levels in female rats, a BMD of 82 80 DRAFT - DO NOT CITE OR QUOTE ------- 1 mg/kg-day and a BMDL of 69 mg/kg-day was derived from the Hill model. For serum SDH in 2 female rats, a BMD of 157 mg/kg-day and a BMDL of 113 mg/kg-day was derived from the 3 power model. The serum ALP data were not amenable to BMD modeling; a LOAEL of 160 4 mg/kg-day was identified. For bile acid levels in female rats, a BMD of 188 mg/kg-day and a 5 BMDL of 170 mg/kg-day were derived from the polynomial model. BMD modeling derived a 6 BMD of 83 mg/kg-day and a BMDL of 60 mg/kg-day from a linear model with a BMR of 1 SD 7 for decreased rat fetal weight. 8 The BMDiso of 22 mg/kg-day and BMDLiso of 15 mg/kg-day based on increased 9 relative liver weight in the female rat was selected as the POD for the subchronic RfD. The 10 observed changes in liver weights, serum liver enzyme levels, and hepatocellular necrosis 11 combine to support hepatotoxicity as the major toxic effect following 1,1,2,2-tetrachloroethane 12 exposure. 13 14 5.1.1.3. RfD Derivation—Including Application of Uncertainty Factors (UFs) 15 To derive the subchronic RfD, the 15 mg/kg-day BMDLiso for increased relative liver 16 weight in female rats is divided by a total UF of 300. The UF of 300 comprises component 17 factors of 10 for interspecies extrapolation, 10 for interhuman variability, and 3 for database 18 deficiencies. 19 A default UF of 10 was selected to account for the interspecies variability in 20 extrapolating from laboratory animals (rats) to humans (i.e., interspecies variability), because 21 information was not available to quantitatively assess toxicokinetic or toxicodynamic differences 22 between animals and humans for 1,1,2,2-tetrachloroethane. 23 A default UF of 10 was selected to account for inter-individual variability (UFH) to 24 account for human-to-human variability in susceptibility in the absence of quantitative 25 information to assess the toxicokinetics and toxicodynamics of 1,1,2,2-tetrachloroethane in 26 humans. However, studies of human GST-zeta polymorphic variants demonstrate different 27 enzymatic activities toward and inhibition by dichloroacetic acid that could affect the 28 metabolism of 1,1,2,2-tetrachloroethane (Lantum et al, 2002; Blackburn et al, 2001, 2000; 29 Tzeng et al., 2000). Populations, or single individuals, may be more sensitive to 1,1,2,2-tetra- 30 chloroethane toxicity depending on which GST-zeta variant they possess. Animal toxicity 31 studies did not show consistent sex-related differences. 32 An UF of 3 was selected to account for deficiencies in the database. The NTP (2004) 33 14-week study provides comprehensive evaluations of systemic toxicity and neurotoxicity in two 34 species. The NTP (2004) study provides information of effects on sperm, estrous cycle, and 35 male and female reproductive tissues in rats and mice, but the database lacks a two-generation 36 reproductive toxicity study. Available developmental toxicity studies provide information on 37 embryo or fetotoxicity in orally exposed rats and mice (Gulati et al., 1991a, b), but the studies 38 did not include skeletal and visceral examinations. 81 DRAFT - DO NOT CITE OR QUOTE ------- 1 An UF for LOAEL-to-NOAEL extrapolation was not used because the current approach 2 is to address this factor as one of the considerations in selecting a BMR for benchmark dose 3 modeling. In this case, a BMR associated with a change of 1 SD from the control mean was 4 selected under an assumption that it represents a minimal biologically significant change. 5 The subchronic RfD for 1,1,2,2-tetrachloroethane is calculated as follows: 6 7 Subchronic RfD = BMDLiso-UF 8 = 15 mg/kg-day-3 00 9 =0.05 mg/kg-day (or 5 x 10"2 mg/kg-day) 10 11 5.1.2. Chronic Oral RfD 12 5.1.2.1. Choice of Principal Study and Critical Effect - with Rationale and Justification 13 Information on the chronic oral toxicity of 1,1,2,2-tetrachloroethane is limited to a 14 78-week cancer bioassay in rats and mice that were exposed by gavage (NCI, 1978). 15 Interpretation of the rat study may be confounded by high incidences of endemic chronic murine 16 pneumonia, although it is unlikely that this contributed to effects observed in the liver. Based on 17 an increased incidence of hepatic fatty changes, the NOAEL and LOAEL for liver effects were 18 62 and 108 mg/kg-day, respectively. In the mouse study, a LOAEL of 142 mg/kg-day was 19 selected for chronic inflammation in the kidneys of males and a NOAEL of 142 mg/kg-day and a 20 LOAEL of 284 mg/kg-day were selected for hydronephrosis and chronic inflammation in the 21 kidneys of females, respectively. 22 The 14-week dietary study in rats and mice (NTP, 2004), used to derive the subchronic 23 RfD, was also considered for the derivation of the chronic RfD. The subchronic NTP (2004) 24 study appears to be a more sensitive assay than the chronic NCI (1978) bioassay. The NTP 25 (2004) study also uses lower dose levels and a wider dose range than the NCI (1978) study, and 26 thereby provides a better characterization of the dose-response curve in the low-dose region. 27 Additionally, dietary exposure is a more relevant route of exposure for the general population 28 exposed to 1,1,2,2-tetrachloroethane in the environment than is gavage exposure. For these 29 reasons, the NTP (2004) subchronic study was selected as the principal study. 30 EPA selected increased liver weight as the critical effect because this effect may 31 represent a potential sensitive endpoint that may occur early in the process leading to 32 hepatocellular necrosis associated with subchronic oral exposure to 1,1,2,2-tetrachloroethane. 33 The increase in relative liver weight was selected as the basis for the selection of the POD 34 because this analysis takes into account the substantive, dose-dependent decreases in body 35 weight that were observed in both sexes of rats. Additional liver effects observed included 36 increased liver weight, increased ALT, ALP, and SDH serum levels, increased serum bile acid 37 levels, and increased incidence of hepatocellular vacuolization and necrosis. 38 39 5.1.2.2. Methodsof Analysis—Including Models (PBPK, BMD, etc.) 82 DRAFT - DO NOT CITE OR QUOTE ------- 1 The subchronic BMDLiso of 15 mg/kg-day based on the increased relative liver weight 2 in female rats was used as the POD for the chronic RfD. The observed increases in liver weights, 3 serum liver enzyme levels, and incidence of hepatocellular necrosis combine to support 4 hepatotoxicity as the critical effect of toxicity of 1,1,2,2-tetrachloroethane. 5 6 5.1.2.3. RfD Derivation—Including Application of UFs 1 To derive the chronic RfD, the subchronic BMDLiso of 15 mg/kg-day, based on 8 increased relative liver weights in female rats, was divided by a UF of 1,000. The UF of 1,000 9 comprises component factors of 10 for interspecies extrapolation, 10 for interhuman variability, 10 3 for subchronic to chronic duration extrapolation, and 3 for database deficiencies, as explained 11 below. 12 A default UF of 10 was selected to account for the interspecies variability in 13 extrapolating from laboratory animals (rats) to humans (i.e., interspecies variability), because 14 information was not available to quantitatively assess toxicokinetic or toxicodynamic differences 15 between animals and humans for 1,1,2,2-tetrachloroethane. 16 A default UF of 10 was selected to account for inter-individual variability (UFn) to 17 account for human-to-human variability in susceptibility in the absence of quantitative 18 information to assess the toxicokinetics and toxicodynamics of 1,1,2,2-tetrachloroethane in 19 humans. However, studies of human GST-zeta polymorphic variants demonstrate different 20 enzymatic activities toward and inhibition by dichloroacetic acid that could affect the 21 metabolism of 1,1,2,2-tetrachloroethane (Lantum et al, 2002; Blackburn et al, 2001, 2000; 22 Tzeng et al., 2000). Populations, or single individuals, may be more sensitive to 1,1,2,2-tetra- 23 chloroethane toxicity depending on which GST-zeta variant they possess. Animal toxicity 24 studies which evaluated both sexes in the same study did not show consistent sex-related 25 differences. Developmental toxicity studies in animals are limited in scope, but have not 26 indicated effects on the offspring at levels that did not also cause maternal effects. 27 An UF of 3 was selected to account for extrapolation from a subchronic exposure 28 duration study to a chronic RfD. The study selected as the principal study was a 14-week study 29 by NTP (2004), a study duration that is minimally past the standard subchronic (90 day) study 30 and falls well short of a standard lifetime study. In addition, some data are available to inform 31 the nature and extent of effects that would be observed with a longer duration of exposure to 32 1,1,2,2-tetrachloroethane. Specifically, the available chronic cancer bioassay data (NCI, 1978) 33 suggest that liver damage observed in F344 rats following subchronic exposure to 1,1,2,2-tetra- 34 chloroethane (NTP, 2004), e.g., increased liver weight and incidence of necrosis, and altered 35 serum enzyme and bile levels, may not progress to more severe effects following chronic 36 exposures. The chronic cancer bioassay was conducted in Osborne-Mendel rats and did not 37 measure liver enzyme levels. However, NCI (1978) observed minimal alterations in liver 38 pathology, including inflammation, fatty metamorphosis, focal cellular change, and angiectasis 83 DRAFT - DO NOT CITE OR QUOTE ------- 1 in rats, and organized thrombus and nodular hyperplasia in mice. NCI (1978) reported that the 2 study authors performed complete histological analysis on the liver, but specific endpoints 3 assessed were not included. The available database does not abrogate all concern associated 4 with using a subchronic study as the basis of the RfD. For these reasons, a threefold UF was 5 used to account for the extrapolation from subchronic to chronic exposure duration for the 6 derivation of the chronic RfD. 7 An UF of 3 was selected to account for deficiencies in the database. The NTP (2004) 8 14-week study provides comprehensive evaluations of systemic toxicity and neurotoxicity in 9 both rats and mice. However, the database is limited by the lack of a two-generation 10 reproductive toxicity study. The NTP (2004) study provides information on effects on sperm, 11 estrous cycle, and male and female reproductive tissues in rats and mice, but the database lacks a 12 two-generation reproductive toxicity study. Available developmental toxicity studies provide 13 information on embryo or fetotoxicity in orally exposed rats and mice (Gulati et al., 199la, b), 14 but the studies did not include skeletal and visceral examinations. 15 An UF for LOAEL-to-NOAEL extrapolation was not used because the current approach 16 is to address this factor as one of the considerations in selecting a BMR for benchmark dose 17 modeling. In this case, a BMR associated with a change of 1 SD from the control mean was 18 selected under an assumption that it represents a minimal biologically significant change. 19 The chronic RfD for 1,1,2,2-tetrachloroethane is calculated as follows: 20 21 Chronic RfD = BMDLiSD-UF 22 =15 mg/kg-day - 1,000 23 = 0.015 mg/kg-day (or 1.5 x 10"2 mg/kg-day) 24 25 5.1.3. RfD Comparison Information 26 Figure 5-1 is an exposure-response array that presents NOAELs, LOAELs, and the dose 27 range tested corresponding to selected health effects. The effects observed in the subchronic and 28 chronic studies were considered candidates for the derivation of the sample subchronic and 29 chronic RfDs. 30 In addition to the increase in relative liver weight and the increased incidence of 31 hepatocellular cytoplasmic vacuolization, changes in absolute liver weight and serum levels of 32 ALT and SDH, bile acid levels, and serum cholesterol levels were considered for comparison. 33 Mean rat fetal weights observed following subchronic or chronic exposure to 1,1,2,2-tetrachloro- 34 ethane were also considered for comparison. Table 5-3 provides a tabular summary of sample 35 PODs and resulting subchronic sample RfDs for these endpoints in female rats. Additionally, 36 Figure 5-2 provides a graphical representation of this information. This figure should be 37 interpreted with caution since the PODs across studies are not necessarily comparable, nor is the 38 confidence the same in the data sets from which the PODs were derived. Figure 5-3 provides a 84 DRAFT - DO NOT CITE OR QUOTE ------- 1 graphical representation of the derivation of sample chronic RfDs for sample PODs from the 2 subchronic data. 3 85 DRAFT - DO NOT CITE OR QUOTE ------- 350 0> (A O O 1 < > > 1 I » LOAEL • NOAEL 1 TU., ,,„,+;„„! lines represent the range of doses tested in a given study. f I T T T T T T T T I 1-^f Iss s|I -- SQ: -~- ifs llo.- Is|^ l|o' = E .0 CM CD ^S = 05 CM <^'-- 051——. T=> — o CD co i _ o ' ~— <-, ^^^CD ^--cor^ o Q_.b! |— — _£= 0- .>i|— coJ«C3 CD^,-, to-o" cOcoQ- o««coo ca^-- OO"±;CD ro o o Z co °>H- ro^Z ^SP^ rorooi ro^l- 2^!~ ro^roOi CD ^^ r -^ E 2 ^ o-^^— -^^5, ^ra^ 2 S^ eroz ""5- 0.°^ oo-ro roS^o CDOOCO rofe C- — CO 0 ^X3 >— ^-. o ^X3- — - £= CO CD 0 o+ CD -Q 2 Q- CD -^^ro ? ra .c " s= ' -<=a> -o a^c' 1 2 Figure 5-1. Exposure response array for subchronic and chronic oral exposure to 1,1^2,2-tetrachloroethane. 86 DRAFT - DO NOT CITE OR QUOTE ------- Table 5-3. Potential PODs with applied UFs and resulting subchronic RfDs Effect Hepatocellular cytoplasmic vacuolization Relative liver weight Absolute liver weight ALT SDH Bile acids Fetal body weight POD (mg/kg-d) l.lb 15C 23C 26C 113C 57C 60d Gender and Species Male Rat Female Rat Male Rat Male Rat Female Rat Male Rat Rat UFsa A 10 10 10 10 10 10 10 H 10 10 10 10 10 10 10 L - - - - - - s - - - - - - D 3 3 3 3 3 3 3 Total 300 300 300 300 300 300 300 Subchronic RfD 4 x 10"3 5 x 10'2 8 x 10'2 9 x 10"2 0.38 0.20 0.20 1 2 aUFs: A = animal to human (interspecies); H = interindividual (intraspecies); L = LOAEL to NOAEL; S = subchronic-to-chronic duration; D = database deficiency. bPOD based on BMDL determined through BMD modeling of a 10% response; source: NTP (2004). °POD based on BMDL determined through BMD modeling of a 1 SD response; source: NTP (2004). dPOD based on BMDL determined through BMD modeling of a 5% response; source: Gulati et al. (1991a). 87 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 1000 100 10 (O T3 0.1 0.01 0.001 if If If POD UFA UFD RfD hepatocellular relative liver weight- absolute liver weight increased ALT-o increased SDH-? increased bile acids - decreased fetal body cytoplasnic ? rats (HIP, 2004) - o rats (NIP, 2004) rats (NTP, 2004) rats (NTP, 2004) o rats (NIP, 2004) weight-rats (Gulati vacuolization - o rats etal., 1991 a) (NTP, 2004) Figure 5-2. PODs for selected endpoints (with critical effect circled) from Table 5-3 with corresponding applied UFs and derived sample subchronic oral reference values (RfVs). DRAFT - DO NOT CITE OR QUOTE ------- 1000 100 10 ra _§> 1 "TO E 0.1 0.01 0.001 %\\\\V %\\\\V POD UFA UFH UFD UFsc RfD \\\\v \\\\v hepatocellular cytoplasmic vacuolization - <$ rats (NTP, 2004) relative liver weight- ? rats (NTP, 2004) absolute liver w eight - (5 rats (NTP, 2004) increased ALT- S rats (NTP, 2004) increased SDH - 2004) increased bile decreased fetal acids - (5 rats body w eight - (NTP, 2004) rats (Gulati et al., 1991 a) 1 2 3 Figure 5-3. PODs for selected endpoints (with critical effect circled) from Table 5-3 with corresponding applied UFs and derived sample chronic oral reference values (RfVs). 89 DRAFT - DO NOT CITE OR QUOTE ------- 1 5.1.4. Previous RfD Assessment 2 An oral assessment for 1,1,2,2-tetrachloroethane was not previously available on IRIS. 3 4 5.2. INHALATION REFERENCE CONCENTRATION (RfC) 5 5.2.1. Choice of Principal Study and Critical Effect—with Rationale and Justification 6 Information on the inhalation toxicity of 1,1,2,2-tetrachloroethane is limited. In Truffert 7 et al. (1977), rats were exposed to a presumed concentration of 560 ppm (3,909 mg/m3) for a 8 TWA duration of 5.1 hours/day, 5 days/week for 15 weeks. Findings included transient 9 histological alterations in the liver, including granular appearance and cytoplasmic vacuolation, 10 which were observed after 9 exposures and were no longer evident after 39 exposures. Because 11 of the uncertainty regarding the actual exposure concentration for the single dose, and a lack of 12 incidence and severity data, this report cannot be used to identify a NOAEL or LOAEL or for 13 possible derivation of an RfC. 14 Horiuchi et al. (1962) observed fatty degeneration of the liver and splenic congestion in a 15 single monkey exposed to a TWA of 1,974 ppm (15,560 mg/m3) 1,1,2,2-tetrachloroethane for 16 2 hours/day, 6 days/week for 9 months. The monkey was weak after approximately seven 17 exposures and had diarrhea and anorexia between the 12th and 15th exposures. Beginning at the 18 15th exposure, the monkey was "almost completely unconscious falling upon his side" for 20- 19 60 minutes after each exposure. Also, hematological parameters demonstrated sporadic changes 20 in hematocrit and RBC and WBC counts, but the significance of these findings cannot be 21 determined. This study cannot be utilized to identify a NOAEL or LOAEL due to the use of a 22 single test animal with no control group. 23 Mellon Institute of Industrial Research (1947) observed an increased incidence of lung 24 lesions and an increase in kidney weight in rats following a 6-month exposure to 200 ppm 25 1,1,2,2-tetrachloroethane, but these results were not evaluated because the control animals 26 experienced a high degree of pathological effects in the kidney, liver, and lung, and because of 27 the presence of an endemic lung infection in both controls and treated groups. MIIR (1947) also 28 observed increased serum phosphatase levels and blood urea nitrogen levels in a dog exposed to 29 200 ppm 1,1,2,2-tetrachloroethane, compared to control values, along with cloudy swelling of 30 the liver and the convoluted tubules of the kidney, and light congestion of the lungs. However, 31 identification of a LOAEL or NOAEL is precluded by poor study reporting, high mortality and 32 lung infection in the rats, and the use of a single treated animal in the dog study. 33 Kulinskaya and Verlinskaya (1972) observed inconsistent changes in acetylcholine levels 34 in Chinchilla rabbits exposed to 10 mg/m3 (1.5 ppm) 1,1,2,2-tetrachloroethane for 3 hours/day, 35 6 days/week for 7-8.5 months. A NOAEL or LOAEL was not identified because the changes in 36 acetylcholine were not consistent across time and incompletely quantified, and the biological 37 significance of the change is unclear. 90 DRAFT - DO NOT CITE OR QUOTE ------- 1 Shmuter (1977) observed increases in antibody levels in Chinchilla rabbits at 2 mg/m3 2 1,1,2,2-tetrachloroethane and decreases in antibody levels at 100 mg/m3. Exposure to 3 100 mg/m3 1,1,2,2-tetrachloroethane also resulted in a decrease in the relative content of 4 antibodies in the y-globulin fraction and an increase in the T and P fractions. This is a poorly 5 reported study that provides inadequate data, including reporting limitations, toxicological 6 uncertainty in the endpoints, and inconsistent patterns of response, which preclude the 7 identification of a NO AEL or LO AEL. 8 Effects following the chronic inhalation toxicity of 1,1,2,2-tetrachloroethane included 9 hematological alterations and increased liver fat content in rats exposed to 1.9 ppm (13.3 mg/m3) 10 4 hours/day for 265 days (Schmidt et al, 1972). Statistically significant changes included 11 increased leukocyte (89%) and pVglobulin (12%) levels compared to controls after 110 days, 12 and an increased percentage of segmented nucleated neutrophils (36%), decreased percentage of 13 lymphocytes (17%), and increased liver total fat content (34%) after 265 days. A statistically 14 significant decrease in y-globulin levels (32%) at 60 days postexposure and a decrease in adrenal 15 ascorbic acid content (a measure of pituitary ACTH activity) were observed at all three time 16 periods (64, 21, and 13%, respectively). This study is insufficient for identification of a NO AEL 17 or LOAEL for systemic toxicity because most of the observed effects occurred at a single dose or 18 time point, or there was a reversal of the effect at the next dose or time point. A reproductive 19 assessment in the Schmidt et al. (1972) study was sufficient for identification of a NO AEL for 20 the single dose tested, 1.9 ppm (13.3 mg/m ), for reproductive effects in male rats, including 21 percentage of mated females having offspring, littering interval, time to 50% littered, total 22 number of pups, pups per litter, average birth weight, postnatal survival on days 1, 2, 7, 14, 21, 23 and 84, sex ratio, and average body weight on postnatal day 84. However, macroscopic 24 malformations or significant group differences in the other indices were not observed at 25 13.3 mg/m3. The lack of information on the reproductive toxicity precludes utilizing the selected 26 NO AEL in the derivation of the RfC. 27 In addition, effects of chronic exposure to 1,1,2,2-tetrachloroethane included alterations 28 in serum acetylcholinesterase activity in rabbits exposed to 1.5 ppm (10 mg/m ) 1,1,2,2-tetra- 29 chloroethane 3 hours/day, 6 days/week for 7-8.5 months (Kulinskaya and Verlinskaya, 1972) 30 and immunological alterations in rabbits exposed to 0.3-14.6 ppm (2-100 mg/m3) 3 hours/day, 31 6 days/week, for 8-10 months (Shmuter, 1977). These studies are inadequate for identification 32 of NOAELs or LOAELs for systemic toxicity due to inadequate study reporting. 33 The inhalation toxicity database lacks a well-conducted study that demonstrates a dose- 34 related toxicological effect following subchronic and/or chronic exposure to 1,1,2,2-tetrachloro- 35 ethane. Therefore, an inhalation RfC was not derived. 36 37 5.2.2. Methods of Analysis—Including Models (PBPK, BMD, etc.) 91 DRAFT - DO NOT CITE OR QUOTE ------- 1 A route-to-route extrapolation using the computational technique of Chiu and White 2 (2006), as described in Section 3.5, was considered. However, U.S. EPA (1994b) recommends 3 not conducting a route-to-route extrapolation from oral data when a first-pass effect by the liver 4 or respiratory tract is expected, or a potential for a portal-of-entry effect in the respiratory tract is 5 indicated following analysis of short-term inhalation, dermal irritation, in vitro studies, or 6 evaluation of the physical/chemical properties. In the case of 1,1,2,2-tetrachloroethane, a first- 7 pass effect by the liver is expected. In addition, the presence of tissue-bound metabolites in the 8 epithelial linings in the upper respiratory tract may demonstrate a first-pass effect by the 9 respiratory tract (Eriksson and Brittebo, 1991). Lehmann et al. (1936) observed irritation of the 10 mucous membranes of two humans following inhalation of 146 ppm (1,003 mg/m3) for 11 30 minutes or 336 ppm (2,308 mg/m ) for 10 minutes, indicating the potential for portal-of-entry 12 effects in the respiratory system. 13 14 5.2.3. Previous RfC Assessment 15 An inhalation assessment for 1,1,2,2-tetrachloroethane was not previously available on 16 IRIS. 17 18 5.3. UNCERTAINTIES IN THE ORAL REFERENCE DOSE (RfD) AND INHALATION 19 REFERENCE CONCENTRATION (RfC) 20 The following discussion identifies some uncertainties associated with the RfD for 21 1,1,2,2-tetrachloroethane. As presented earlier (Sections 5.1.2 and 5.1.3; 5.2.2 and 5.2.3), EPA 22 standard practices and RfC and RfD guidance (U.S. EPA, 1994b) were followed in applying an 23 UF approach to a POD, a BMDLiso for the subchronic and chronic RfDs. Factors accounting 24 for uncertainties associated with a number of steps in the analyses were adopted to account for 25 extrapolating from an animal bioassay to human exposure, a diverse human population of 26 varying susceptibilities, and to account for database deficiencies. These extrapolations are 27 carried out with standard approaches given the lack of extensive experimental and human data on 28 1,1,2,2-tetrachloroethane to inform individual steps. 29 An adequate range of animal toxicology data is available for the hazard assessment of 30 1,1,2,2-tetrachloroethane, as described in Section 4. Included in these studies are short-term and 31 long-term bioassays and a developmental toxicity bioassay in rats and mice, as well as numerous 32 supporting genotoxicity and metabolism studies. Toxicity associated with oral exposure to 33 1,1,2,2-tetrachloroethane is observed in the liver, kidney, and developing organism, including 34 decreased fetal body weight and increased number of litter resorptions. 35 Consideration of the available dose-response data to determine an estimate of oral 36 exposure that is likely to be without an appreciable risk of adverse health effects over a lifetime 37 led to the selection of the 14-week oral dietary study in rats (NTP, 2004) and increased relative 38 liver weight in females as the principal study and critical effect, respectively, for deriving the 92 DRAFT - DO NOT CITE OR QUOTE ------- 1 subchronic and chronic RfDs for 1,1,2,2-tetrachloroethane. The NTP (2004) data demonstrate 2 hepatocellular damage, including increased liver weight, increased serum liver enzyme levels, 3 and increased incidence of hepatic necrosis. Increased liver weight was chosen as the critical 4 effect because it may represent a sensitive indicator of 1,1,2,2,-tetrachloroethane-induced 5 hepatoxicity and occurs at a dose lower than the observed overt liver necrosis. The increase in 6 relative liver weight was selected as the basis for the selection of the POD because this analysis 7 takes into account the substantive, dose-dependent decreases in body weight that were observed 8 in both sexes of rats. The dose-response relationships between oral exposure to 1,1,2,2-tetra- 9 chloroethane and fetal body weight in rats and mice are also suitable for deriving an RfD, but are 10 associated with BMDLs that are less sensitive than the selected critical effect and corresponding 11 BMDL. 12 For comparison purposes, Figure 5-2 presents potential PODs, applied UFs, and derived 13 potential RfDs for the additional endpoints that were modeled using the EPA's BMDS, version 14 2.1.1. The additional endpoints included increased absolute liver weight, changes in serum ALT 15 and SDH, increased bile acids, and increased incidence of hepatocellular necrosis, all of which 16 support the liver as the target of 1,1,2,2-tetrachloroethane-induced toxicity following oral 17 exposure. A decrease in rat fetal weight was also modeled. The change in serum ALP was 18 modeled, but a model with adequate fit was not available. 19 The selection of the BMD model for the quantitation of the RfD does not lead to 20 significant uncertainty in estimating the POD, since benchmark effect levels were within the 21 range of experimental data. However, the selected model, the polynomial model, does not 22 represent all possible models one might fit, and other models could be selected to yield more 23 extreme results, both higher and lower than those included in this assessment. 24 Extrapolating from animals to humans embodies further issues and uncertainties. An 25 effect and its magnitude associated with the concentration at the POD in rodents are extrapolated 26 to human response. Pharmacokinetic models are useful in examining species differences in 27 pharmacokinetic processing, however, dosimetric adjustment using pharmacokinetic modeling 28 was not possible for the toxicity observed following oral and inhalation exposure to 1,1,2,2-tetra- 29 chloroethane. Additional interspecies uncertainty may arise from the rate of metabolism across 30 species, as it has been demonstrated that mice have greater metabolic capacity following 31 exposure to tetrachloroethylene than rats and humans (Reitz et al, 1996). Reitz et al. (1996) 32 demonstrated that mice possessed a greater relative ability to metabolize tetrachloroethylene than 33 rats and humans, and, although data are not available, a similar situation may exist for 1,1,2,2- 34 tetrachloroethane. 35 Heterogeneity among humans is another uncertainty associated with extrapolating from 36 animals to humans. Uncertainty related to human variation needs to be considered; also, 37 uncertainties in extrapolating from a subpopulation, say of one sex or a narrow range of life 38 stages typical of occupational epidemiologic studies, to a larger, more diverse population need to 93 DRAFT - DO NOT CITE OR QUOTE ------- 1 be addressed. In the absence of 1,1,2,2-tetrachloroethane-specific data on human variation, a 2 factor of 10 was used to account for uncertainty associated with human variation in the 3 derivation of the RfD. Human variation may be larger or smaller; however, 1,1,2,2-tetrachloro- 4 ethane-specific data to examine the potential magnitude of over- or under-estimation are 5 unavailable. 6 Extrapolating from subchronic PODs to derive chronic reference values is also an 7 uncertainty encountered in this assessment. A threefold UF was selected to account for 8 extrapolation from a subchronic exposure duration study to a chronic RfD. Based on the 9 available data for 1,1,2,2-tetrachloroethane, the toxicity observed in the liver does not appear to 10 increase over time. The use of data from a subchronic study to derive a chronic RfD becomes a 11 concern when the damage, in this case hepatoxicity, has the potential to accumulate; however, if 12 the progression of the effect is not apparent, a reduced UF may be considered (U.S. EPA, 1994b). 13 Specifically, liver damage observed in F344 rats following subchronic exposure to 1,1,2,2-tetra- 14 chloroethane (NTP, 2004), e.g., increased incidence of necrosis or altered serum enzyme and bile 15 levels, did not progress to more severe effects such as cirrhosis or major liver disease following 16 chronic exposures (NCI, 1978). NCI (1978) observed minimal alterations in liver pathology, 17 including inflammation, fatty metamorphosis, focal cellular change, and angiectasis in rats, and 18 organized thrombus and nodular hyperplasia in mice. Therefore, the available database does not 19 abrogate all concern associated with using a subchronic study as the basis of the RfD, but 20 supports the utilization of a database UF of 3. 21 Data gaps have been identified that are associated with uncertainties in database 22 deficiencies specific to the developmental and reproductive toxicity of 1,1,2,2-tetrachloroethane 23 following oral exposure. The developing fetus may be a target of toxicity, and the absence of a 24 study specifically evaluating the full range of developmental toxicity endpoints represents an 25 area of uncertainty or gap in the database. The database of inhalation studies is of particular 26 concern due to the paucity of studies, especially subchronic and chronic studies, a multi- 27 generational reproductive study, and a developmental toxicity study. 28 29 5.4. CANCER ASSESSMENT 30 As discussed in Section 4.7, under U.S. EPA's Guidelines for Carcinogen Risk 31 Assessment (U.S. EPA, 2005a), 1,1,2,2-tetrachloroethane is "likely to be carcinogenic to 32 humans" based on data from an oral cancer bioassay in male and female Osborne-Mendel rats 33 and B6C3Fi mice (NCI, 1978) demonstrating an increase in the incidence of hepatocellular 34 carcinomas in both sexes of mice. In this study, the incidence of hepatocellular carcinomas was 35 statistically significantly increased in both sexes of B6C3Fi mice at 142 (13/50 males; 30/48 36 females) and 284 mg/kg-day (44/49 males; 43/47 females), with incidences in the male and 37 female controls of 3/36 and 1/40, respectively. NCI (1978) also demonstrated a decrease in the 38 time to tumor in both sexes of mice. Male rats exhibited an increased incidence in hepatocellular 94 DRAFT - DO NOT CITE OR QUOTE ------- 1 carcinomas, characterized as rare tumors, but the increased incidence was not statistically 2 significantly different from controls. NCI (1978) has characterized the carcinogenic results in 3 male rats as "equivocal." 4 The epidemiological human data available are inadequate for evaluation for cancer risk 5 (IARC, 1999). There are a limited number of positive results from genotoxicity studies which 6 suggest that 1,1,2,2-tetrachloroethane treatment in animals can result in UDS (Miyagawa et al., 7 1995), chromosomal aberrations (McGregor, 1980), SCE (NTP, 2004; Colacci et al., 1992), and 8 micronucleus formation (NTP, 2004). The ability of 1,1,2,2,-tetrachloroethane to alkylate 9 enzymatically purified hepatic DNA was observed following a single oral dose of 150 mg/kg of 10 1,1,2,2-tetrachloroethane in B6C3Fi mice (Dow Chemical Company, 1988). 1,1,2,2-Tetra- 11 chloroethane may have tumor initiating and promoting activity in mammalian cells (Colacci et 12 al., 1996, 1992; Milrnan et al., 1988; Story et al., 1986). 13 14 5.4.1. Choice of Study/Data—with Rationale and Justification 15 The only carcinogenicity bioassay for 1,1,2,2-tetrachloroethane is a chronic gavage study 16 in Osborne-Mendel rats and B6C3Fi mice performed by NCI (1978). This study was conducted 17 in both sexes in two species with an adequate number of animals per dose group, with 18 examination of appropriate toxicological endpoints in both sexes of rats and mice. Selection of 19 doses was aided by range-finding toxicity tests. The rat study did not identify statistically 20 significant increases in tumor incidences in males or females. Three rare liver tumors in high- 21 dose male rats were noted. 22 The mouse study identified statistically significant, dose-related increases in the 23 incidences of hepatocellular carcinomas in both sexes. Based on these increases in 24 hepatocellular carcinomas, NCI (1978) concluded that orally administered 1,1,2,2-tetrachloro- 25 ethane is a liver carcinogen in male and female B6C3Fi mice. NCI (1978) stated that there was 26 no evidence for carcinogenicity of 1,1,2,2-tetrachloroethane in Osborne-Mendel rats (NCI, 1978). 27 The tumor data in mice from the NCI study was used for dose-response analysis for oral 28 exposure. 29 30 5.4.2. Dose-response Data 31 Data on the incidences of hepatocellular carcinomas in male and female mice from the 32 NCI (1978) study were used for cancer dose-response assessment. These data are shown in 33 Table 5-4. The control data were pooled from vehicle control groups. The cancer bioassay for 34 1,1,2,2-tetrachloroethane demonstrated evidence of increased incidence of tumors in both sexes 35 of one species. 36 95 DRAFT - DO NOT CITE OR QUOTE ------- Table 5-4. Incidences of hepatocellular carcinomas in B6C3Fi mice used for dose-response assessment of 1,1^2,2-tetrachloroethane Sex Male Female Dose (mg/kg-d)a 0 3/36b l/40b 142 13/50 30/48 284 44/49 43/47 aTWA dose administered by gavage on 5 d/wk for 78 wks. Pooled vehicle (corn oil) control groups from this and another, concurrent, bioassay. Pooling based on identical housing and care, similar spontaneous tumor rates, placed on test at about the same time, and examined by the same pathologists. Source: NCI (1978). 1 2 5.4.3. Dose Adjustments and Extrapolation Method(s) 3 Conversion of the doses in the NCI (1978) mouse study to human equivalent doses 4 (HEDs) to be used for dose-response modeling was accomplished in three steps. The mice were 5 treated with 1,1,2,2-tetrachloroethane by gavage 5 days/week for 78 weeks and then observed 6 untreated for 12 weeks for a total study duration of 90 weeks. Because the reported TWA doses 7 were for a 5 day/week, 78 week exposure, they were duration-adjusted to account for the partial 8 week exposure (by multiplying by 5 days/7 days) and untreated observation period (by 9 multiplying by 78 weeks/90 weeks). These duration-adjusted animal doses were then converted 10 to HEDs by adjusting for differences in body weight and lifespan between humans and mice. In 11 accordance with the U.S. EPA (2005a) Guidelines for Carcinogen Risk Assessment, a factor of 12 BW3/4 was used for cross-species scaling. Because the study duration (90 weeks) was less than 13 the animal lifespan (104 weeks), the scaled dose was then multiplied by the cubed ratio of 14 experimental duration to animal lifespan to complete the extrapolation to a lifetime exposure in 15 humans. The equation and data used to calculate the HEDs are presented below, and the 16 calculated HEDs are presented in Table 5-5. 17 18 HED = Dose* x (W/70 kg)1/4 x (Le/L)3 19 Where: 20 Dose = average daily animal dose (* TWA converted for 5/7 days, 78/90 weeks) 21 W = average animal body weight (0.030 kg for male and female B6C3Fi mice [U.S. EPA, 22 1988]). 23 70 kg = reference human body weight (U. S. EPA, 1988) 24 Le = duration of experiment (90 weeks) 25 L = reference mouse lifespan (104 weeks) (U.S. EPA, 1988) 26 96 DRAFT - DO NOT CITE OR QUOTE ------- Table 5-5. HEDs corresponding to duration-adjusted TWA doses in mice Duration-adjusted dose in male and female mice (mg/kg-d) HED for use with both male and female mouse incidence data (mg/kg-d) Dose (mg/kg-d) 0 0 87.9 8.22 175.8 16.5 1 2 The mode of action of 1,1,2,2-tetrachloroethane carcinogenicity is unknown. It appears 3 that metabolism to one or more active compounds is likely to play a role in the development of 4 the observed liver tumors, but insufficient data preclude proposing a specific mode of action. 5 Dichloroacetic acid, a metabolite of 1,1,2,2-tetrachloroethane, induces hepatocellular carcinomas 6 in male and female B6C3Fi mice and F344 rats. Trichloroethylene (NTP, 1990; NCI, 1976) and 7 tetrachloroethylene (NTP, 1996; NCI, 1977), also metabolites of 1,1,2,2-tetrachloroethane, have 8 also been shown to be hepatocarcinogens in rodents. 9 Results of genotoxicity and mutagenicity studies of 1,1,2,2-tetrachloroethane are mixed 10 and insufficient for informing whether 1,1,2,2-tetrachloroethane carcinogenicity is associated 11 with a mutagenic mode of action. Given that the mechanistic and other information available on 12 cancer risk from exposure to 1,1,2,2-tetrachloroethane is sparse and that the existing data do not 13 inform the mode of action of carcinogenicity, a linear low-dose extrapolation was conducted as a 14 default option for the derivation of the oral slope factor. 15 Dose-response modeling was performed to obtain a POD for quantitative assessment of 16 cancer risk. The data sets for hepatocellular carcinoma in both sexes of mice were modeled for 17 determination of the POD. In accordance with the U.S. EPA (2005a) cancer guidelines, the 18 BMDLio (lower bound on dose estimated to produce a 10% increase in tumor incidence over 19 background) was estimated by applying the multistage cancer model in the EPA's BMDS 20 (version 2.1.1.) for the dichotomous incidence data, and selecting the results of the model that 21 best characterizes the cancer incidences. The BMD modeling of the male mouse data did not 22 achieve adequate model fit for any of the dichotomous models; thus, a cancer slope factor was 23 not derived from the male data. The 1° multistage model was selected for the derivation of the 24 cancer slope factor from the female data because this model provided adequate model fit and the 25 lowest Akaike's Information Criterion (AIC) when compared to the results of the 2° multistage 26 model. In addition, the 2° multistage model had insufficient degrees of freedom to test the 27 goodness-of-fit. The BMDL of 0.65 mg/kg-day from the modeling of the tumor incidence data 28 in female mice is selected as the POD for use in calculation of an oral slope factor (Table 5-6). 29 Details of the BMD modeling are presented in Appendix C. 30 97 DRAFT - DO NOT CITE OR QUOTE ------- Table 5-6. Summary of human equivalent BMDs and BMDLs based on hepatocellular carcinoma incidence data in female B6C3Fi mice Female mice BMR (% extra risk) 10 HMD (mg/kg-d)a 0.81 BMDL10 (mg/kg-d)a 0.65 aHED. 1 2 5.4.4. Oral Slope Factor and Inhalation Unit Risk 3 The oral slope factor was derived from the BMDLio (the lower bound on the exposure 4 associated with a 10% extra cancer risk) by dividing the BMR by the BMDLio, and represents an 5 upper bound on cancer risk associated with a continuous lifetime exposure to 1,1,2,2-tetrachloro- 6 ethane. In accordance with the U.S. EPA (2005a) guidelines, an oral slope factor (mg/kg-day)"1 7 was calculated by dividing the human equivalent BMDLio into 0.1 (10%) (Appendix C). The 8 BMDLio, the lower 95% bound on exposure at 10% extra risk, is 0.65 mg/kg-day, and the cancer 9 slope factor, the slope of the linear extrapolation from the BMDLio to 0, is 0.10/0.65 = 0.15 per 10 mg/kg-day. The slope of the linear extrapolation from the central estimate (i.e., BMD) is 11 0.1/0.81 mg/kg-day or 0.12 (mg/kg-day)"1. 12 In the absence of any suitable data on the carcinogenicity of 1,1,2,2-tetrachloroethane via 13 the inhalation route, an inhalation unit risk has not been derived in this evaluation. 14 15 5.4.5. Uncertainties in Cancer Risk Values 16 Extrapolation of data from animals to estimate potential cancer risks to human 17 populations from exposure to 1,1,2,2-tetrachloroethane yields uncertainty. Several types of 18 uncertainties may be considered quantitatively, but other important uncertainties cannot be 19 considered quantitatively. Thus, an overall integrated quantitative uncertainty analysis is not 20 presented. This section and Table 5-7 summarize the principal uncertainties. 21 98 DRAFT - DO NOT CITE OR QUOTE ------- Table 5-7. Summary of uncertainty in the l,l?2,2-tetrachloroethane cancer risk assessment Consideration/ approach Low-dose extrapolation procedure Dose metric Cross-species scaling Statistical uncertainty at POD Bioassay Species/gender combination Human relevance of mouse tumor data Human population variability in metabolism and response/sensitive subpopulations Impact on oral slope factor Departure from U.S. EPA's Guidelines for Carcinogen Risk Assessment POD paradigm, if justified, could J, or 1 slope factor an unknown extent Alternatives could f or I slope factor by an unknown extent Alternatives could J, or 1 slope factor (e.g., 3. 5 -fold J, [scaling by BW] or t twofold (scaling by BW2/3]) 1 slope factor if MLE used rather than lower bound on POD Alternatives could f or J, slope factor by an unknown extent Human risk could J, or 1, depending on relative sensitivity Human relevance of mouse tumor data could I slope factor Low-dose risk f or J, to an unknown extent Decision Multistage cancer model to determine POD, linear low- dose extrapolation from POD Used administered exposure BW3/4 LEC (method for calculating reasonable upper bound slope factor) NCI study Female mice liver cancer Liver tumors in mice are relevant to human exposure Considered qualitatively Justification Available mode of action data do not inform selection of dose-response model; linear approach used in absence of an alternative as per U.S. EPA's Guidelines for Carcinogen Risk Assessment. Experimental evidence supports a role for metabolism in toxicity, but actual responsible metabolites are not clearly identified. There are no data to support alternatives. Because the dose metric was not an AUC, BW3 4 scaling was used to calculate equivalent cumulative exposures for estimating equivalent human risks. Limited size of bioassay results in sampling variability; lower bound is 95% confidence interval on administered exposure. Alternative bioassays were unavailable. There are no mode of action data to guide extrapolation approach for any choice. It was assumed that humans are as sensitive as the most sensitive rodent gender/species tested; true correspondence is unknown. The carcinogenic response occurs across species. Generally, direct site concordance is not assumed; consistent with this view, some human tumor types are not found in rodents and rat and mouse tumor types also differ. 1,1,2,2-tetrachloroethane is carcinogenic through an unknown mode of action. No data to support range of human variability/sensitivity, including whether children are more sensitive. Metabolic activation mode of action (if fully established) could indicate | or { early-life susceptibility. 1 2 3 4 5 6 Choice of low-dose extrapolation approach. The mode of action is a key consideration in clarifying how risks at low-dose exposures should be estimated. A linear low-dose extrapolation approach was used to estimate human carcinogenic risk associated with 1,1,2,2-tetrachloroethane exposure due to the unavailability of data that supports any specific mode of carcinogenic action for 1,1,2,2-tetrachloroethane. 99 DRAFT - DO NOT CITE OR QUOTE ------- 1 The extent to which the overall uncertainty in low-dose risk estimation could be reduced 2 if the mode of action for 1,1,2,2-tetrachloroethane were known is of interest, but data on the 3 mode of action of 1,1,2,2-tetrachloroethane are not available. 4 Dose metric. 1,1,2,2-Tetrachloroethane is metabolized to intermediates with 5 carcinogenic potential. Dichloroacetic acid is recognized as hepatocarcinogenic in male B6C3Fi 6 mice and F344 rats (U.S. EPA, 2003). However, it is unknown whether a metabolite or some 7 combination of parent compound and metabolites is responsible for the observed toxicity. If the 8 actual carcinogenic moiety is proportional to administered exposure, then use of administered 9 exposure as the dose metric is the least biased choice. On the other hand, if this is not the correct 10 dose metric, then the impact on the slope factor is unknown. 11 Cross-species scaling. An adjustment for cross-species scaling (BW3/4) was applied to 12 address toxicological equivalence of internal doses between the rodent species and humans, 13 consistent with the 2005 Guidelines for Carcinogen Risk Assessment (U.S. EPA, 2005a). It is 14 assumed that equal risks result from equivalent constant lifetime exposures. 15 Statistical uncertainty at the POD. Parameter, or probabilistic, uncertainty can be 16 assessed through confidence intervals. Each description of parameter uncertainty assumes that 17 the underlying model and associated assumptions are valid. For the multistage cancer model 18 applied to the female mice data, there is a reasonably small degree of uncertainty at a 10% 19 increase in tumor incidence (the POD for linear low-dose extrapolation). 20 Bioassay selection. The study by NCI (1978) was used for development of an oral slope 21 factor. This study was conducted in both sexes in two species with an adequate number of 22 animals per dose group, with examination of appropriate toxicological endpoints in both sexes of 23 rats and mice. Alternative bioassays were unavailable. Both genders of mice exhibited liver 24 tumors. Uncertainties associated with the use of this study in the derivation of the oral slope 25 factor arise, primarily, from the study design. The dose levels used in the study were poorly 26 selected and were modified over the exposure duration, and the exposure duration of the study 27 (78 weeks) was less then the standard 104 week chronic exposure duration. In addition, the bolus 28 nature of the 1,1,2,2-tetrachloroethane gavage exposures in NCI (1978) may lead to more 29 pronounced irritation, inflammation, cell death, and an eventual increase in tumor incidence at 30 portals of entry because of direct contact of the test chemical with the gastroinstestinal tissues. There 31 was also an increased incidence of endemic chronic murine pneumonia in male and female rats and 32 mice, and while interpretation of this study is complicated by the chronic murine pneumonia, it is 33 unlikely to have contributed to the carcinogenicity results observed in male and female rats. 34 Choice of species/gender. The oral slope factor for 1,1,2,2-tetrachloroethane was 35 quantified using the tumor incidence data for female mice. The hepatocelluar carcinoma data in 36 male mice demonstrated tumorigenicity, but the data in male mice did not achieve adequate 37 model fit for any of the dichotomous models when BMD modeled. The male and female rat 38 tumor incidence data were not suitable for deriving low-dose quantitative risk estimates, and NCI 100 DRAFT - DO NOT CITE OR QUOTE ------- 1 described the rat strain as relatively insensitive to the carcinogenic effects of chlorinated organic 2 compounds. 3 Relevance to humans. The oral slope factor is derived from the incidence of 4 hepatocellular carcinomas in female mice. Using liver tumors in B6C3Fi mice as the model for 5 human carcinogenesis is a concern because of the prevalence of and susceptibility to developing 6 liver tumors in this strain of mice. Hasemen et al. (1998) reported an increased liver carcinoma rate 7 of 17.9 and 8.4% for male and female B6C3F1 mice, respectively, from NTP carcinogenicity feeding 8 bioassays, and a combined adenoma and carcinoma rate of 42 and 24% for male and female B6C3F1 9 mice, respectively. The B6C3F1 mouse was also used in the NCI (1978) study and may be 10 excessively sensitive to the development of hepatocellular tumors. 11 Additional interspecies uncertainty may arise from the rate of metabolism across species, 12 as it has demonstrated that mice have greater metabolic capacity following exposure to 13 tetrachloroethylene than rats and humans (Reitz et al., 1996). Reitz et al. (1996) demonstrated 14 that mice possessed a greater relative ability to metabolize tetrachloroethylene than rats and 15 humans, and, although data are not available, a similar situation may exist for 1,1,2,2- 16 tetrachloroethane. 17 In addition, the genotoxicity and mutagenicity studies provide limited evidence of a 18 mutagenic mode of action, with 1,1,2,2-tetrachloroethane displaying equivocal results of 19 mutagenic activity. In addition, there are inadequate data to support any mode of action 20 hypothesis. 21 Human population variability. The extent of inter-individual variability in animals for 22 1,1,2,2-tetrachloroethane metabolism has not been characterized. A separate issue is that the 23 human variability in response to 1,1,2,2-tetrachloroethane is also unknown. This lack of 24 understanding about potential differences in metabolism and susceptibility across exposed 25 animal and human populations thus represents a source of uncertainty. 26 27 5.4.6. Previous Cancer Assessment 28 In the previous IRIS assessment, posted to the IRIS database in 1987, 1,1,2,2-tetrachloro- 29 ethane was characterized as "Classification — C; possible human carcinogen" based on the 30 increased incidence of hepatocellular carcinomas in mice observed in the NCI (1978) bioassay 31 (U.S. EPA, 1987). An oral slope factor of 0.2 (mg/kg-day)"1 was derived using the increased 32 incidence of hepatocellular carcinomas in female mice (NCI, 1978) and a linear multistage 33 extrapolation method. 34 101 DRAFT - DO NOT CITE OR QUOTE ------- 1 6. MAJOR CONCLUSIONS IN THE CHARACTERIZATION OF HAZARD AND DOSE 2 RESPONSE 3 4 5 6.1. HUMAN HAZARD POTENTIAL 6 1,1,2,2-Tetrachloroethane (CAS No. 79-34-5) has been used as an insecticide, fumigant, 7 and weed killer (Hawley, 1981), although it presently is not registered for any of these purposes. 8 It was once used as an ingredient in an insect repellent, but registration was canceled in the late 9 1970s. In the past, the major use for 1,1,2,2-tetrachloroethane was in the production of 10 trichloroethylene, tetrachloroethylene, and 1,2-dichloroethylene (Archer, 1979). It was also used 11 as a solvent, in cleaning and degreasing metals, in paint removers, varnishes, and lacquers, in 12 photographic films, and as an extractant for oils and fats (Hawley, 1981). With the development 13 of new processes for manufacturing chlorinated ethylenes, the production of 1,1,2,2-tetrachloro- 14 ethane as a commercial end-product in the United States and Canada had steadily declined since 15 the late 1960s and had ceased by the early 1990s (HSDB, 2009; Environment Canada and Health 16 Canada, 1993). 1,1,2,2-Tetrachloroethane may still appear as a chemical intermediate in the 17 production of a variety of other common chemicals. 18 1,1,2,2-Tetrachloroethane is well absorbed from the respiratory and gastrointestinal 19 tracts, is rapidly and extensively metabolized, and is eliminated mainly as metabolites in the 20 urine and breath. Both reductive and oxidative metabolisms occur, producing reactive radical 21 and organochlorine intermediates, respectively. Trichloroethanol, trichloroacetic acid, and 22 dichloroacetic acid are initial metabolites that subsequently yield glyoxalic acid, oxalic acid, and 23 carbon dioxide. 24 A limited amount of information is available addressing the toxicity of 1,1,2,2-tetra- 25 chloroethane in humans. CNS depression was the predominant effect of high-dose acute oral 26 and inhalation exposures, although acute inhalation also caused irritation of the mucous 27 membranes. Occupational studies suggest that repeated exposure to 1,1,2,2-tetrachloroethane 28 can affect the liver and the nervous system. 29 Animal studies have established that the CNS and liver are the main targets of toxicity at 30 high levels of oral and inhalation exposures. Death in laboratory animals typically was preceded 31 by signs of CNS depression (e.g., lethargy, incoordination, loss of reflexes, depressed 32 respiration, prostration, and loss of consciousness), and postmortem examinations mainly 33 showed fatty degeneration in the liver. The most sensitive target of sublethal ingestion and 34 inhalation appears to be the liver, and short-term and subchronic exposures caused hepatic 35 effects that included serum chemistry changes, hepatocellular degeneration, and other 36 histopathological alterations. Comprehensive neurobehavioral testing in 14-week feeding studies 37 showed no effects in rats or mice, indicating that the liver was more sensitive than the nervous 38 system for subchronic oral exposure (Chan, 2004). A limited amount of information is available 102 DRAFT - DO NOT CITE OR QUOTE ------- 1 on other effects of 1,1,2,2-tetrachloroethane. Reduced body weight gain and weight loss were 2 effects of repeated oral exposures in rats and mice that generally occurred at high doses and may 3 have contributed to mild anemia and atrophy in the spleen, bone, bone marrow, and reproductive 4 tissues in these animals. Kidney lesions (acute toxic tubular necrosis and chronic inflammation) 5 occurred in mice that were chronically exposed to oral doses that also caused reduced survival. 6 Adequate immunological testing of 1,1,2,2-tetrachloroethane has not been performed. 7 The reproductive and developmental toxicity of 1,1,2,2-tetrachloroethane has not been 8 adequately evaluated. Significant decreases in maternal and fetal body weights were observed in 9 rats. In mice, litter resorption was observed along with high maternal mortality. Toxicity to 10 reproductive tissues following 1,1,2,2-tetrachloroethane exposure to adult rats and mice was 11 observed in F344 rats and B6C3Fi mice. Effects observed in rats and/or mice include: 12 decreased sperm and spermatozoal motility; decreased testis and epididymal weight; increased 13 atrophy of the preputial and prostate gland, seminal vesicle, testicular germinal epithelium, 14 uterus, and clitoral gland; ovarian interstitial cell cytoplasmic alterations; and lengthened estrus 15 cycle. Chronic low-level inhalation caused no effects on reproductive function in male mice, but 16 multigeneration or other tests of reproductive function in females have not been conducted for 17 any route of exposure. Developmental toxicity was assessed in rats and mice that were 18 gestationally exposed to 1,1,2,2-tetrachloroethane in the diet. These studies did not include 19 examinations for skeletal or visceral abnormalities, although effects that included reduced fetal 20 body weight gain in rats and litter resorptions in mice occurred at doses that were maternally 21 toxic. 22 The carcinogenicity of 1,1,2,2-tetrachloroethane was evaluated in a chronic gavage study 23 in rats and mice conducted by NCI (1978). Hepatocellular carcinomas were induced in male and 24 female mice, but there were no statistically significant increases in tumor incidences in the rats. 25 Three rare tumors in high dose male rats were noted. Thus, 1,1,2,2-tetrachloroethane is "likely 26 to be carcinogenic to humans" by any route of exposure, according to the Guidelines for 27 Carcinogen Risk Assessment (U. S. EPA, 2005a). 28 29 6.2. DOSE RESPONSE 30 6.2.1. Noncancer/Oral 31 The NTP (2004) study was selected as the principal study because it was a well-designed 32 subchronic dietary study, conducted in both sexes in two rodent species with a sufficient number 33 of animals per dose group. The number of test animals allocated among three dose levels and an 34 untreated control group was acceptable, with examination of appropriate toxicological endpoints 35 in both sexes of rats and mice. The liver was the most sensitive target in both species and the 36 rats were more sensitive than the mice. In addition to the observed liver weight increases, there 37 is evidence of hepatocellular effects, including increased serum liver enzyme levels and an 38 increased incidence of both hepatocellular cytoplasmic vacuolization and necrosis, from the 103 DRAFT - DO NOT CITE OR QUOTE ------- 1 subchronic NTP (2004) study. EPA selected increased liver weight as the critical effect because 2 this effect may represent an indicator of liver toxicity that occurs early in the process leading to 3 hepatocellular necrosis associated with subchronic oral exposure to 1,1,2,2-tetrachloroethane. 4 Potential PODs for a subchronic RfD were derived by BMD modeling of dose-response 5 data for increases in liver weight, increases in serum levels of ALT, SDH, and ALP, increased 6 levels of bile acids, and increased incidence of hepatocellular cytoplasmic vacuolization in rats. 7 All available dichotomous models in the EPA's BMDS (version 2.1.1) were fit to the incidence 8 data for hepatocellular cytoplasmic vacuolization, and all available continuous models in the 9 software were applied to the data for liver weight and serum enzyme levels, as well as the data 10 for rat fetal body weight. A BMR of 10% (10% extra risk above control) was selected for 11 derivation of the BMDL for hepatocellular cytoplasmic vacuolization in female rats, and a BMR 12 of 1 SD (a change in the mean equal to 1 SD from the control mean) was selected for the 13 derivation of the BMDL for the continuous female rat liver weight and rat fetal body weight data. 14 The BMDiso of 22 mg/kg-day and BMDLiso of 15 mg/kg-day based on the relative liver 15 weight effects seen in the female rat represents a reasonable POD for the derivation of the RfD. 16 To derive the subchronic RfD, the 15 mg/kg-day BMDLiso based on female rat relative liver 17 weight was divided by a total UF of 300, yielding a subchronic RfD of 0.05 mg/kg-day. The UF 18 of 300 comprises component factors of 10 for interspecies extrapolation, 10 for interhuman 19 variability, and 3 for database deficiencies. 20 The choice of BMD model is not expected to introduce a considerable amount of 21 uncertainty in the risk assessment since the chosen response rate of 1 SD is within the observable 22 range of the data. Additional BMD modeling for other amenable data sets, including serum liver 23 enzyme levels, liver lesions, and fetal body weight, were also conducted to provide other PODs 24 for comparison purposes (see Appendix B). A graphical representation of these potential PODs 25 and resulting subchronic reference values is shown below in Figure 6-1. 104 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 1000 100 (O T3 0.1 0.01 0.001 If If POD UFA UFD RID hepatocellular relative liver weight- absolute liver weight increased ALT-o increased SDH-? increased bile acids - decreased fetal body cytoplasnic ? rats (HIP, 2004) - o rats (HIP, 2004) rats (NTP,2004) rats (NTP, 2004) o rats (NTP, 2004) weight-rats (Gulati vacuolization - o rats etal., 1991a) (NTP, 2004) Figure 6-1. PODs for selected endpoints (with critical effect circled) with corresponding applied UFs and derived sample subchronic oral RfVs. 105 DRAFT - DO NOT CITE OR QUOTE ------- 1 The default UF of 10 for the extrapolation from animals and humans is a composite of 2 uncertainty to account for toxicokinetic differences and toxicodynamic differences between the 3 animal species in which the POD was derived and humans. 4 PBTK models can be useful for the evaluation of interspecies toxicokinetics; however, 5 information was unavailable to quantitatively assess toxicokinetic or toxicodynamic differences 6 between animals and humans and the potential variability in human susceptibility; thus, the 7 interspecies and intraspecies UFs of 10 were applied for a total UF of 100. Human variation may 8 be larger or smaller; however, 1,1,2,2-tetrachloroethane-specific data to examine the potential 9 magnitude of human variability of response are unknown. 10 In addition, a threefold database UF was applied due to the lack of information 11 addressing the potential reproductive toxicity associated with 1,1,2,2-tetrachloroethane. 12 Uncertainties associated with data gaps in the 1,1,2,2-tetrachloroethane database have been 13 identified, specifically, uncertainties associated with database deficiencies characterizing 14 reproductive toxicity associated with oral exposure to 1,1,2,2-tetrachloroethane. The developing 15 fetus may be a target of toxicity (Gulati et al, 1991a), and the absence of a study specifically 16 evaluating the full range of developmental toxicity represents an additional area of uncertainty or 17 gap in the database. 18 The overall confidence in this subchronic RfD assessment is medium. Confidence in the 19 principal study (NTP, 2004) is high. Confidence in the database is medium. Reflecting high 20 confidence in the principal study and medium confidence in the database, confidence in the 21 subchronic RfD is medium. 22 Information on the chronic oral toxicity of 1,1,2,2-tetrachloroethane consists of a limited 23 78-week gavage study in rats and mice (NCI, 1978). The high incidences of hepatocellular 24 tumors in all treated groups of mice precluded evaluation of noncancer effects in the liver and 25 identification of a NOAEL or LOAEL. Additionally, the NCI (1978) study performed 26 histological examinations on the animals when they died or at the termination of the study, which 27 was beyond the point at which more sensitive hepatotoxic effects, including nonneoplastic 28 effects, would be expected. The 14-week dietary study (NTP, 2004) was used to derive the 29 subchronic oral RfD. The NTP (2004) study also utilized a more relevant type of exposure (i.e., 30 oral feeding) for the general population exposed to 1,1,2,2-tetrachloroethane in the environment. 31 The chronic RfD of 0.015 mg/kg-day was calculated by dividing the subchronic 32 BMDLiso of 15 mg/kg-day for increased relative liver weight by a total UF of 1,000: 10 for 33 interspecies extrapolation, 10 for interhuman variability, 3 for subchronic to chronic duration 34 extrapolation, and 3 for database deficiencies. 35 The choice of BMD model is not expected to introduce a considerable amount of 36 uncertainty in the risk assessment since the chosen BMR of 1 SD from the control mean is within 37 the observable range of the data. Additional BMD modeling for other amenable data sets, 38 including serum liver enzyme levels, liver lesions, and fetal body weight, were also conducted to 106 DRAFT - DO NOT CITE OR QUOTE ------- 1 provide other PODs for comparison purposes (see Appendix B). A graphical representation of 2 these potential PODs and resulting chronic reference values is shown below in Figure 6-2. 107 DRAFT - DO NOT CITE OR QUOTE ------- 1000 100 10 co 0.1 0.01 0.001 *\\\vo A\\\V — w 1 1 1 1 1 1 hepatocellular relative liver weight-absolute liver weight increased ALT -$ increased SDH-? increased bile acids - decreased fetal body cytoplasnic ? rats (NTP, 2004) - $ rats (NTP, 2004) rats (NTP, 2004) rats (NTP, 2004) $ rats (NTP, 2004) weight-rats (Gulati vacuolization - o rats etal., 1991 a) (NTP, 2004) POD UFA UFH UFD \\\\v \\\\v RfD I 2 3 Figure 6-2. PODs for selected endpoints (with critical effect circled) from Table 5-3 with corresponding applied UFs and derived sample subchronic oral RfVs. 108 DRAFT - DO NOT CITE OR QUOTE ------- 1 The default UF of 10 for the extrapolation from animals and humans is a composite of 2 uncertainty to account for toxicokinetic differences and toxicodynamic differences between the 3 animal species in which the POD was derived and humans. 4 PBTK models can be useful for the evaluation of interspecies toxicokinetics; however, 5 information was unavailable to quantitatively assess toxicokinetic or toxicodynamic differences 6 between animals and humans and the potential variability in human susceptibility, thus, the 7 interspecies and intraspecies UFs of 10 were applied for a total UF of 100. Human variation may 8 be larger or smaller; however, 1,1,2,2-tetrachloroethane-specific data to examine the potential 9 magnitude of human variability of response are unknown. 10 A threefold UF was applied for extrapolation from a subchronic exposure duration study 11 to a chronic RfD. Based on the available data for 1,1,2,2-tetrachloroethane, the toxicity observed 12 in the liver does not appear to increase over time. Specifically, liver damage observed in 13 F344 rats following subchronic exposure to 1,1,2,2-tetrachloroethane (NTP, 2004), e.g., 14 increased incidence of necrosis or altered serum enzyme and bile levels, did not progress to more 15 severe effects such as cirrhosis or major liver disease following chronic exposures (NCI, 1978). 16 Therefore, the available database does not abrogate all concern associated with using a 17 subchronic study as the basis of the RfD but supports the utilization of a database UF of 3. 18 In addition, a threefold database UF was applied due to the lack of information 19 addressing the potential reproductive toxicity associated with 1,1,2,2-tetrachloroethane. 20 Uncertainties associated with data gaps in the 1,1,2,2-tetrachloroethane database have been 21 identified, specifically, uncertainties associated with database deficiencies characterizing 22 reproductive toxicity associated with oral exposure to 1,1,2,2-tetrachloroethane. The developing 23 fetus may be a target of toxicity (Gulati et al, 199la), and the absence of a study specifically 24 evaluating the full range of developmental toxicity represents an additional area of uncertainty or 25 gap in the database. 26 The overall confidence in this chronic RfD assessment is medium. Confidence in the 27 principal study (NTP, 2004) is high. Confidence in the database is medium. Reflecting high 28 confidence in the principal study and medium confidence in the database, confidence in the 29 chronic RfD is medium. 30 31 6.2.2. Noncancer/Inhalation 32 An RfC was not calculated due to insufficient data. Information on the subchronic and 33 chronic inhalation toxicity of 1,1,2,2-tetrachloroethane is limited to the results of one study in 34 rats that found transient liver effects (Truffert et al., 1977). Reporting inadequacies preclude 35 identification of a NOAEL or LOAEL and derivation of an RfC in the usual manner. 36 A route-to-route extrapolation using the computational technique of Chiu and White 37 (2006), as described in Section 3.5, was considered. However, U.S. EPA (1994b) recommends 38 not conducting a route-to-route extrapolation from oral data when a first-pass effect by the liver 109 DRAFT - DO NOT CITE OR QUOTE ------- 1 or respiratory tract is expected, or a potential for portal-of-entry effects in the respiratory tract is 2 indicated following analysis of short-term inhalation, dermal irritation, in vitro studies, or 3 evaluation of the physical properties of the chemical. In the case of 1,1,2,2-tetrachloroethane, a 4 first-pass effect by the liver is expected. In addition, the presence of tissue-bound metabolites in 5 the epithelial linings in the upper respiratory tract may demonstrate a first-pass effect by the 6 respiratory tract (Eriksson and Brittebo, 1991). Lehmann et al. (1936) observed irritation of the 7 mucous membranes of two humans following exposure to 1,1,2,2-tetrachloroethane air 8 concentrations of 146 ppm (1,003 mg/m ) for 30 minutes or 336 ppm (2,308 mg/m ) for 9 10 minutes, indicating the potential for portal-of-entry effects in the respiratory system. 10 Information regarding the chronic inhalation toxicity of 1,1,2,2-tetrachloroethane is 11 available from four animal studies that include limited data on liver effects and serum 12 acetylcholinesterase, hematological, and immunological alterations (Shmuter, 1977; Kulinskaya 13 and Verlinskaya, 1972; Schmidt et al., 1972; Mellon Institute of Industrial Research, 1947). 14 However, the reporting of results from these chronic bioassays is inadequate for identification of 15 NOAELs or LOAELs for systemic toxicity. A chronic NOAEL was identified for reproductive 16 effects in male rats (Schmidt et al., 1972); however, macroscopic malformations or significant 17 group differences in the other indices were not observed at 13.3 mg/m3. This lack of information 18 on reproductive toxicity precludes utilizing this selected NOAEL in the derivation of an RfC. 19 20 6.2.3. Cancer/Oral and Inhalation 21 Under the Guidelines for Carcinogen Risk Assessment (U. S. EPA, 2005a), 1,1,2,2-tetra- 22 chloroethane is characterized as "likely to be carcinogenic to humans", based on the existence of 23 evidence of the compound's tumorigenicity in a single study in a single animal species (NCI, 24 1978) and the induction of hepatocellular carcinomas in both rats and mice by the main 25 metabolite, 1,2-dichloroacetic acid (U.S. EPA, 2003). The epidemiological human data available 26 are inadequate for evaluation of cancer risk (IARC, 1999). The NCI (1978) provided evidence 27 that 1,1,2,2-tetrachloroethane causes hepatocellular tumors in male and female mice. A few, 28 statistically nonsignificant, rare tumors were seen in high-dose male rats (NCI, 1978). The NCI 29 concluded that 1,1,2,2-tetrachloroethane causes cancer in mice. 30 The only carcinogenicity bioassay for 1,1,2,2-tetrachloroethane was a chronic gavage 31 study in Osborne-Mendel rats and B6C3Fi mice performed by NCI (1978). This was a well- 32 designed study, conducted in both sexes in two rodent species with an adequate number of 33 animals per dose group and with examination of appropriate toxicological endpoints in both 34 sexes of rats and mice. The rat study found no statistically significant increases in tumor 35 incidences in males or females. Three rare hepatocellular tumors in high-dose male rats were 36 noted and NCI (1978) characterized the carcinogenic results in male rats as "equivocal." The 37 mouse study found significant, dose-related increases in the incidences of hepatocellular 38 carcinomas in both sexes. Based on the increased incidences of hepatocellular carcinomas, NCI 110 DRAFT - DO NOT CITE OR QUOTE ------- 1 (1978) concluded that orally administered 1,1,2,2-tetrachloroethane is a liver carcinogen in male 2 and female B6C3Fi mice. This NCI study was used for dose-response analysis for oral exposure. 3 Data on the incidences of hepatocellular carcinomas in male and female mice from the 4 NCI (1978) study were used for cancer dose-response assessment. Conversion of the doses in 5 the NCI (1978) mouse study to HEDs to be used for dose-response modeling was accomplished 6 in two steps. The mice were treated with 1,1,2,2-tetrachloroethane by gavage 5 days/week for 7 78 weeks, and then observed untreated for 12 weeks for a total study duration of 90 weeks. 8 Because the reported TWA doses were doses for 5 days/week for 78 weeks, they were duration- 9 adjusted to account for the partial week exposure (by multiplying by 5 days/7 days) and 10 untreated observation period (by multiplying by 78 weeks/90 weeks). The duration-adjusted 11 animal doses were converted to HEDs by adjusting for differences in body weight and lifespan 12 between humans and mice. In accordance with U.S. EPA (2005a) Guidelines for Carcinogen 13 Risk Assessment, a factor of BW3 4 was used for cross-species scaling. Because the study 14 duration (90 weeks) was less than the animal lifespan (104 weeks), the scaled dose was then 15 multiplied by the cubed ratio of experimental duration to animal lifespan to complete the 16 extrapolation to a lifetime exposure in humans. 17 The mode of action of 1,1,2,2-tetrachloroethane carcinogenicity is unknown. It appears 18 that metabolism to one or more active compounds is likely to play a role in the development of 19 the observed liver tumors, but insufficient data preclude proposing this as a mode of action. 20 Results of genotoxicity and mutagenicity studies of 1,1,2,2-tetrachloroethane are mixed and 21 insufficient for informing the mode of action. Given that the mechanistic and other information 22 available on cancer risk from exposure to 1,1,2,2-tetrachloroethane is sparse and that the data 23 that does exist is equivocal, there is inadequate information to inform the low dose extrapolation. 24 Dose-response modeling was performed to obtain a POD for quantitative assessment of 25 cancer risk. The incidences of hepatocellular carcinomas in both sexes of mice were modeled for 26 determination of the POD. In accordance with the U.S. EPA (2005a) cancer guidelines, the 27 BMDLio (lower bound on dose estimated to produce a 10% increase in tumor incidence over 28 background) was estimated by applying the multistage cancer model in the the EPA's BMDS 29 (version 2.1.1) for the dichotomous incidence data and selecting the results for the model that 30 best fits the data. The BMD modeling of the male mouse data did not achieve adequate fit for 31 any of the dichotomous models; thus, a cancer slope factor was not derived from the male data. 32 The 1° multistage model was selected for the derivation of the cancer slope factor from the 33 female data because this model provided adequate model fit and the lowest AIC when compared 34 to the results of the 2° multistage model. In addition, the 2° multistage model had insufficient 35 degrees of freedom to test the goodness-of-fit. The BMDLio of 0.65 mg/kg-day from the 36 modeling of the tumor incidence data in female mice is selected as the POD for use in 37 calculation of an oral slope factor. Details of the BMD modeling are presented in Appendix C. 111 DRAFT - DO NOT CITE OR QUOTE ------- 1 In accordance with the U. S. EPA (2005a) guidelines, an oral slope factor of 0.15 (mg/kg- 2 day)"1 is calculated by dividing the human equivalent BMDLio of 0.65 mg/kg-day into 0.1 (10%) 3 (Appendix C). 4 In the absence of any data on the carcinogenicity of 1,1,2,2-tetrachloroethane via the 5 inhalation route, an inhalation unit risk has not been derived in this evaluation. 6 7 112 DRAFT - DO NOT CITE OR QUOTE ------- 7. REFERENCES Amoore, JE; Hautala E. (1983) Odor as an aid to chemical safety: odor thresholds compared with threshold limit values and volatilities for 214 industrial chemicals in air and water dilution. J Appl Toxicol 3(6):272-290. Anderson, WB; Board, PG; Gargano, B; et al. (1999) Inactivation of glutathione transferase zeta by dichloroacetic acid and other fluorine-lacking a-haloalkanoic acids. Chem Res Toxicol 12:1144-1149. Andrews, JE; Nichols, H; Hunter, ES. (2002) Developmental toxicity of di- and tetrachloroethane and dichloropropane in the rat whole embryo culture system. Toxicologist 66(1-S):23. Archer, WL. (1979) Other chloroethanes. In: Grayson, H; Eckroth, D; eds. Kirk-Othmer encyclopedia of chemical technology. New York, NY: John Wiley & Sons, 3rd edition, Vol 5, pp. 722-742. Arthur Little, Inc. (1983) Cell transformation assays of 11 chlorinated hydrocarbon analogs. Final report. ICAIR Work Assignment No. 10. EPA Document No. 40-8324457; NTIS No. OTS0509392. ASTER (Assessment Tools for the Evaluation of Risk). (1995) ASTER ecotoxicity profile. U.S. Environmental Protection Agency, Environmental Research Laboratory, Duluth, MN. AT SDR (Agency for Toxic Substances and Disease Registry). (1996) Toxicological profile for 1,1,2,2-tetrachloroethane (update). U.S. Department of Health and Human Services, Public Health Service. Blackburn, AC; Coggan, M; Tzeng, HF; et al. (2001) GSTZld: a new allele of glutathione transferase zeta and maleylacetoacetate isomerase. Pharmacogenetics 11:671-678. Blackburn, AC; Tzeng, HF; Anders, MW; et al. (2000) Discovery of a functional polymorphism in human glutathione transferase zeta by expressed sequence tag database analysis. Pharmacogenetics 10:49-57. Brem, H; Smith, AB; Rosenkranz, HS. (1974) The mutagenicity and DNB-modifying effect of haloalkanes. Cancer Res 34:2576-2579. Bull, RJ; Sanchez, IM; Nelson, MA; et al. (1990) Liver tumor induction in B6C3F1 mice by dichloroacetate and trichloroacetate. Toxicology 63:341-359. Callen, DF; Wolf, CR; Richard, MP. (1980) Cytochrome P-450 mediated genetic activity and cytotoxicity of seven halogenated aliphatic hydrocarbons in Saccharomyces cerevisiae. Mutat Res 77:55-63. Carpenter, CP; Smyth, HF, Pozzani, UC. (1949) The assay of acute vapor toxicity, and the grading and interpretation of results on 96 chemical compounds. J Ind Hyg Toxicol 31:343-346. CAS. (Chemical Abstracts Service). (1994). CA Registry File. Computer printout. Casciola, LAF; Ivanetich, KM. (1984) Metabolism of chloroethanes by rat liver nuclear cytochrome P-450. Carcinogenesis 5:543-548. Chan, PC. (2004) NTP technical report on the toxicity studies of 1,1,2,2-tetrachloroethane (CAS No. 79-34-5) administered in microcapsules in feed to F344/N rats and B6C3F1 mice. Toxic Rep Ser (49):6-Fl 1. Chiou, CT; Peters, LJ; Freed, VH. (1979) A physical concept of soil-water equilibria for nonionic organic compounds. Science 206:831-832. Chiu, WA; White, P. (2006) Steady-state solutions to PBPK models and their applications to risk assessment I: Route-to-route extrapolation of volatile chemicals. Risk Anal 26:769-780. Colacci, A; Grilli, S; Lattanzi, G; et al. (1987) The covalent binding of 1,1,2,2-tetrachloroethane to macromolecules of rat and mouse organs. Teratog Carcinog Mutagen 7:465-474. 113 DRAFT - DO NOT CITE OR QUOTE ------- Colacci, A; Perocco, P; Vaccari, M; et al. (1990) In vitro transformation of BALB/c 3T3 cells by 1,1,2,2-tetrachloroethane. Jpn J Cancer Res 81:786-792. Colacci, A; Perocco, P; Bartoli, S; et al. (1992) Initiating activity of 1,1,2,2-tetrachloroethane in two-stage BALB/c 3T3 cell transformation. Cancer Lett 64:145-153. Colacci, A; Vaccari, M; Perocco, P; et al. (1996) Enhancement of BALB/c 3T3 cells transformation by 1,2-dibromoethane promoting effect. Carcinogenesis 17:225-231. Cottalasso, D; Bellocchio, A; Domenicotti, C; et al. (1998) 1,1,2,2-Tetrachloroethane-induced early decrease of dolichol levels in rat liver microsomes and Golgi apparatus. J Toxicol Environ Health 54:133-144. Coyer, HA. (1944) Tetrachloroethane poisoning. Ind Med 13:230-233. Crebelli, R; Franekic, J; Conti, G; et al. (1988) Induction of chromosome malsegregation by halogenated organic solvents in Aspergillus nidulans: unspecific or specific mechanism? Mutat Res 201:401-411. Daniel, FB; DeAngelo, AB; Stober, JA; et al. (1992) Hepatocarcinogenicity of chloral hydrate, 2-chloroacetaldehyde, and dichloroacetic acid in male B6C3F1 mouse. Fundam Appl Toxicol 19:159-168. DeAngelo, AB; Daniel, FB; Stober, JA; et al. (1991) The carcinogenicity of dichloroacetic acid in the male B6C3F1 mouse. Fundam Appl Toxicol 16:337-347. DeAngelo, AB; Daniel, FB; Most, BM; et al. (1996) The carcinogenicity of dichloroacetic acid in the male Fischer 344 rat. Toxicology 114:207-221. DeAngelo, AB; George, MH; House, DE. (1999) Hepatocarcinogenicity in the male B6C3F1 mouse following a life-time exposure to dichloroacetic acid in the drinking water: dose-response determination and modes of action. J Toxicol Environ Health 58:485-507. Deguchi, T. (1972) A fundamental study of the threshold limit values for solvent mixtures in the air. OsakB-shiritsu Daigaku Igaku Zasshi 21:187-209. DeMarini, DM; Brooks, HG. (1992) Induction of prophage lambda by chlorinated organics: detection of some single-species/single-site carcinogens. Environ Mol Mutagen 19:98-111. Dow Chemical Company. (1988) The metabolism and hepatic micromolecular interactions of 1,1,2,2-tetrachloroethane (TCE) in mice and rats. Document D002628. Elliott, JM. (1933) Report of a fatal case of poisoning by tetrachloroethane. JR Army Med Corps 60:373-374 Environment Canada and Health Canada. (1993). Canada environmental protection act. Priority substances list assessment report. 1,1,2,2,-Tetrachlorethane. Ottawa, Canada. Eriksson, C; Brittebo, EB. (1991) Epithelial binding of 1,1,2,2-tetrachloroethane in the respiratory and upper alimentary tract. Arch Toxicol 65:10-14. Ferreira-Gonzalez, A; DeAngelo, AB; Nasim, S; et al. (1995) Ras oncogene activation during hepatocarcinogenesis in B6C3F male mice by dichloroacetic and trichloroacetic acid. Carcinogenesis 16:495-500. Flick, EW. (1985) Industrial solvents handbook. Park Ridge, NJ: Noyes Publication, p. 134. Forbes, G. (1943) Tetrachloroethane poisoning. Br Med J 1:348-350. Galloway, SM; Armstrong, MJ; Reuben, C; et al. (1987) Chromosome aberrations and sister chromatid exchanges in Chinese hamster ovary cells: evaluations of 108 chemicals. Environ Mol Mutagen 10:1-175. 114 DRAFT - DO NOT CITE OR QUOTE ------- Gargas, ML; Anderson, ME. (1989) Determining kinetic constants of chlorinated metabolism in the rat from rates of exhalation. Toxicol Appl Pharmacol 99:344-353. Gargas, ML; Burgess, RJ; Voisard, DE; et al. (1989) Partition coefficients of low-molecular-weight volatile chemicals in various liquids and tissues. Toxicol Appl Pharmacol 98:87-99. Gohlke, R; Schmidt, P. (1972) Subacute action of low concentrations of chlorinated ethane with and without additional ethanol treatment in the rat. Int Arch Arbeitsmed 30:299-312. Gohlke, R; Schmidt, P; Bahmann, H. (1977) 1,1,2,2-Tetrachloroethane and heat stress in animal experiment. Morphological results. Z Gesamte Hyg 20: 278-282. (German) Grasso, P. (2002) Essentials of pathology for toxicologists. Taylor and Francis, London. Gulati, DK; Grimes, LK; Smith, MR; et al. (199la) Range finding studies: developmental toxicity. 1,1,2,2-Tetrachloroethane (repeat) then administered via feed in CD Sprague-Dawley rats. Study No: NTP-91- RF/DT-017. Gulati, DK; Grimes, LK; Smith, MR; et al. (1991b) Range finding studies: developmental toxicity. 1,1,2,2-Tetrachloroethane (repeat) then administered via feed in Swiss CD-I mice. Study No: NTP-91-RF/DT-020. Halpert, J. (1982) Cytochrome P-450 dependent covalent binding of 1,1,2,2-tetrachloroethane in vitro. Drug Metab Dispos 10:465-468. Halpert, J; Neal, RA. (1981) Cytochrome P-450-dependent metabolism of 1,1,2,2-tetrachloroethane in dichloroacetic acid in vitro. Biochem Pharmacol 30:1366-1368. Halpert, JR; Balfour, C; Miller, NE; et al. (1986) Dichloromethyl compounds as mechanism-based inactivators of rat liver cyrochromes P-450 in vitro. Mol Pharmacol 30:19-24. Hamilton, A. (1917) Military medicine and surgery. JAMA 69:2037-2039. Hansch, C; Leo, AJ. (1985) Medchem project. Issue no. 26. Claremont, CA: Pomona College. Harris, RZ; Benet, LZ; Schwartz, JB. (1995) Gender effects in pharmacokinetics and pharmacodynamics. Drugs 50(2):222-239. Haseman, JK; Hailey, JR; Morris, RW. (1998) Spontaneous neoplasm incidences in Fischer 344 rats and B6C3F1 mice in two-year carcinogenicity studies: a National Toxicology Program update. Toxicol Pathol 26(3):428-441. Hawley, GG. (1981) Condensed chemical dictionary. New York, NY: VanNostrand Reinhold, p. 1003. Haworth, S; Lawlor, T; Mortelmans, K; et al. (1983) Salmonella mutagenicity test results for 250 chemicals. Environ Mutag 5(Suppl 1):3-142. Hayes, W. (2001) Principles and methods of toxicology. 4th edition. Philadelphia, PA: Taylor and Francis. Henics, T; Wheatley, DN. (1999) Cytoplasmic vacuolation, adaptation and cell death: a view on new perspectives and features. Biol Cell. 91:485-498. Hepple, RA. (1927) An unusual case of poisoning. J R Army Med Corps 49:442-445. Herren-Freund, SL; Pereira, MA; Khoury, DK; et al. (1987) The carcinogenicity of trichloroethylene and its metabolites, trichloroacetic acid and dichloroacetic acid, in mouse liver. Toxicol Appl Pharm 90:183-189. Horiguchi, S; Morioka, S; Utsunomiya, T; et al. (1964) A survey of the actual conditions of artificial pearl factories with special reference to work using tetrachloroethane. Jpn J Ind Health 6:251-256. 115 DRAFT - DO NOT CITE OR QUOTE ------- Horiuchi, K; Horiguchi, S; Hashimoto, K; et al. (1962) Studies on the industrial tetrachloroethane poisoning. Osaka City Med 18:29-38. Horvath, M; Frantik, E. (1973) To the relative sensitivity of nervous functions and behavior to nonspecific effects of foreign substances. Act Nerv Super (Praha) 15:25-27. HSDB (Hazardous substance data bank). (2009) National Library of Medicine, Bethesda, MD. http://toxnet.nlm.nih.gov/cgi-bin/sis/search (August 10, 2009). IARC (International Agency for Research on Cancer). (1999) 1,1,2,2-Tetrachloroethane. IARC Monogr Eval Carcmog Risks Hum 71(Pt 2):817-827. Ikeda, M; Ohtsuji, H. (1972) A comparative study of the excretion of Fujiwara reaction-positive substances in urine of humans and rodents given trichloro- or tetrachloro-derivatives of ethane and ethylene. Br J Ind Med 29:99-104. Jeney, E; Bartha, F; Kondor, L; et al. (1957) Prevention of industrial tetrachloroethane intoxication—Part III. Egeszsegtudomany 1:142-164. (Hungarian) Koizumi, A; Kumai, M; Ikeda, M. (1982) Enzymatic formation of an olefin in the metabolism of 1,1,2,2-tetrachloroethane: an in vitro study. Bull Environ Contam Toxicol 29:562-565. Kulinskaya, IL; Verlinskaya, RV. (1972) Comparative effect of low concentrations of di-, tetra-, and pentachloroethane on the blood acetylcholine system. Gig Tr Prof Zabol 16:56-58. (Russian) Kunkel, GH; Hoagland, CL. (1947) Mechanism and significance of the thymol turbidity test for liver disease. J Clin Invest 26(6): 1060-1071. Lantum, HB; Board, PG; Anders, MW. (2002) Kinetics of the biotransformation of maleylacetone and chlorofluoroacetic acid by polymorphic variants of human glutathione transferase zeta (hGSTZl-1). Chem Res Toxicol 15:957-963. Lehmann, KB; Schmidt-Kehl, L; Ruf, H; et al. (1936) The thirteen most important chlorinated aliphatic hydrocarbons from the standpoint of industrial hygiene. Arch Hyg 116:132-200. Lide, DR, ed. (1993) CRC handbook of chemistry and physics, Boca Raton, FL: CRC Press Inc., pp. 4-39. Lilliman, B. (1949) Suggested mechanism of poisoning by liquid tetrachloroethane. Analyst 74:510-511. Lobo-Mendonca, R. (1963) Tetrachloroethane -a survey. Br J Ind Med 20:51-56 Mackay, D; Shiu, WY. (1981) A critical review of Henry's Law constants for chemicals of environmental interest. J Phys Chem Ref Data 10:1175-1199. Mant, AK. (1953) Acute tetrachloroethane poisoning: a report on two fatal cases. Br Med J 1:655-656. Maronpot, RR; Shimkin, MB; Witschi, HP; et al. (1986) Strain a mouse pulmonary tumor test results for chemicals previously tested in the national cancer institute carcinogenicity tests. J Natl Cancer Inst 76:1101-1112. McGregor, DB. (1980) Tier II mutagenic screening of 13 NIOSH priority compounds, individual compound report, 1,1,2,2-tetrachloroethane . Inveresk Research International Limited, Musselburgh EH21 7UB Scotland NIOSH, Cincinnati, OH. , Report No 26. McKim, JM; Lien, GJ; Hoffman, AD; et al. (1999) Respiratory-cardiovascular physiology and xenobiotic gill flux in the lake trout (Salvelinus namaycush). Comp Biochem Physiol A Mol Integr Physiol 123:69-81. Mellon Institute of Industrial Research. (1947) Repeated exposure of rats and dogs to vapors of eight chlorinated hydrocarbons. Union Carbide Corp. Submitted under TSCA Section 8D; EPA Document No. 86-870001397; NTIS No. OTS0515559. 116 DRAFT - DO NOT CITE OR QUOTE ------- Meulenberg, CJW; Vijverberg, HPM. (2000) Empirical relations predicting human and rat tissue:air partition coefficients of volatile organic compounds. Toxicol Appl Pharmacol 165:206-216. Meulenberg, CJ; Wijnker, AG; Vijverberg, HPM. (2003) Relationship between olive oil:air, saline:air, and rat brain:air partition coefficients of organic solvents in vitro. J Toxicol Environ Health, Part A 66:1985-1998. Milman, HA; Story, DL; Riccio, ES; et al. (1988) Rat liver foci and in vitro assays to detect initiating and promoting effects of chlorinated ethanes and ethylenes. Ann NY Acad Sci 534:521-530. Minot, GR; Smith, LW. (1921) The blood in tetrachloroethane poisoning. Arch Intern Med 28:687-702. Mirsalis, JC; Tyson, CK; Steinmetz, KL; et al. (1989) Measurement of unscheduled DNA synthesis and S-phase synthesis in rodent hepatocytes following in vivo treatment: testing of 24 compounds. Environ Mol Mutagen 14:155-164. Mitoma, C; Tyson, CA; Riccio, ES. (1984) Investigations of the species sensitivity and mechanism of carcinogenicity of halogenated hydrocarbons. Standford Research Institute International, Menlo Park, CA; Submitted under TSCA Section 4; EPA Document No. 40-8424225; NTIS No. OTS0509408. Mitoma, C; Steeger, T; Jackson, SE; et al. (1985) Metabolic disposition study of chlorinated hydrocarbons in rats and mice. Drug Chem Toxicol 8:183-194. Miyagawa, M; Takasawa, H; Sugiyama, A; et al. (1995) The in vivo-in vitro replicative DNA synthesis (RDS) test with hepatocytes prepared from male B6C3F1 mice as an early prediction assay for putative nongenotoxic (Ames- negative) mouse hepatocarcinogens. Mutat Res 343:157-183. Morgan, A; Black, A; Belcher, DR. (1970) The excretion in breath of some aliphatic halogenated hydrocarbons following administration by inhalation. Ann Occup Hyg 13:219-233. Mugford, CA; Kedderis, GL. (1998) Sex-dependent metabolism of xenobiotics. Drug Metab Rev 30:441-498. NCI (National Cancer Institute). (1976) Carcinogenesis bioassay of trichloroethylene. Natl Cancer Inst Carcinogen Tech Rep SerNo. 2; NCI-CG-TR-2. NIH Publication No. 76-802. NCI (National Cancer Institute). (1977) Bioassay of tetrachloroethylene for possible carcinogenicity. Natl Cancer Inst Tech Rep SerNo. 13; NCI-CG-TR-13. NIH Publication No. 77-813. NCI (National Cancer Institute). (1978) Bioassay of 1,1,2,2-tetrachloroethane for possible carcinogenicity. Natl Cancer Inst Carcmog Tech Rep Ser 27:1-86.:l-86; NIH Publication No. 78-827. PB2774537GA. NRC (National Research Council). (1983) Risk assessment in the federal government: managing the process. Washington, DC: National Academy Press. NTP (National Toxicology Program). (1986) NTP technical report on the toxicology and carcinogenesis tetrachloroethylene (perchloroethylene) (CAS No. 1127-18-4) in F344/N and B6C3F1 mice (inhalation studies). U.S. DHHS, Public Health Service, National Institute of Health. Technical Report Series, Number 311; NIH Publication No. 86-2567. NTP (National Toxicology Program). (1990) Carcinogenesis studies of trichloroethylene (without epichlorohydrin) (CAS No. 79-01-6) in F344/N rats and B6C3F1 mice (gavage studies). U.S. DHHS, Public Health Service, National Institute of Health. Technical Report Series No. 243; NIH Publication No. 90-1779. NTP. (National Toxicology Program). (1996) NTP technical report on renal toxicity studies of selected halogenated ethanes administered by gavage to F344/Nrats. U.S. DHHS, Public Health Service, National Institute of Health. Toxicity Report Series, Number 45. NTP. (National Toxicology Program). (2004) NTP technical report on the toxicity studies of 1,1,2,2-tetrachloroethane administered in microcapsules in feed to F344/N rats and B6C3F1 mice. U.S. DHHS, Public Health Service, National Institute of Health. Toxicity Report Series, Number 49. 117 DRAFT - DO NOT CITE OR QUOTE ------- Nebert, DW; Adesnik, M; Coon, MJ; et al. (1987) The P450 gene superfamily: recommended nomenclature. DNA 6:1-11. Nestmann, ER; Lee, EGH. (1983) Mutagenicity of constituents of pulp and paper mill effluent in growing cells of Saccharomyces cerevisiae. Mutat Res 119:273-280. Nestmann, ER; Lee, EG-H; Matula, TI; et al. (1980) Mutagenicity of constituents identified in pulp and paper mill effluents using the salmonella/mammalian-microsome assay. Mutat Res 79:203-212. Nichols, JW; Mckim, JM; Lien, GJ; et al. (1993) Physiologically-based toxicokinetic modeling of three waterborne chloroethanes in channel catfish, Ictaluruspunctatus. Aqua Toxicol 27:83-111. Norman, JE; Robinette, CD; Fraumeni, IF. (1981) The mortality experience of army World War II chemical processing companies. J Occup Med 23:818-822. Omiecinski, CJ; Remmel, RP; Hosagrahara, VP. (1999) Concise review of the cytochrome P450s and their roles in toxicology. Toxicol Sci 48:151-156. Ono, Y; Kobayashi, U; Somiya, I; et al. (1996) Evaluation of DNA damage by active oxygen induced by organochlorine compounds and nitroarenes. Mizu Kankyo Gakkaishi 19:871-877. Osol, A. (1972) Blakiston's gould medical dictionary. 3rd edition. New York, NY: McGraw-Hill, Inc., p. 1828. Pantelitsch, M. (1933) Experiments concerning the effect of chlorinated methane and ethane on mice: the relative sensitivity of mice and cats to poisons. Wurzburg: Julius Maximilian University, pp. 1-13. Paolini, M; Sapigni, E; Mesirca, R; et al. (1992) On the hepatotoxicity of 1,1,2,2-tetrachloroethane. Toxicology 73:101-115. Parmenter, DC. (1921) Tetrachloroethane poisoning and its prevention. J Ind Hyg 2:456-465. Pereira, MA. (1996) Carcinogenic activity of dichloroacetic acid and trichloroacetic acid in the liver of female B6C3F1 mice. Fundam Appl Toxicol 31:192-199. Pereira, MA; Phelps, JB. (1996) Promotion by dichloroacetic acid and trichloroacetic acid of N-methyl-N- nitrosourea-initiated cancer in the liver of female B6C3F1 mice. Cancer Lett 102:133-141. Plaa, GL; Hewitt, WR. (1998) Toxicology of the liver. 2nd edition. Washington, DC: Taylor and Francis, p. 431. Price, NH; Allen, SD; Daniels, AU; et al. (1978) Toxicity data for establishing "immediately dangerous to life or health" (IDLH) values. Cincinnati, OH: National Institute for Occupational Safety andHealth; PB87163531. Reitz, RH; Gargas, ML; Mendrala, AL; et al. (1996) In vivo and in vitro studies of perchloroethylene metabolism for physiologically based pharmacokinetic modeling in rats, mice, and humans. Toxicol and App Pharm 136:289- 306. Richmond, RE; Carter, JH; Carter, HW; et al. (1995) Immunohistochemical analysis of dichloroacetic acid (DCA)- induced hepatocarcinogenesis in male Fischer (F344) rats. Cancer Lett 92:67-76. Riddick, JA; Bunger, WB; Sakano, TK. (1986) 1,1,1-Trichloroethane. In: Organic solvents. Physical properties and methods of purification. New York, NY: John Wiley and Sons; pp. 358-359. Robbins, SL; Angell M. (1976) Disease at the cellular level. In: Basic pathology, 2nd edition. Philadelphia, PA: Saunders; pp. 3-30. Roldan-Arjona, T; GarciB-Pedrajas, MD; Luque-Romero, FL. (1991) An association between mutagenicity of the Ara test of Salmonella typhimurium and carcinogenicity in rodents for 16 halogenated aliphatic hydrocarbons. Mutagenesis 6:199-205. 118 DRAFT - DO NOT CITE OR QUOTE ------- Rosenkranz, HS. (1977) Mutagenicity of halogenated alkanes and their derivatives. Environ Health Perspect 21:79- 84. Sato, A; Nakajima, T; Koyama, Y. (1980) Effects of chronic ethanol consumption on hepatic metabolism of aromatic and chlorinated hydrocarbons in rats. Br J Ind Med 37:382-386. Sato, A; Nakajima, T. (1987) Pharmacokinetics of organic solvent vapors in relation to their toxicity. Scand J Work Environ Health 13:81-93. Schmidt, P; Binnewies, S; Gohlke, R; et al. (1972) Subacute action of low concentrations of chlorinated ethanes on rats with and without additional ethanol treatment. 1. Biochemical and toxicometric aspects, especially results in subacute and chronic toxicity studies with 1,1,2,2-tetrachloroethane. Int Arch Arbeitsmed 30:283-298. (German) Schmidt, P; Burck, D; Buerger, A; et al. (1980a) On the hepatotoxicity of benzene, 1,1,2,2-tetrachloroethane and carbon tetrachloride. Z Gesamte Hyg 26:167-172. (German) Schmidt, P; Gohlke, R; Just, A; et al. (1980b) Combined action of hepatotoxic substances and increased environmental temperature on the liver of rats. J Hyg Epidemiol Microbiol Immunol 24:271-277. Sherman, JB. (1953) Eight cases of acute tetrachloroethane poisoning. J Trop Med Hyg 56:139-140. Shimada, T; Yamazaki, H; Mimura, M; et al. (1994) Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 Caucasians. J Pharmacol Exp Ther 270:414-423. Shmuter, LM. (1977) The effect of chronic exposure to low concentrations of ethane series chlorinated hydrocarbons on specific and nonspecific immunological reactivity in animal experiments. Gig Tr Prof Zabol 20:38-43. Smyth, HF; Carpenter, CP; Weil, CS; et al. (1969) Range-finding toxicity data: list VII. Am Ind Hyg Assoc J 30:470-476 Story, DL; Meierhenry, EF; Tyson, CA; et al. (1986) Differences in rat liver enzyme-altered foci produced by chlorinated aliphatics and phenobarbital. Toxicol Ind Health 2:351-362. Theiss, JC; Stoner, GD; Shimkin, MB; et al. (1977) Test for carcinogenicity of organic contaminants of United States drinking waters by pulmonary tumor response in strain A mice. Cancer Res 37(8):2717-2720. Tomasi, A; Albano, E; Bini, A; et al. (1984) Free radical intermediates under hypoxic conditions in the metabolism of halogenated carcinogens. Toxicol Pathol 12:240-246. Tomokuni, K. (1969) Studies on hepatotoxicity induced by chlorinated hydrocarbons. Lipid and ATP metabolisms in the liver of mice exposed to 1,1,2,2-tetrachloroethane. Acta Med Okayama 23:273-282. Tomokuni, K. (1970) Hepatotoxicity induced by chlorinated hydrocarbons. II. Lipid metabolism and absorption spectrum of microsomal lipids in the mice exposed to 1,1,2,2-tetrachloroethane. Acta Med Okayama 24:315-322. TOXNET (Toxicology Data Network). (2009). National Library of Medicine, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD. Available online at http://toxnet.nlm.nih.gov (accessed August 4, 2009). Truffert, L; Girard-Wallon, C; Emmerich, E; et al. (1977) Early experimental demonstration of the hepatotoxicity of some chlorinated solvents by the study of the synthesis of hepatic DNA. Arch Mai Prof 38:261-263. TSI Mason Laboratories. (1993a) 14 Day pilot gavage toxicity study of 1,1,2,2-tetrachloroethane in male F344/N rats. Contract NO1-ES-15326. MLI-NTP-1-93-1. Submitted to NTP. 119 DRAFT - DO NOT CITE OR QUOTE ------- TSI Mason Laboratories. (1993b) 14 Day pilot gavage toxicity study of 1,1,2,2-tetrachloroethane in male F344/N rats. Contract NO1-ES-15326. MLI-NTP-13-93-13. Submitted to NTP. TSI Mason Laboratories. (1993c) 14 Day pilot gavage toxicity study of 1,1,2,2-tetrachloroethane in B6C3F1 mice. Contract NO1-ES-15326. MLI-NTP-10-93-10. Submitted to NTP. TSI Mason Laboratories. (1993d) 14 Day pilot dosed feed toxicity study of microencapsulated 1,1,2,2-tetrachloro- ethane in B6C3F1 mice. Contract NO1-ES-15326. MLI-NTP-9-93-9. Submitted to NTP. Tu, AS; Murray, TA; Hatch, KM; et al. (1985) In vitro transformations of BALB/C-3T3 cells by chlorinated ethanes and ethylenes. Cancer Lett 28:85-92. Tzeng, HF; Blackburn, AC; Board, PG; et al. (2000) Polymorphism- and species-dependent inactivation of glutathione transferase zeta by dichloroacetate. Chem Res Toxicol 13:231-236. U.S. EPA (Environmental Protection Agency). (1986) Guidelines for mutagenicity risk assessment. Federal Register 51(185):34006-34012. U.S. EPA (Environmental Protection Agency). (1987) 1,1,2,2-tetrachloroethane (CASRN 79-34-5). Available online at http:// www.epa.gov/iris (accessed August 4, 2009). U.S. EPA (Environmental Protection Agency). (1988) Recommendations for and documentation of biological values for use in risk assessment. EPA 600/6-87/008. Available from: National Technical Information Service, Springfield, VA; PB88-179874/AS. U.S. EPA (Environmental Protection Agency). (1991a) Guidelines for developmental toxicity risk assessment. Federal Register 56(234):63798-63826. U.S. EPA (Environmental Protection Agency). (1991b) Toxicology of the chloroacetic acids by-products of the drinking water disinfection process. II. The comparative carcinogenicity of dichloroacetic and trichloroacetic acid: implication for risk assessment. Document No. HERL-0820. Research Triangle Park, NC: Health Effects Research Laboratory. U.S. EPA (Environmental Protection Agency). (1994a) Interim policy for particle size and limit concentration issues in inhalation toxicity studies. Federal Register 59(206):53799. U.S. EPA (Environmental Protection Agency). (1994b) Methods for derivation of inhalation reference concentrations and application of inhalation dosimetry. EPA/600/8-90/066F. Available from: National Technical Information Service, Springfield, VA; PB2000-500023. Available online at http://www.epa.gov/iris/backgr-d.htm (accessed August 4, 2009). U.S. EPA (Environmental Protection Agency). (1995) Use of the benchmark dose approach in health risk assessment. EPA/630/R-94/007. Available from: National Technical Information Service (NTIS) , Springfield, VA; PB95-213765. Available online at http://www.epa.gov/iris/backgr-d.htm (accessed August 4, 2009). U.S. EPA (Environmental Protection Agency). (1996) Guidelines for reproductive toxicity risk assessment. Federal Register 61(212):56274-56322. U.S. EPA (Environmental Protection Agency). (1998a) Guidelines for neurotoxicity risk assessment. Federal Register 63(93):26926-26954. U.S. EPA (Environmental Protection Agency). (2000a) Science policy council handbook: risk characterization. Prepared by the Office of Science Policy, Office of Research and Development, Washington, DC. EPA 100-B-OO- 002. Available online at http://www.epa.gov/iris/backgr-d.htm (accessed August 4, 2009). U.S. EPA (Environmental Protection Agency). (2000b) Benchmark dose technical guidance document [external review draft]. EPA/630/R-00/001. Available online at http://www.epa.gov/iris/backgr-d.htm (accessed August 4, 2009). 120 DRAFT - DO NOT CITE OR QUOTE ------- U.S. EPA (Environmental Protection Agency). (2000c) Supplemental guidance for conducting health risk assessment of chemical mixtures. EPA/630/R-00/002. Available online at http://www.epa.gov/iris/backgr-d.htm (accessed August 4, 2009). U.S. EPA (Environmental Protection Agency). (2003) Toxicological review of dichloroacetic acid. In support of the Integrated Risk Information System (IRIS). August 2003. EPA 635/R-03/007. U.S. EPA (Environmental Protection Agency). (2005a) Guidelines for carcinogen risk assessment. Risk Assessment Forum, Washington, DC; EPA/630/P-03/00IB. Available online at http://www.epa.gov/iris/backgrd.htm (accessed January 15, 2009). U.S. EPA (Environmental Protection Agency). (2005b) Supplemental guidance for assessing susceptibility from early-life exposure to carcinogens. Risk Assessment Forum, Washington, DC; EPA/630/R-03/003F. Available online at http://www.epa.gov/iris/backgr-d.htm (accessed January 15, 2009). U.S. EPA (Environmental Protection Agency). (2006a) Peer review handbook. 3rd edition. Review draft. Science Policy Council, Washington, DC. Available online at http://www.epa.gov/peerreview (accessed August 4, 2009). U. S. EPA (Environmental Protection Agency). (2006b) A framework for assessing health risk of environmental exposures to children. National Center for Environmental Assessment, Washington, DC, EPA/600/R-05/093F. Available online at http://cfpub.epa.gov/ncea/cfm/recordisplay.cfm?deid=l58363 (accessed August 4, 2009). Vogel, EW; Nivard, MJM. (1993) Performance of 181 chemicals in a Drosophila assay predominantly monitoring interchromosomal mitotic recombination. Mutagenesis 8:57-81. Ward, JM. (1955) Accidental poisoning with tetrachloroethane. Br Med J 1:1136. Warner, JR; Hughes, TJ; Claxton, LD. (1988) Mutagenicity of 16 volatile organic chemicals in a vaporization technique with Salmonella typhimurium TA100. Environ Mol Mutagen 11:111-112. Willcox, WH; Spilsbury, BH; Legge, TM. (1915) An outbreak of toxic jaundice of a new type amongst aeroplane workers-its clinical and toxological aspects. Trans Med Soc London 38:129-156. Williams, GM. (1983) DNA repair tests of 11 chlorinated hydrocarbon analogs. Final report. U.S. Environmental Protection Agency. NTIS No. OTS408324292. Wolff, L. (1978) The effect of 1,1,2,2,-tetrachloroethane on passive avoidance learning and spontaneous locomotor activity. Act Nerv Super (Praha) 20:14-16. Woodruff, RC; Mason, JM; Valencia, R; et al. (1985) Chemical mutagenesis testing in drosophila. V. Results of 53 coded compounds tested for the National Toxicology Program. Environ Mutagen 7:677-702. Yllner S. (1971) Metabolism of l,l,2,2-tetrachloroethane-14C in the mouse. Acta Pharmacol Toxicol (Copenh) 29:499-512. 121 DRAFT - DO NOT CITE OR QUOTE ------- 1 APPENDIX A: SUMMARY OF EXTERNAL PEER REVIEW AND PUBLIC 2 COMMENTS AND DISPOSITION 3 4 The Toxicological Review of 1,1,2,2-tetrachloroethane (dated August, 2009) has 5 undergone a formal external peer review performed by scientists in accordance with EPA 6 guidance on peer review (U.S. EPA, 2006a, 2000a). The external peer reviewers were tasked 7 with providing written answers to general questions on the overall assessment and on chemical- 8 specific questions in areas of scientific controversy or uncertainty. A summary of significant 9 comments made by the external reviewers and EPA's responses to these comments arranged by 10 charge question follow. In many cases, the comments of the individual reviewers have been 11 synthesized and paraphrased in development of Appendix A. An external peer-review workshop 12 was held January 27, 2010. EPA did not receive any scientific comments from the public. 13 14 EXTERNAL PEER REVIEW PANEL COMMENTS 15 The reviewers made several editorial suggestions to clarify specific portions of the text. 16 These changes were incorporated in the document as appropriate and are not discussed further. 17 In addition, the reviewers provided comments specific to particular decisions and 18 analyses presented in the Toxicological Review under multiple charge questions. These 19 comments were organized and responded to under the most appropriate charge question. 20 21 A. General Comments 22 23 1. Is the Toxicological Review logical, clear and concise? Has EPA clearly synthesized the 24 scientific evidence for noncancer and cancer hazard? 25 26 Comments: The reviewers, generally, commented that the Toxicological Review was 27 logically written. One reviewer recommended an improvement to the clarity of the document 28 by reducing the text describing the available studies and presenting the individual study data 29 in a bulleted format, and this was echoed by another reviewer who recommended condensing 30 the study summaries and discussions. 31 32 Response: The content of the Toxicological Review is consistent with the current outline for 33 IRIS Toxicological Reviews, although an effort has been made to streamline the document and 34 reduce the redundancy. The general structure of a Toxicological Review is to present a factual 35 summary of toxicity studies in Section 4 and critical interpretation/synthesis in Section 5. 36 37 2. Please identify any additional studies that should be considered in the assessment of the 38 noncancer and cancer health effects of l,!92,2-tetrachloroethane. 39 A-l B-l DRAF1 ------- 1 Comments: One reviewer identified the following studies: 2 3 Matsuoka A, Yamakage K, Kusakabe H, Wakuri S, Asakura M, Noguchi T, Sugiyama T, 4 Shimada H, Nakayama S, Kasahara Y, Takahashi Y, Miura KF, Hatanaka M, Ishidate M 5 Jr, Monta T, Watanabe K, Hara M, Odawara K, Tanaka N, Hayashi M, Sofuni T. Re- 6 evaluation of chromosomal aberration induction on nine mouse lymphoma assay "unique 7 positive'NTP carcinogens. 1996. Mutat Res. Aug 12;369(3-4):243-52. 8 9 Sofuni T, Honma M, Hayashi M, Shimada H, Tanaka N, Wakuri S, Awogi T, Yamamoto 10 KI, Nishi Y, Nakadate M. Detection of in vitro clastogens and spindle poisons by the 11 mouse lymphoma assay using the microwell method: interim report of an international 12 collaborative study. Mutagenesis. 1996 Jul;ll(4):349-55. 13 14 Ashley DL, Bonin MA, Cardinali FL, McCraw JM, Wooten JV. Blood concentrations of 15 volatile organic compounds in a nonoccupationally exposed US population and in groups 16 with suspected exposure. Clin Chem. 1994 Jul;40(7Pt 2):1401-4. 17 18 Response: The references [Matsuoka et al. (1996), Sofuni et al. (1996), Ashley et al. (1994)] 19 were examined but have not been added to the Toxicological Review, as these references do not 20 contribute significant information to the discussion and analysis in the document. 21 22 B. Oral Reference Dose (RfD) for 1,1,2,2-tetrachloroethane 23 24 1. Subchronic and chronic RfDs for l,l?2,2-tetrachloroethane have been derived from a 25 13-week oral gavage study (NTP, 2004) in rats and mice. Please comment on whether 26 the selection of this study as the principal study has been scientifically justified. Please 27 identify and provide the rationale for any other studies that should be selected as the 28 principal study. 29 30 Comment: The reviewers generally agreed that the selection of the NTP (2004) report as the 31 principal study was scientifically justified. 32 33 Response: No response. 34 35 Comment: One reviewer commented that the Gulati et al. (1991a,b) is the only other study 36 that could be a candidate principal study and provides what may be a more significant 37 endpoint for human health protection; but also states that EPA has made a reasonable 38 selection in the NTP study. A-2 B-l DRAF1 ------- 1 2 Response: The Gulati et al. developmental studies were conducted at doses higher than the 3 subchronic NTP (2004) study, which demonstrated liver effects at lower doses. Therefore, 4 the Gulati et al. studies were not selected as the principal studies. However, potential points 5 of departure (PODs) based on the observed developmental effects from Gulati et al. (1991 a) 6 were provided in the document for comparison purposes. 7 8 Comment: One reviewer requested additional explanation regarding the statement that high 9 incidences of hepatocellular tumors in all mouse groups of the NCI (1978) study precluded 10 evaluation of noncancer effects in the liver. 11 12 Response: A LOAEL of 142 mg/kg-day was selected for chronic inflammation in the 13 kidneys of male mice, while a NOAEL of 142 mg/kg-day and a LOAEL of 284 mg/kg-day 14 were selected for hydronephrosis and chronic inflammation in the kidneys of female mice. 15 The text in Section 5.1.2.1., Choice of Principal Study and Critical Effect - with Rationale 16 and Justification, addressing the high incidence of hepatocellular tumors in all mouse dose 17 groups and the evaluation of noncancer effects in the liver was deleted. 18 19 2. Increased relative liver weight was selected as the critical effect for the derivation of the 20 subchronic and chronic RfDs. Please comment on whether the rationale for the 21 selection of this critical effect has been scientifically justified. Please provide a detailed 22 explanation. Please identify and provide the rationale for any other endpoints that 23 should be considered in the selection of the critical effect. 24 25 Comment: The reviewers generally agreed that the selection of increased relative liver 26 weight as the critical effect for the derivation of the subchronic and chronic RfDs was 27 justified. One reviewer commented that increased relative liver weight is a less 28 toxicologically significant index of liver change than increased absolute liver weight, due to 29 the treatment-induced loss of body weight; whereas another reviewer believed the change in 30 relative liver weight is more appropriate than absolute liver weight where body weights in 31 general are being affected. Another reviewer commented that increased serum enzyme 32 activity is an alternative critical effect and a true measure of hepatocellular damage, and the 33 most toxicologically-significant endpoint should be selected as the critical effect. A reviewer 34 commented that the only other endpoint that is a candidate critical effect is reduced fetal 35 body weight in the Gulati et al. studies, but also states that EPA's selection of the relative 36 liver weight as the critical effect is reasonable. 37 Two reviewers questioned the statement in the Toxicological Review that the critical 38 effect was selected "because this effect may represent a sensitive endpoint that occurs early A-3 B-l DRAF1 ------- 1 in the process leading to hepatocellular necrosis." The reviewers questioned whether 2 increases in liver weight reflect other, earlier changes that have been going on long enough to 3 cause the cell proliferation, inflammation, or other effects responsible for the observed 4 weight gain. 5 6 Response: The increase in relative liver weight was selected as the basis for the selection of 7 the POD because the relative liver weight analysis takes into account the substantive, dose- 8 dependent decreases in body weight that were observed in both sexes of rats. 9 The reduction in fetal body weight was observed at doses higher than the 10 demonstrated liver effects from the subchronic NTP (2004) study. Therefore, the decrease in 11 fetal body weight was not selected as the critical effect. However, potential points of 12 departure (PODs) based on the observed developmental effects from Gulati et al. (199la) 13 were provided in the document for comparison purposes. 14 EPA considered that, given the available data, increased liver weight represents the most 15 sensitive effect observed in the liver and that it may occur early in the process of liver toxicity 16 associated with oral exposure to 1,1,2,2-tetrachloroethane. In addition to increased liver weight 17 following subchronic exposure, the evidence of hepatocellular damage includes; increased serum 18 concentrations of hepatocellular enzymes (ALT and SDH), decreased serum cholesterol, and 19 increased incidences of hepatocellular necrosis, bile duct hyperplasia, hepatocelluar mitotic 20 alterations, and hepatic pigmentation. In addition, evidence of the 'earlier changes' reflected by 21 the increase in liver weight as suggested by two reviewers is unavailable. Thus, EPA concluded 22 that the observed increase in liver weight may represent the most sensitive effect that occurs early 23 in the process of 1,1,2,2-tetrachloroethane-induced hepatoxicity following subchronic oral 24 exposure. 25 26 3. Hepatocellular vacuolization was observed at the lowest dose in the principal study 27 (NTP, 2004). This effect was not selected as the critical effect for the determination of 28 the POD for derivation of the subchronic and chronic RfDs. Please comment on the 29 rationale and justification for not selecting this endpoint as the critical effect. 30 31 Comment: The reviewers generally considered the rationale and justification for not 32 selecting hepatocellular vacuolization as the critical effect as reasonable, justified, logical, 33 and comprehensive. One reviewer recommended slight refinements to the justification, and 34 questioned whether the comments that vacuolization was not observed across species and the 35 severity was not dose-dependent supported the conclusion. Another reviewer asked if NTP 36 (2004) specified the lobular distribution of the vacuoles. 37 A-4 B-l DRAF1 ------- 1 Response: The decision to not select hepatocellular vacuolization as the critical effect 2 involved more than a consideration of cross species observations and severity (see Section 3 5.1.1.1., Choice of Principal Study and Critical Effect - with Rationale and Justification). 4 The biological significance of the hepatocellular vacuolization observed following 5 1,1,2,2-tetrachloroethane exposure was unclear based on the paucity of information provided 6 by NTP (2004). 7 NTP did not specify the lobular distribution of the observed vacuoles. 8 9 4. The subchronic and chronic RfDs have been derived utilizing benchmark dose (BMD) 10 modeling to define the point of departure (POD). All available models were fit to the 11 data in both rats and mice for increased absolute and relative liver weight, increased 12 incidence of hepatocellular cytoplasmic vacuolization (rats only), increased levels of 13 ALT, SDH, and bile acids, and decreased fetal body weight. Has the BMD modeling 14 been appropriately conducted? Is the benchmark response (BMR) selected for use in 15 deriving the POD (i.e., one standard deviation from the control mean) scientifically 16 justified? Please identify and provide the rationale for any alternative approaches 17 (including the selection of the BMR, model, etc.) for the determination of the POD and 18 discuss whether such approaches are preferred to EPA's approach. 19 20 Comment: Three reviewers stated that the BMD modeling was appropriate. One reviewer 21 disagreed with the reasoning provided in the document for eliminating the two highest dose 22 groups from the BMD modeling analysis for all of the endpoints, and stated that dropping 23 doses is typically only done when the issues of model fit are encountered. A second reviewer 24 commented that EPA should at least show earlier BMD modeling results with the highest 25 doses included and show the lack of model fit that led to the elimination of the two highest 26 doses. 27 28 Response: In agreement with the reviewers' comments, the current reasoning, provided in 29 Section 5.1.1.2 of the document, Methods of Analysis—Including Models (PBPK, BMD, etc.), 30 for dropping the two highest dose groups (exceeding the MTD) was removed. In its place, a 31 rationale for dropping dose groups based on adequacy of model fit was employed. In 32 addition, as requested by two of the external peer reviewers, the endpoints in Table 5-1 were 33 remodeled using the most recent version of BMDS (i.e., 2.1.1). Because of these changes, 34 Appendix B was essentially replaced with a new version showing BMD modeling results 35 (generated using version 2.1.1 of BMDS) with the highest dose groups included to 36 demonstrate that lack of model fit led to the elimination of one or more of these dose groups 37 in order to obtain adequate fit. As a result of this remodeling, a new critical effect was A-5 B-l DRAF1 ------- 1 selected, relative liver weight in female rats, where before, relative liver weight in male rats 2 had been selected. 3 4 5. Please comment on the selection of the uncertainty factors applied to the POD for the 5 derivation of the RfDs. For instance, are they scientifically justified? If changes to the 6 selected uncertainty factors are proposed, please identify and provide a rationale(s). 7 8 Please comment specifically on the following uncertainty factor: 9 • A database uncertainty factor of 3 was used to account for the lack of oral 10 reproductive and developmental toxicity data for 1,1,2,2-tetrachloroethane. 11 Please comment on whether the application of this uncertainty factor has 12 been scientifically justified. 13 14 Comment: The reviewers generally considered the applications of the uncertainty factors to 15 be adequate, acceptable, reasonable, and appropriate. 16 17 Response: No response. 18 19 Comment: One reviewer requested a comparison between the RfD derived from the 20 subchronic NTP study and an approximate RfD derived from the chronic NCI study. 21 22 Response: A comparison between the RfD derived from the subchronic NTP (2004) study 23 and an approximate RfD derived from the chronic NCI (1978) study was considered. The 24 RfD from the subchronic NTP study was based on a study that used lower dose levels and a 25 wider dose range than the NCI (1978) study, and thereby provided a better characterization 26 of the dose-response curve in the low-dose region. Additionally, the route of exposure used 27 in the NTP study, dietary exposure, is a more relevant route of exposure for the general 28 population exposed to 1,1,2,2-tetrachloroethane in the environment than the gavage exposure 29 used in the NCI study. However, if one were to use the observance of chronic inflammation 30 in the kidneys of male mice in the NCI study as a LOAEL, for purposes of comparison, the 31 POD of 142 mg/kg-day could be divided by a total UF of 3 00 to yield an RfD of 0.5 mg/kg- 32 day. 33 34 Comment: A reviewer recommended the addition of text addressing the major metabolites of 35 1,1,2,2-tetrachloroethane (dichloroacetic acid, trichloroethylene, perchloroethylene) and how 36 the results of these assessments compare to those derived for 1,1,2,2-tetrachloroethane. 37 A-6 B-l DRAF1 ------- 1 Response: This comparison was considered outside of the scope of the IRIS assessment for 2 1,1,2,2-tetrachloroethane. 3 4 Comment: One reviewer commented that there is a considerable amount of information 5 about the toxicokinetics of related halocarbons [e.g., trichloroethylene (TCE), 6 perchloroethylene (PERC), chloroform, 1,1,1-trichloroethane] in rodents and humans, and 7 that the rank of metabolic activation of the compounds is: mice » rats > humans. Therefore, 8 the toxicokinetic component of the interspecies UF of 10 could be reduced, resulting in a 9 interspecies uncertainty factor of 3. 10 11 Response: The potential difference between animal and human toxicokinetics following 1,1,2,2- 12 tetrachloroethane exposure based on information from related halocarbons was added to Section 13 5.3, Uncertainties in the Oral Reference Dose (RfD) and Inhalation Reference Concentration 14 (RfC)- Upon further evaluation, this information was not considered sufficient to reduce the UF 15 for 1,1,2,2-tetrachloroethane and the UF of 10 was retained. 16 17 Comment: A reviewer commented that Section 5.3 is a restatement of the features that 18 contributed to the valuation of the standard uncertainty factors, and recommended a 19 consideration of what additional uncertainties are present that might impact the results. 20 21 Response: Additional text was added to this section in response to the reviewer's comment. 22 23 C. Inhalation Reference Concentration (RfC) for l,l?2,2-tetrachloroethane 24 25 1. An RfC for l,l?2,2-tetrachloroethane has not been derived. Has the scientific 26 justification for not deriving an RfC been described in the document? Please identify 27 and provide the rationale for any studies that should be selected as the principal study. 28 Please identify and provide the rationale for any endpoints that should be considered in 29 the selection of the critical effect. 30 31 Comment: The reviewers agreed with the decision not to derive an RfC. One reviewer 32 comment that a comparison to metabolically-related compounds is useful and recommended 33 including this information in the discussion of the uncertainties associated with not deriving 34 an RfC. 35 36 Response: Most reviewers were in agreement with the decision to not derive an RfC based 37 on the available data. Additional text related to uncertainties was added to Section 5.3. 38 A-7 B-l DRAF1 ------- 1 D. Carcinogenicity of l,l?2,2-tetrachloroethane 2 3 1. Under EPA's 2005 Guidelines for carcinogen risk assessment (www.epa.gov/iris/backgr- 4 d.htm), the Agency concluded that l,l?2,2-tetrachloroethane is likely to be carcinogenic 5 to humans by all routes of exposure. Please comment on the cancer weight of the 6 evidence characterization. Is the cancer weight of evidence characterization 7 scientifically justified? 8 9 Comment: One reviewer commented that the conclusion that 1,1,2,2-tetrachloroethane is 10 likely to be carcinogenic to humans is one of the weakest likely to be carcinogenic to humans 11 characterizations demonstrated when the data is singularly considered; in addition, given the 12 prevalence of and susceptibility to developing liver tumors in B6C3Fi mice, the reviewer 13 questioned whether a slope factor should be derived from this study. A second reviewer did 14 not concur with the conclusion that 1,1,2,2-tetrachloroethane is likely to be carcinogenic to 15 humans, and thought it would be more accurate to characterize 1,1,2,2-tetrachloroethane as a 16 possible human carcinogen. Several reviewers recommended incorporating the carcinogenic 17 conclusions for related compounds/major metabolites (dichloroacetic acid, trichloroethylene, 18 and perchloroethylene) to make a stronger case for the likely to be carcinogenic to humans 19 determination. 20 21 Response: The cancer weight of evidence descriptor for 1,1,2,2-tetrachloroethane is based 22 on the statistically significant increase in the incidence of hepatocellular carcinomas in both 23 male and female B6C3F1 mice, and the rare hepatocellular tumors observed in the male 24 Osborne-Mendel rats (NCI, 1978). According to the Guidelines for Carcinogen Risk 25 Assessment (U.S. EPA, 2005a), the likely to be carcinogenic to humans descriptor is 26 supported when an agent has tested positive in animal experiments in more than one species, 27 sex, strain, site, or exposure route with or without evidence of carcinogenicity in humans, and 28 in the case of 1,1,2,2-tetrachloroethane, a positive tumor response was observed in both male 29 and female mice. This descriptor is also supported when a rare animal tumor is observed in a 30 single experiment that is assumed to be relevant to humans, and in the case of 1,1,2,2- 31 tetrachloroethane, NCI (1978) considered the liver tumors observed in male rats to be a rare 32 tumor response. 33 Additional text was added to the discussion of the potential susceptibility of B6C3F1 34 mice to developing hepatocellular carcinomas following 1,1,2,2-tetrachloroethane exposure 35 is included in Section 5.4.5, Uncertainties in Cancer Risk Values. 36 Section 4.7.1, Summary of Overall Weight of Evidence, presents the carcinogenicity 37 data available for 1,1,2,2-tetrachloroethane. This section also includes a discussion of the A-8 B-l DRAF1 ------- 1 carcinogenicity data available for dichloroacetic acid, trichloroethylene, and 2 perchloroethylene. 3 4 2. A two-year oral gavage cancer bioassay (NCI, 1978) was selected as the principal study 5 for the derivation of an oral slope factor. Please comment on the appropriateness of the 6 selection of the principal study. 7 8 Comment: The reviewers generally agreed with the selection of the NCI (1978) study as the 9 principal study for the development of an oral slope factor, although the reviewers highlighted 10 that this was the only study available for this purpose. 11 12 Response: No response. 13 14 Comment: One reviewer commented that the NCI study used poorly selected dose levels that 15 were adjusted during the course of the study, the exposure duration was 78 weeks as opposed 16 to the more standard 104 weeks, that there was also a concurrent disease (pneumonia) 17 observed, and that these deficiencies and resulting uncertainties need to be stated in the 18 document. 19 20 Response: Text was added to Section 5.4.5, Uncertainties in Cancer Risk Values, to address 21 the concern associated with the doses selection and modification and the increased incidence 22 of chronice murine pneumonia in the rats. 23 24 Comment: A reviewer expressed concerns that gavage dosing may deliver the chemical in a 25 short term bolus dose and may not provide the same results as a dietary or other oral dosing 26 method that delivers the chemical more gradually over time. 27 28 Response: The potential effect of the corn oil vehicle, as well as the bolus nature of the 29 gavage dose, on the effects observed in the liver following 1,2,3-trichloropropane exposure 30 has been added to Section 5.4.5, Uncertainties in Cancer Risk Values. 31 32 3. An increased incidence of hepatocellular carcinomas in B6C3F1 mice was used to 33 estimate the oral cancer slope factor. Please comment on the scientific justification of 34 this analysis. Has the BMD modeling been appropriately conducted? 35 36 Comment: Several reviewers considered the modeling of the increased incidence of 37 hepatocellular tumors in B6C3F1 mice to be justified and appropriate. One reviewer 38 commented that maybe an oral slope factor should not be derived given the prevalence of and A-9 B-l DRAF1 ------- 1 susceptibility to developing liver tumors in this strain of mice. A reviewer commented that 2 both sexes of B6C3F1 mice have a high spontaneous cancer incidence and referenced a study 3 by Haseman et al. (1998) which reported that male B6C3F1 control mice have a 42% liver 4 cancer incidence. 5 6 Response: The U.S. EPA considers liver tumors in mice to be relevant to humans unless 7 chemical-specific information is available to indicate otherwise. Text addressing this issue is 8 included in Section 5.4.5, Uncertainties in Cancer Risk Values. 9 Text was also added to Section 5.4.5, Uncertainties in Cancer Risk Values, 10 addressing the high spontaneous cancer incidence of liver cancer in male B6C3F1 mice. The 11 42% liver cancer rate for male B6C3F1 mice was for liver adenomas and carcinomas 12 combined, but the NCI (1978) study analysis was for hepatocellular carcinomas, only. 13 Haseman et al. (1998) reported a 17.9 and 8.4% hepatocellular carcinoma rate in feeding 14 studies for male and female B6C3F1 mice, respectively. 15 It should be noted, that even though the B6C3F1 strain may have a high 16 spontaneous cancer incidence, the incidence in the control mice in NCI (1978) was 1/18 in 17 the male vehicle controls and 0/20 in the female vehicle controls, and 3/36 and 1/40 in male 18 and female pooled-vehicle controls, respectively. Comparison of an experimental group is 19 with its concurrent controls was considered to be the most appropriate comparison, and in 20 this case, the control values were considered low (Haseman et al., 1992; Tarone et al., 1981; 21 Gart et al., 1979 referenced in Haseman et al., 1998). 22 23 Comment: One reviewer requested additional model output information, in Appendix C, 24 describing how the multi-stage model fit the data points, even if the reported goodness-of-fit 25 p-value was provided as "NA" because of too many model parameters. 26 27 Response: In response to this comment, the incidence of hepatocellular carcinomas in male 28 and female mice were remodeled using the most recent version of BMDS (version 2.1.1), and 29 the relevant information describing the fit of both the one- and two-stage multistage models 30 to these incidence data have now been included in Appendix C. 31 32 Comment: A reviewer requested additional analysis of the mode of action of carcinogenesis, 33 as the preponderance of genotoxicity data suggest that 1,1,2,2-tetrachloroethane is not 34 genotoxic and the data available indicate promotion potential. This reviewer recommended 35 an uncertainty factor approach for the cancer assessment. A second reviewer also 36 commented that it is more likely that 1,1,2,2-tetrachloroethane may act as a tumor promoter, 37 provided that the majority of the in vitro and in vivo genotoxicity and mutagenicity studies 38 yielded non-positive results. A-10 B-l DRAF1 ------- 1 2 Response: The two available studies providing some evidence to support the promotion 3 potential of 1,1,2,2-tetrachloroethane were added to Section 4.7.3, Mode of action 4 Information. However, the key events associated with any hypothesized mode of action of 5 carcinogenesis of 1,1,2,2-tetrachloroethane cannot be determined provided the information 6 available. 7 8 Comment: A reviewer commented that mice and other rodents metabolize a considerably 9 larger portion of high doses of halocarbons than humans, and, therefore, experience more 10 severe hepatocellular injury, greater formation of covalent adducts, and higher cancer 11 incidences. This reviewer also commented that male B6C3F1 mice have a very high 12 spontaneous liver cancer incidence as indicated by Haseman et al. (1998). The reviewer 13 recommended including a discussion addressing this in the uncertainty section. 14 15 Response: Text was added to Section 5.4.5, Uncertainties in Cancer Risk Values, addressing 16 the potential difference between animal and human toxicokinetics following 1,1,2,2- 17 tetrachloroethane exposure based on information from related halocarbons demonstrating 18 increased metabolic activation in mice compared with humans. In addition, text was also added 19 to Section 5.4.5, Uncertainties in Cancer Risk Values, addressing the high spontaneous 20 cancer incidence of liver cancer in male B6C3F1 mice. 21 22 Comment: A reviewer commented that the document should recognize that administration of 23 large quantities of corn oil promotes lipid accumulation and lipoperoxidative damage of 24 hepatocytes, and that corn oil is believed to be tumorigenic in rats and humans through 25 increased expression of protooncogenes, decreased apotosis, mitogenesis, etc. The reviewer 26 recommended including a discussion addressing this in the uncertainty section. 27 28 Response: EPA has included text in Section 5.4.5, Uncertainties in Cancer Risk Values, that 29 addresses that the bolus administration of 1,2,3-trichloropropane was in corn oil. 30 31 A-11 B-l DRAF1 ------- APPENDIX B. BENCHMARK DOSE MODELING RESULTS FOR THE DERIVATION OF THE RfD Dichotomous Endpoints Incidence of hepatocellular cytoplasmic vacuolization in male and female rats (NTP, 2004) Table B-l. Incidences of hepatocellular cytoplasmic vacuolization in rats exposed to dietary I,l92,2-tetrachlorethane for 14 weeks Nonneoplastic lesion Dose (mg/kg-d) Vehicle control 20 40 80 170 320 Males3 Hepatocellular cytoplasmic vacuolization 0/10 7/1 Ob (1.3) 9/1 Ob (2.0) 10/10b (1.9) 8/1 Ob (1.4) 0/10 Females3 Hepatocellular cytoplasmic vacuolization 0/10 0/10 10/10b (1.7) 10/10b (2.2) 4/1 Ob (1.3) 0/10 a Values represent proportion of animals with the lesion; for those dose groups in which lesions were found, the average severity score is in parenthesis; severity grades were as follows: 1 = minimal, 2 = mild, 3 = moderate, 4 = severe. b Statistically significantly different from vehicle control group. Source: NTP (2004). All available dichotomous models (except the "quantal-linear" and "quantal-quadratic") in the EPA's BMDS (version 2.1.1) were fit to the incidence of hepatocellular cytoplasmic vacuolization in male and female rats administered 1,1,2,2-tetrachloroethane in the diet for 14 weeks. Table B-l displays the incidence data for this endpoint for both males and females. BMDs and their associated 95 percent lower confidence limits (i.e., BMDLs) at an extra risk of 10% were estimated by each model. The results of this BMD modeling for male and female rats are summarized in Tables B-2 and B-3, respectively, and the BMDS output from the selected model are displayed following each table. A-A B-l DRAF1 ------- Table B-2. Summary of BMD modeling results for the incidence of hepatocellular cytoplasmic vacuolization in male rats Model DF x2 5£2 Goodness of fit />-valuea Scaled residuals of interest1" AIC BMD10 (mg/kg- day) BMDL10 (mg/kg- day) All dose groups included BMDS was unable to generate model outputs Highest dose group dropped Gamma0 Logistic Log-logistic ' Log-probit Multistage (1 -degree)6 Probit Weibull0 4 3 4 4 4 3 4 57.61 22.78 6.78 36.46 57.61 20.45 57.61 O.001 O.001 0.15 O.001 O.001 O.001 O.001 0.00/1.66 -2.77/1.01 0.00/-0.06 0.00/0.85 0.00/1.66 3.00/0.94 0.00/1.66 47.97 57.05 36.14 41.77 47.97 58.24 47.97 3.64 10.59 0.91 4.70 3.64 13.29 3.64 2.60 6.70 0.40 3.03 2.60 8.99 2.60 Two highest dose groups dropped Gamma0 Logistic Log-logistic Log-probit Multistage (l-degree)e'g Multistage (2 -degree)0 Multistage (3 -degree)0 Probit Weibull0 2 2 2 2 3 2 2 2 2 0.10 2.50 0.25 0.18 0.10 0.08 0.06 2.56 0.10 0.95 0.29 0.88 0.92 0. 99 0.96 0.97 0.28 0.95 0.00/0.08 -0.82/0.81 0.00/0.09 0.00/0.10 0.00/-0.02 0.00/0.12 0.00/0.13 -0.81/1.03 0.00/0.10 22.87 25.51 23.09 22.98 20.89 22.83 22.80 25.71 22.86 2.47 6.78 6.16 5.49 1.73 1.99 1.89 6.45 2.32 1.12 3.67 0.31 1.80 1.12 1.12 1.13 3.73 1.12 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; DF = degrees of freedom aValues < 0.1 fail to meet conventional goodness-of-fit criteria. Scaled residuals at doses immediately below and immediately above the benchmark dose. °Power restricted to >1. dSlope restricted to >1. "Betas restricted to >0. fAlthough the overall goodness of flip-value suggested adequate fit of this model to the data, the model was rejected because the very high residual at the high dose (-2.32) suggested that fit of the model to the data would be improved by dropping that dose. 8Selected model is displayed in boldface type. BMDLs for models with adequate fit differed by > threefold. However, the results from the log-logistic model were rejected as unreliable due to the large spread between BMD and BMDL (20-fold) and because the BMDL from this model was an outlier in relation to the results of the other models. After dropping this model, the results of the other models were within approximately threefold. Among the remaining models, the 1-degree polynomial had the lowest AIC and also produced the lowest BMDL and was therefore selected as the most suitable model for this dataset. A-B B-l ------- As shown in Table B-2, in attempting to model the incidence of hepatocellular cytoplasmic vacuolization in male rats with all six dose groups included, the BMDS failed to generate any output because response was not a monotonically increasing function of dose (i.e., the response in the penultimate dose group was 80%, while the response in the highest dose group was 0). A key underlying assumption for the fitting of the dichotomous models in BMDS is that response must be a monotonically non-decreasing function of dose. Therefore, the highest dose group was dropped and the models were fit to the data again. In this instance, the chi- square goodness-of-fit test found that all models exhibited inadequate fit (i.e., p < 0.1). Finally, in an attempt to find a model that fit, the two highest dose groups were dropped and the models were refit to these data. In this case, all of the models exhibited adequate fit (p > 0.10). Of these models exhibiting adequate fit, a "best-fit" model was selected consistent with the EPA's Benchmark Dose Technical Guidance Document (USEPA 2000), as follows. If the BMDL estimates from the models exhibiting adequate fit were "sufficiently close," then the model with the lowest AIC is to be used to estimate the BMDL from which the POD will be derived. In this particular case, as explained in the footnote in Table B-2, BMDLs for models with adequate fit differed by greater than threefold. However, the results from the log-logistic model were rejected as unreliable due to the large spread between BMD and BMDL (20-fold) and because the BMDL from this model was an outlier in relation to the results from the other models. After dropping the log-logistic model, the BMDLs from the other models were within approximately threefold. Among the remaining models, the one-stage multistage model had the lowest AIC, and also produced the lowest BMDL, and was therefore selected as the most suitable model for this dataset. The BMDLio from this model (i.e., 1.12 mg/kg-day) was then selected as a possible POD. The standard BMDS output from the one-stage multistage model is displayed below. A-C B-l DRAF1 ------- Multistage Model with 0.95 Confidence Level a o '•8 0.8 0.6 0.4 0.2 Multistage 3MDL BMD 0 10 20 30 40 50 60 70 80 dose 11:41 03/302010 Multistage Model. (Version: 3.0; Date: 05/16/2008) Input Data File: C:\USEPA\lRlS\TCE\NTP\hepcytvac\male\mst_hepcytvacM2HDD_MS_l.(d) Gnuplot Plotting File: C:\USEPA\lRlS\TCE\NTP\hepcytvac\male\mst_hepcytvacM2HDD_MS_l.pit Tue Mar 30 12:41:48 2010 HMDS Model Run The form of the probability function is: P[response] = background + (1-background)* [1-EXP( -betal*doseAl)] The parameter betas are restricted to be positive Dependent variable = incidence Independent variable = dose Total number of observations = 4 A-4 B-l DRAF1 ------- Total number of records with missing values Total number of parameters in model = 2 Total number of specified parameters = 0 Degree of polynomial = 1 = 0 Maximum number of iterations = 250 Relative Function Convergence has been set to: le-008 Parameter Convergence has been set to: le-008 Default Initial Parameter Values Background = 0 Beta(l) = 1.28571e+018 Asymptotic Correlation Matrix of Parameter Estimates Beta(l) ( *** The model parameter(s) -Background have been estimated at a boundary point, or have been specified by the user, and do not appear in the correlation matrix ) Beta(l) 1 Parameter Estimates Variable Background Beta(l) Estimate 0 0. 0607678 Std. Err. 95.0% Wald Confidence Interval Lower Conf. Limit Upper Conf. Limit * - Indicates that this value is not calculated. Model Full model Fitted model Reduced model Analysis of Deviance Table Log(likelihood) -9.35947 -9.44611 -25.8979 # Param's 4 1 1 Deviance Test d.f. P-value 0 .173273 33.0768 0.9818 < .0001 AIC: 20.8922 Goodness of Fit Scaled 0 20 40 80 Dose .0000 .0000 .0000 .0000 Est 0. 0. 0. 0. . Prob. 0000 7034 9120 9923 Expected 0. 7 . 9. 9. .000 .034 .120 .923 Observed 0. 7. 9. 10. .000 .000 .000 .000 Size 10 10 10 10 Residual 0. -0. -0. 0. .000 .024 .134 .279 ChiA2 = 0.10 d.f. = 3 P-value = 0.9922 A-5 B-l DRAF1 ------- Benchmark Dose Computation Specified effect = 0.1 Risk Type = Extra risk Confidence level = 0.95 HMD = 1.73382 BMDL = 1.11682 BMDU = 2.71595 Taken together, (1.11682, 2.71595) is a 90 % two-sided confidence interval for the BMD A-6 B-l DRAF1 ------- Table B-3. Summary of benchmark dose model results for the incidence of hepatocellular cytoplasmic vacuolization in female rats Model DF 7.2 X2 Goodness of fit />-valuea Scaled residuals of interest1" AIC BMD10 (mg/kg-day) BMDL10 (mg/kg-day) All dose groups included BMDS was unable to generate model outputs Highest dose group dropped Gamma0 Logistic Log-logistic Log-probit Multistage (1 -degree polynomial)6 Probit Weibull0 4 3 4 4 4 3 4 45.13 38.70 31.61 49.11 45.13 38.70 45.13 O.001 O.001 O.001 O.001 O.001 O.001 O.001 0.00/-1.66 -2.52/3.63 0.00/-2.36 0.00/-1.61 0.00/-1.66 -2.50/3.65 0.00/-1.66 61.33 69.75 53.57 58.57 61.33 69.79 61.33 8.65 30.61 3.99 12.62 8.65 31.28 8.65 6.18 18.21 2.24 8.86 6.18 19.39 6.18 Two highest dose groups dropped Gamma0 Logistic Log-logistic"1 Log-probitd Multistage (1 -degree polynomial)6 Multistage (2 -degree polynomial)6 Multistage (3 -degree polynomial)6 Probit WeibulF'1 3 2 3 3 3 3 3 2 3 1.56 0.00 0.04 0.00 13.83 7.48 4.41 0.00 0.00 0.67 1.00 1.00 1.00 0.003 0.06 0.22 1.00 1.00 -0.95/0.82 0.00/0.00 -0.14/0.14 0.00/0.00 0.00/-3.09 0.00/-2.24 0.00/-1.78 0.00/0.00 -0.02/0.01 5.00 4.00 2.08 2.00 22.89 14.54 9.85 4.00 2.00 20.59 29.46 25.03 26.36 3.14 10.17 14.53 28.77 30.68 17.05 19.38 19.51 19.56 2.05 5.95 9.15 19.85 19.16 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the concentration associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; DF = degrees of freedom aValues < 0.1 fail to meet conventional goodness-of-fit criteria. bScaled residuals at doses immediately below and immediately above the benchmark dose. °Power restricted to >1. Slope restricted to >1. 6Betas restricted to >0. Selected model is displayed in boldface type. BMDLs for models with adequate fit differed by < threefold, so the models with the lowest AIC (Log-probit and Weibull models) were initially selected as the best fitting. The Weibull model had a slightly lower BMDL between the two models; thus the Weibull was selected. 1 B-l DRAFT - DO NOT CITE OR QUOTE ------- 1 As shown in Table B-3, in attempting to model the incidence of hepatocellular 2 cytoplasmic vacuolization in female rats with all six dose groups included, the BMDS failed to 3 generate any output because response was not a monotonically increasing function of dose (i.e., 4 the response in the penultimate dose group was 40%, while the response in the highest dose 5 group was 0). A key underlying assumption for the fitting of the dichotomous models in BMDS 6 is that response must be a monotonically non-decreasing function of dose. Therefore, the highest 7 dose group was dropped and the models were fit to the data again. In this instance, the chi- 8 square goodness-of-fit test showed that all models exhibited inadequate fit (i.e., p < 0.1). Finally, 9 in an attempt to find a model that fit, the two highest dose groups were dropped and the models 10 were refit to these data. In this case, all of the models exhibited adequate fit, except for the one- 11 and two-stage multistage models (p > 0.10). 12 Of the models exhibiting adequate fit, a "best-fit" model was selected consistent with the 13 EPA's Benchmark Dose Technical Guidance Document (USEPA 2000), as follows. If the 14 BMDL estimates from the models exhibiting adequate fit were "sufficiently close," then the 15 model with the lowest AIC is to be used to estimate the BMDL from which the POD will be 16 derived. In this particular case, as explained in the footnote in Table B-3, BMDLs for models 17 with adequate fit differed by less than threefold. Among these models, the log-probit and 18 Weibull models shared the lowest AIC, and thus the average BMDLio from these two models 19 (i.e., 19.36 mg/kg-day) was used to derive a possible POD. The standard BMDS outputs from 20 the log-probit and Weibull models are displayed below. B-2 DRAFT - DO NOT CITE OR QUOTE ------- LogProbit Model with 0.95 Confidence Level 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 C o • 0.8 0.6 0.4 0.2 LogProbit BMDL BMD 10 20 30 40 dose 50 60 70 80 11:5403/302010 Probit Model. (Version: 3.1; Date: 05/16/2008) Input Data File: C:\USEPA\lRlS\TCE\NTP\hepcytvac\female\lnp_hepcytvacF2HDD_logprobit.(d) Gnuplot Plotting File: C:\USEPA\lRlS\TCE\NTP\hepcytvac\female\lnp_hepcytvacF2HDD_logprobit.plt Tue Mar 30 12:54:34 2010 HMDS Model Run The form of the probability function is: P[response] = Background + (1-Background) * CumNorm(Intercept+Slope*Log(Dose) where CumNorm(.) is the cumulative normal distribution function Dependent variable = incidence Independent variable = dose Slope parameter is restricted as slope >= 1 Total number of observations = 4 Total number of records with missing values = 0 B-3 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 6 7 c o 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 43 44 45 46 47 48 49 50 51 52 53 J -J 54 55 56 57 58 59 60 61 62 63 64 Maximum number of iterations = 250 Relative Function Convergence has been set to: le-008 Parameter Convergence has been set to: le-008 User has chosen the log transformed model Default Initial (and Specified) Parameter Valu background = 0 intercept = -8.43383 slope = 2.43905 Asymptotic Correlation Matrix of Parameter Estimates ( *** The model parameter (s) -background -slope have been estimated at a boundary point, or have been specified and do not appear in the correlation matrix ) intercept intercept 1 Parameter Estimates 95.0% Wald Confidence I Variable Estimate Std. Err. Lower Conf. Limit Upper background 0 NA intercept -60.1746 2420.13 -4803.54 slope 18 NA NA - Indicates that this parameter has hit a bound implied by some inequality constraint and thus has no standard error. Analysis of Deviance Table Model Log (likelihood) # Param's Deviance Test d.f. P- value Full model 0 4 Fitted model -4 .43789e-009 1 8.87578e-009 3 1 Reduced model -27.7259 1 55.4518 3 <.0001 AIC: 2 Goodness of Fit Dose Est. Prob. Expected Observed Size 0.0000 0.0000 0.000 0.000 10 20.0000 0.0000 0.000 0.000 10 -o.ooo 40.0000 1.0000 10.000 lO.ooo 10 o.ooo 80.0000 1.0000 10.000 10.000 10 ChiA2 = 0.00 d.f. = 3 P-value = 1.0000 Benchmark Dose Computation Specified effect = 0.1 es by the user nterval Conf. Limit 4683 .19 Scaled Residual 0.000 0.000 B-4 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 Risk Type = Extra risk 3 4 Confidence level = 0.95 5 6 BMD = 26.3597 7 8 BMDL = 19.557 B-5 DRAFT - DO NOT CITE OR QUOTE ------- Weibull Model with 0.95 Confidence Level 2 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 C o • 0.8 0.6 0.4 0.2 Weibull BMDL BMD 10 20 30 40 dose 50 60 70 80 11:5403/302010 Weibull Model using Weibull Model (Version: 2.12; Date: 05/16/2008) Input Data File: C:\USEPA\lRlS\TCE\NTP\hepcytvac\female\wei_hepcytvacF2HDD_weibull.(d) Gnuplot Plotting File: C:\USEPA\lRlS\TCE\NTP\hepcytvac\female\wei_hepcytvacF2HDD_weibull.plt Tue Mar 30 12:54:37 2010 HMDS Model Run The form of the probability function is: P[response] = background + (1-background)*[1-EXP(-slope*doseApower)] Dependent variable = incidence Independent variable = dose Power parameter is restricted as power >=1 Total number of observations = 4 Total number of records with missing values = 0 Maximum number of iterations = 250 Relative Function Convergence has been set to: le-008 Parameter Convergence has been set to: le-008 B-6 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 6 7 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 43 44 45 46 47 48 T-O 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 Default Initial (and Specified) Parameter Valu Background = 0.0454545 Slope = 0.00369372 Power = 1.53227 Asymptotic Correlation Matrix of Parameter Estimates ( *** The model parameter (s) -Background -Power have been estimated at a boundary point, or have been specified and do not appear in the correlation matrix ) Slope Slope -1.$ Parameter Estimates 95.0% Wald Confidence I Variable Estimate Std. Err. Lower Conf. Limit Upper Background 0 NA Slope 1.815596-028 1 . #QNAN 1 . #QNAN Power 18 NA NA - Indicates that this parameter has hit a bound implied by some inequality constraint and thus has no standard error. Analysis of Deviance Table Model Log (likelihood) # Param's Deviance Test d.f. P- value Full model 0 4 Fitted model -0.000514093 1 0.00102819 3 1 Reduced model -27.7259 1 55.4518 3 <.0001 AIC: 2.00103 Goodness of Fit Dose Est. Prob. Expected Observed Size 0.0000 0.0000 0.000 0.000 10 20.0000 0.0000 0.000 0.000 10 40.0000 1.0000 10.000 10.000 10 80.0000 1.0000 10.000 10.000 10 ChiA2 = 0.00 d.f. = 3 P-value = 1.0000 Benchmark Dose Computation Specified effect = 0.1 Risk Type = Extra risk es by the user nterval Conf. Limit 1 . #QNAN Scaled Residual 0.000 -0.022 0.006 0.000 B-7 DRAFT - DO NOT CITE OR QUOTE ------- 1 Confidence level = 0.95 2 3 HMD = 30.681 4 5 BMDL = 19.1631 6 B-8 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 Continuous Endpoints Organ weight and serum chemistry changes in male and female rats (NTP, 2004) Table B-4. Selected organ weight and serum chemistry changes in male and female F344 rats administered l,l?2,2-tetrachlroethane in the diet for 14 weeks Endpoint Absolute liver wt (g) Relative liver wt (mg organ wt / g body wt) Serum ALT activity (IU/L) Serum SDH activity (IU/L) Serum bile acid levels (umol/L) Sex M F M F M F M F M F Dose (mg/kg-day) 0 12.74 ±0.26a 6.84±0.17 34.79 ±0.42 35.07 ±0.56 48 ±2 46 ±2 23 ±1 27 ±1 29.2 ±2.9 37.0±7.1 20 12. 99 ±0.35 7.03 ±0.13 36.72 ±0.44 36.69 ±0.36 49 ±2 42 ±1 27 ±1 27 ±1 27.5 ±2.7 46.6 ±6.5 40 14.47 ±0.44 7.14±0.16 41.03 ±0.85 37.84±0.51 53±2 41±2 26 ±2 28 ±2 27.2 ±2.7 39.1 ±5.6 80 15. 54 ±0.40 7. 80 ±0.08 45.61 ±0.52 44.20 ±0.27 69 ±3 49 ±2 31±1 25 ±1 35.9±3.9 36. 3 ±3. 9 170 11. 60 ±0.44 6.66 ±0.22 44.68 ±0.45 48.03 ±0.89 115±8 112±7 47 ±2 45 ±3 92.0 ±16.6 39. 3 ±7.9 320 6.57±0.18 4.94±0.12 52.23 ±1.42 58.40 ±1.42 292 ±18 339±18 74 ±4 82 ±3 332.4 ±47.4 321. 5 ±50.6 9 10 11 12 13 14 15 16 17 18 19 20 21 22 "Values are means ± SE for 10 animals. Source: NTP (2004). All available continuous models in theEPA's BMDS (version 2.1.1) were fit to each of the endpoints listed in Table B-4 for both male and female rats administered 1,1,2,2-tetrachloro- ethane in the diet for 14 weeks. BMDs and their 95 percent lower confidence limits (i.e., BMDLs) associated with a change in the response of one standard deviation from the control were estimated by each model. The results of this BMD modeling for male and female rats are summarized in Tables B-5 through B-14. Following each table is the BMDS output for the selected model. The model fitting procedure for continuous data was as follows. The simplest model (linear) is first applied to the data while assuming constant variance. If the data are consistent with the assumption of constant variance (p > 0.1), then the fit of the linear model to the means is evaluated and the polynomial, power, and Hill models are fit to the data while assuming constant variance. In accordance with U.S. EPA (2000) guidance, BMDs and BMDLs are estimated employing a BMR that represents a change of 1 standard deviation from the control. Adequate model fit is judged primarily by the goodness-of-fit/>-value (p > 0.1), but visual inspection of the dose-response curve and the examination of scaled residual at the data point (except the control) closest to the predefined BMR also play a role. If the test for constant variance is negative, the linear model is run again while applying the power model integrated into BMDS to account for B-9 DRAFT - DO NOT CITE OR QUOTE ------- 1 nonhomogeneous variance. If the nonhomogeneous variance model provides an adequate fit (p > 2 0.1) to the variance data, then the fit of the linear model to the means is evaluated and the 3 polynomial, power, and Hill models are fit to the data and evaluated while the variance model is 4 applied. If no model provides adequate fit to the data based on these criteria, then the highest 5 dose is dropped, if appropriate, and the continuous modeling procedure is repeated. 6 7 Absolute liver weights in male and female rats (Tables B-5 andB-6) 8 No adequate fit to the data for absolute liver weight in males or females was achieved 9 until the two highest doses were dropped. After dropping the two highest doses, the assumption 10 of constant variance was met and all models provided adequate fit (except the Hill model, which 11 has too many parameters for the number of remaining data points). BMDL estimates across the 12 models with adequate fit differed by less than threefold. In accordance with U.S. EPA (2000), 13 the model with the lowest AIC (linear, for both males and females) was selected as the basis for 14 the BMDiso and BMDLiso estimates for these endpoints (respectively, 30 and 23 mg/kg-day for 15 males, and 36 and 26 mg/kg-day for females). 16 B-10 DRAFT - DO NOT CITE OR QUOTE ------- Table B-5. Summary of benchmark dose modeling results for absolute liver weight in male rats Model Test for significant difference />-valuea Variance />-valueb Means />-valueb Scaled residuals of interest0 AIC BMD1SD (mg/kg- day) BMDL1SD (mg/kg- day) All dose groups included Constant variance Lineard O.0001 0.07 O.0001 NA 198.13 NA 3,925.92 Non-constant variance Hille Lineard Polynomial (2 -degree/ Polynomial (3 -degree) Polynomial (4-degree)d Polynomial (5-degree)d Power6 O.0001 O.0001 <0.0001 O.0001 <0.0001 <0.0001 <0.0001 0.39 0.39 0.39 0.39 0.39 0.39 0.39 O.0001 O.0001 O.0001 <0.0001 <0.0001 O.0001 <0.0001 -0.7/1.81 NA NA NA NA NA -1.43/0.08 160.48 200.13 200.13 200.13 200.13 200.13 106.77 36.49 NA NA NA NA NA 173.92 NA 10.43 10.45 733.03 595.06 533.37 141.52 Highest dose group dropped Constant variance Hill6 Lineard Polynomial (2-degree)d Polynomial (3 -degree) Polynomial (4-degree)d Power6 <0.0001 O.0001 <0.0001 <0.0001 <0.0001 <0.0001 0.49 0.49 0.49 0.49 0.49 0.49 <0.0001 <0.0001 <0.0001 O.0001 <0.0001 <0.0001 3.3/0.00 NA NA NA NA 3.3/0.00 100.95 112.67 112.67 112.67 112.67 98.95 165.58 NA NA NA NA 166.09 94.36 606.09 416.42 326.66 282.11 145.65 Two highest dose groups dropped Constant variance Hill6 LinearAf Power6 O.0001 <0.0001 <0.0001 0.41 0.41 0.41 NA 0.32 0.13 0.00/0.00 -1.07/0.97 -1.03/1.01 57.97 56.26 58.25 32.10 30.40 31.30 20.62 22.92 22.93 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; NA = not applicable (BMD/BMDL computation failed or insufficient degrees of freedom to fit model); SD = standard deviation aValues >0.05 fail to meet conventional goodness-of-fit criteria. Values <0.10 fail to meet conventional goodness-of-fit criteria. °Scaled residuals at doses immediately below and immediately above the benchmark dose. dCoefficients restricted to be positive. ePower restricted to >1. fBest-fitting model is displayed in boldface type. BMDLs for models providing adequate fit differed by < threefold, so the model with the lowest AIC was selected. B-ll DRAFT - DO NOT CITE OR QUOTE ------- Linear Model with 0.95 Confidence Level c o Q_ (/) 0 ce c ro 0 16 15 14 13 12 Linear BMDL BMD 10 20 30 40 dose 50 60 70 80 14:1203/262010 B-12 DRAFT - DO NOT CITE OR QUOTE ------- 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 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 Polynomial Model. (Version: 2.13; Date: 04/08/2008) Input Data File: C:\USEPA\lRlS\TCE\NTP\abslivwt\male\lin_abslivwtM2HDD_linear.(d) Gnuplot Plotting File: C:\USEPA\IRIS\TCE\NTP\abslivwt\male\lin_abslivwtM2HDD_linear.plt Fri Mar 26 15:12:39 2010 HMDS Model Run The form of the response function is: Y[dose] = beta 0 + beta l*dose + beta 2*doseA2 + ... Dependent variable = mean Independent variable = dose rho is set to 0 The polynomial coefficients are restricted to be positive A constant variance model is fit Total number of dose groups = 4 Total number of records with missing values = 0 Maximum number of iterations = 250 Relative Function Convergence has been set to: le-008 Parameter Convergence has been set to: le-008 Default Initial Parameter Values alpha = 1.35605 rho = 0 Specified beta_0 = 12.626 beta 1 = 0.0374 Asymptotic Correlation Matrix of Parameter Estimates ( *** The model parameter(s) -rho have been estimated at a boundary point, or have been specified by the user, and do not appear in the correlation matrix ) alpha alpha 1 beta_0 -6.9e-010 beta 1 -4.8e-011 Variable alpha beta 0 Estimate 1.29235 12.626 beta_0 beta_l -6.96-010 -4.88-011 1 -0.76 -0.76 1 Parameter Estimates 95.0% Wald Confidence Interval Std. Err. Lower Conf. Limit Upper Conf. Limit 0.288979 0.725966 1.85874 0.278462 12.0802 13.1718 B-13 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 beta 1 0.0374 0.00607655 0.0254902 0.0493098 Table of Data and Estimated Values of Interest Dose N Obs Mean Est Mean Obs Std Dev Est Std Dev Scaled Res. 0 20 40 80 10 10 10 10 12 .7 13 14 .5 15 .5 12 .6 13 .4 14 .1 15 .6 0.82 1.11 1.39 1.26 1.14 1 .14 1 .14 1 .14 0.317 -1 .07 0. 968 -0.217 Model Descriptions for likelihoods calculated Model Al: Yij = Mu(i) + e(ij) Var{e(ij) } = SigmaA2 Model A2: Yij = Mu(i) + e(ij) Var{e(ij) } = Sigma(i) A2 Model A3: Yij = Mu(i) + e(ij) Var{e(ij)} = SigmaA2 Model A3 uses any fixed variance parameters that were specified by the user Yi = Mu + e(i) Var{e (i)} = SigmaA2 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 MO we Model Test 1 Test 2 Test 3 Test 4 (Note: Likelihoods of Interest Model Al A2 A3 fitted R Log (likelihood) -23.984311 -22.556035 -23.984311 -25.129323 -38 .455553 # Param' s 5 8 5 3 2 AIC 57.968622 61.112070 57.968622 56.258645 80.911106 Explanation of Tests Do responses and/or variances differ among Dose levels? (A2 vs. R) Are Variances Homogeneous? (Al vs A2) Are variances adequately modeled? (A2 vs. A3) Does the Model for the Mean Fit? (A3 vs. fitted) When rho=0 the results of Test 3 and Test 2 will be the same.) B-14 DRAFT - DO NOT CITE OR QUOTE ------- 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 Test Test 1 Test 2 Test 3 Test 4 Tests of Interest -2*log (Likelihood Ratio) Test df 31.799 2.85655 2.85655 2.29002 p-value <.0001 0.4143 0.4143 0.3182 The p-value for Test 1 is less than .05. There appears to be a difference between response and/or variances among the dose levels It seems appropriate to model the data The p-value for Test 2 is greater than .1. A homogeneous variance model appears to be appropriate here The p-value for Test 3 is greater than .1. to be appropriate here The p-value for Test 4 is greater than .1. to adequately describe the data The modeled variance appears The model chosen seems Benchmark Dose Computation Specified effect = 1 Risk Type = Estimated standard deviations from the control mean Confidence level = 0.95 HMD = 30.3962 BMDL = 22.9198 B-15 DRAFT - DO NOT CITE OR QUOTE ------- Table B-6. Summary of benchmark dose modeling results for absolute liver weight in female rats Model Test for significant difference />-valuea Variance />-value Means />-value Scaled residuals of interest0 AIC BMD1SD (mg/kg- day) BMDL1SD (mg/kg- day) All dose groups included Constant variance Linear O.0001 0.05 O.0001 NA 62.98 NA 3,632.46 Non-constant variance Linear O.0001 0.02 O.0001 NA 64.98 NA 24.07 Highest dose group dropped Constant variance Lineard O.0001 0.04 O.0001 NA 5.69 NA 377.10 Non-constant variance Hille Lineard Polynomial (2 -degree) Polynomial (3-degree)d Polynomial (4-degree)d Power6 O.0001 O.0001 O.0001 <0.0001 O.0001 <0.0001 0.84 0.84 0.84 0.84 0.84 0.84 <0.0001 <0.0001 O.0001 <0.0001 <0.0001 <0.0001 0.00f NA NA NA NA 0.00f 4.52 7.69 7.69 7.69 7.69 2.52 170.20 NA NA NA NA 170.19 NA 397.23 343.87 290.54 67.91 153.95 Two highest dose groups dropped Constant variance Hill6 Linear11* Polynomial (2-degree)d Polynomial (3-degree)d Power6 <0.0001 <0.0001 <0.0001 O.0001 <0.0001 0.11 0.11 0.11 0.11 0.11 NA 0.55 0.63 0.71 0.57 -0.30/0.05 0.05/-0.91 -0.28/0.05 -0.19/0.02 -0.30/0.05 -19.17 -22.27 -21.25 -21.35 -21.17 48.28 35.62 48.21 49.83 48.28 25.37 26.10 27.58 27.77 27.44 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; NA = not applicable (BMD/BMDL computation failed or insufficient degrees of freedom to fit model); SD = standard deviation aValues >0.05 fail to meet conventional goodness-of-fit criteria. Values <0.10 fail to meet conventional goodness-of-fit criteria. °Scaled residuals at doses immediately below and immediately above the benchmark dose. dCoefficients restricted to be positive. ePower restricted to >1. Residual at highest dose tested. gBest-fitting model displayed in boldface type. BMDLs for models providing adequate fit differed by < threefold, so the model with the lowest AIC was selected. B-16 DRAFT - DO NOT CITE OR QUOTE ------- Linear Model with 0.95 Confidence Level 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 c o Q_ (/) 0 ce c ro 0 7.8 7.6 7.4 7.2 6.8 6.6 6.4 14:5803/262010 70 80 Polynomial Model. (Version: 2.13; Date: 04/08/2008) Input Data File: C:\USEPA\lRlS\TCE\NTP\abslivwt\female\lin_abslivwtF2HDD_linear.(d) Gnuplot Plotting File: C:\USEPA\lRlS\TCE\NTP\abslivwt\female\lin_abslivwtF2HDD_linear.plt Fri Mar 26 15:58:54 2010 HMDS Model Run The form of the response function is: Y[dose] = beta 0 + beta l*dose + beta 2*doseA2 Dependent variable = mean Independent variable = dose rho is set to 0 The polynomial coefficients are restricted to be positive A constant variance model is fit Total number of dose groups = 4 Total number of records with missing values = 0 Maximum number of iterations = 250 Relative Function Convergence has been set to: le-008 B-17 DRAFT - DO NOT CITE OR QUOTE ------- 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 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 Parameter Convergence has been set to: le-008 Default Initial Parameter Values alpha = 0.195575 rho = 0 Specified beta_0 = 6.784 beta 1 = 0.0119571 Asymptotic Correlation Matrix of Parameter Estimates ( *** The model parameter(s) -rho have been estimated at a boundary point, or have been specified by the user, and do not appear in the correlation matrix ) alpha beta_0 beta 1 alpha 1 -8e-009 8.2e-009 beta_0 -8e-009 1 -0.76 beta_l 8.2e-009 -0.76 1 Variable alpha beta_0 beta 1 Estimate 0 .181435 6.784 0. 0119571 Parameter Estimates 95.0% Wald Confidence Interval Std. Err. Lower Conf. Limit Upper Conf. Limit 0.04057 0.101919 0.26095 0.104336 6.5795 6.9885 0.00227681 0.00749468 0.0164196 Table of Data and Estimated Values of Interest Dose N Obs Mean Est Mean Obs Std Dev Est Std Dev Scaled Res. 0 20 40 80 10 10 10 10 6.84 7. 03 7.14 7 .8 6.78 7. 02 7.26 7.74 0.54 0.41 0.51 0.25 0.426 0.426 0.426 0.426 0.416 0 .0509 -0. 908 0.441 Model Descriptions for likelihoods calculated Model Al: Yij = Mu(i) + e(ij) Var{e(ij) } = SigmaA2 Model A2: Yij = Mu(i) + e(ij) Var{e(ij) } = Sigma(i)A2 Model A3: Yij = Mu(i) + e(ij) Var{e(ij)} = SigmaA2 Model A3 uses any fixed variance parameters that were specified by the user Model R: Yi = Mu B-18 DRAFT - DO NOT CITE OR QUOTE ------- 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 Test 1 Test 2 Test 3 Test 4 (Note: Test Var{e (i) } = SigmaA2 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 Likelihoods of Interest Log (likelihood) 14 .743437 17 .781442 14 .743437 14.137196 3.648385 # Param' s 5 8 5 3 2 AIC -19.486874 -19.562884 -19.486874 -22.274391 -3.296770 Model Al A2 A3 fitted R Explanation of Tests Do responses and/or variances differ among Dose levels? (A2 vs. R) Are Variances Homogeneous? (Al vs A2) Are variances adequately modeled? (A2 vs. A3) Does the Model for the Mean Fit? (A3 vs. fitted) When rho=0 the results of Test 3 and Test 2 will be the same.) Tests of Interest -2*log (Likelihood Ratio) Test df p-value 28 .2661 6.07601 6.07601 1.21248 6 3 3 2 <.0001 0.108 0.108 0.5454 Test 1 Test 2 Test 3 Test 4 The p-value for Test 1 is less than .05. There appears to be a difference between response and/or variances among the dose levels It seems appropriate to model the data The p-value for Test 2 is greater than .1. model appears to be appropriate here The p-value for Test 3 is greater than .1. to be appropriate here The p-value for Test 4 is greater than .1. to adequately describe the data A homogeneous variance The modeled variance appears The model chosen seems B-19 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 Benchmark Dose Computation 3 4 Specified effect = 1 5 6 Risk Type = Estimated standard deviations from the control mean 7 8 Confidence level = 0.95 9 10 HMD = 35.6232 11 12 13 BMDL = 26.1046 14 15 16 Relative liver weights in male and female rats (Tables B-7 and B-8) 17 No model provided an adequate fit to the relative liver weight data in male rats even after 18 dropping the two highest dose groups. Therefore, these data are considered unsuitable for BMD 19 modeling. For the relative liver weight data in females, the assumption of constant variance was 20 satisfied and the power and 2- and 3-degree polynomial models provided adequate fit to the data 21 after the highest two dose groups were dropped. BMDL estimates across these models differed 22 by less than threefold. In accordance with U. S. EPA (2000), the model with the lowest AIC (3- 23 degree polynomial) was selected as the basis for the BMDiso and BMDLiso estimates of 22 and 24 15 mg/kg-day, respectively, for this endpoint. B-20 DRAFT - DO NOT CITE OR QUOTE ------- Table B-7. Summary of benchmark dose modeling results for relative liver weight in male rats Model Test for significant difference />-valuea Variance />-valueb Means />-valueb Scaled residuals of interest0 AIC BMD1SD (mg/kg- day) BMDL1SD (mg/kg- day) All dose groups included Constant variance Linear O.0001 <0.0001 O.0001 1.6/4.15 208.74 68.02 56.64 Non-constant variance Lineard O.0001 0.03 O.0001 1.93/4.36 208.89 55.05 37.77 Highest dose group dropped Constant variance Lineard O.0001 0.09 <0.0001 1.84/4.25 165.27 51.62 40.95 Non-constant variance Lineard O.0001 0.06 <0.0001 -0.79/-0.95 157.11 12.93 8.10 Two highest dose groups dropped Constant variance Lineard O.0001 0.07 0.15 0.25/-1.24 94.60 13.14 10.76 Non-constant variance Linear O.0001 0.08 0.09 0.35/-1.32 95.74 10.97 7.77 3 highest doses dropped Constant variance Linear O.0001 0.03 0.10 0.66/-1.32 74.39 12.16 9.27 Non-constant variance Hill6 Linear Polynomial (2-degree)d Power" NA O.0001 O.0001 O.0001 0.52 0.52 0.52 0.05 NA NA 0.45/-1.32 -0.07/0.12 -0.07/0.12 71.18 69.32 69.32 8.47 15.27 15.50 6.05 8.46 9.02 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; NA = not applicable (insufficient degrees of freedom to fit the model); SD = standard deviation aValues >0.05 fail to meet conventional goodness-of-fit criteria. Values <0.10 fail to meet conventional goodness-of-fit criteria. °Scaled residuals at doses immediately below and immediately above the benchmark dose. dCoefficients restricted to be positive. ePower restricted to >1. B-21 DRAFT - DO NOT CITE OR QUOTE ------- Table B-8. Summary of benchmark dose modeling results for relative liver weight in female rats Model Test for significant difference />-valuea Variance />-valueb Means />-valueb Scaled residuals of interest0 AIC BMD1SD (mg/kg-day) BMDL1SD (mg/kg-day) All dose groups included Constant variance Linear O.0001 <0.0001 0.01 -0.66/-1.01 181.20 36.16 30.95 Non-constant variance Lineard O.0001 0.01 O.0001 <-10/<-10 6.00 0.003 NA Highest dose group dropped Constant variance Lineard O.0001 0.002 <0.0001 -0.52/-1.19 129.06 26.16 21.87 Non-constant variance Lineard O.0001 0.01 0.001 -0.12/-0.30 123.73 16.52 12.39 Two highest dose groups dropped Constant variance Hille Lineard Polynomial (2 -degree) Polynomial (3-degree)A'f Power6 O.0001 O.0001 <0.0001 <0.0001 <0.0001 0.11 0.11 0.11 0.11 0.11 NA 0.005 0.22 0.38 0.15 1.12/-0.72 1.31/-0.09 0.94/-0.70 0.69/-0.43 1.12/-0.72 74.32 78.98 71.76 70.98 72.32 25.33 13.20 23.57 21.90 25.31 17.12 10.81 15.68 14.78 17.12 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; NA= not applicable (BMD/BMDL computation failed or insufficient degrees of freedom to fit model); SD = standard deviation aValues >0.05 fail to meet conventional goodness-of-fit criteria. bValues <0.10 fail to meet conventional goodness-of-fit criteria. °Scaled residuals at doses immediately below and immediately above the benchmark dose. Coefficients restricted to be positive. "Power restricted to >1. Best-fitting model is displayed in boldface type. BMDLs for models providing adequate fit differed by < threefold, so the model with the lowest AIC was selected. B-22 DRAFT - DO NOT CITE OR QUOTE ------- Polynomial Model with 0.95 Confidence Level 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 c o Q_ (/) 0 ce c ro 0 44 42 40 38 36 34 Polynomial BMDL BMD 10 20 30 40 dose 50 60 70 80 08:3403/292010 Polynomial Model. (Version: 2.13; Date: 04/08/2008) Input Data File: C:\USEPA\IRIS\TCE\NTP\rellivwt\female\ply_rellivwtF2HDD_Poly_3.(d) Gnuplot Plotting File: C:\USEPA\IRIS\TCE\NTP\rellivwt\female\ply_rellivwtF2HDD_Poly_3.pit Mon Mar 29 09:34:20 2010 HMDS Model Run The form of the response function is: Y[dose] = beta 0 + beta l*dose + beta 2*doseA2 Dependent variable = mean Independent variable = dose rho is set to 0 The polynomial coefficients are restricted to be positive A constant variance model is fit Total number of dose groups = 4 Total number of records with missing values = 0 Maximum number of iterations = 250 Relative Function Convergence has been set to: le-008 B-23 DRAFT - DO NOT CITE OR QUOTE ------- 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 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 Parameter Convergence has been set to: le-008 Default Initial Parameter Values beta_0 = beta 1 = alpha rho 35.07 0.115542 beta_2 beta 3 1.93677 0 0 = 2.848966-005 Specified Asymptotic Correlation Matrix of Parameter Estimates ( *** The model parameter(s) -rho -beta_2 have been estimated at a boundary point, or have been specified by the user, and do not appear in the correlation matrix ) alpha beta_0 beta_l beta_3 alpha 1 -6e-009 3.2e-oo9 -i.ve-009 beta_0 -6e-009 1 -0.76 0.56 beta_l 3.2e-009 -0.76 1 -0.92 beta_3 -1.76-009 0.56 -0.92 1 Parameter Estimates Variable alpha 1.77636 beta_0 35.1967 beta_l beta_2 beta 3 Estimate Std. Err. 0.397207 0.997852 0.395218 34.4221 0.0567055 0.0185417 1.598986-026 NA 8.68894e-006 2.57808e-006 95.0% Wald Confidence Interval Lower Conf. Limit Upper Conf. Limit 2 .55487 35.9713 0.0203645 0.0930465 3.636e-006 1.374196-005 NA - Indicates that this parameter has hit a bound implied by some inequality constraint and thus has no standard error. Table of Data and Estimated Values of Interest Dose N Obs Mean Est Mean Obs Std Dev Est Std Dev Scaled Res. 0 20 40 80 10 10 10 10 35 .1 36 .7 37 .8 44 .2 35 .2 36 .4 38 44 .2 1.77 1.14 1.61 0.85 1.33 1.33 1.33 1 .33 -0.301 0.687 -0 .43 0. 043 Model Descriptions for likelihoods calculated Model Al: Yij var{e(ij) } Mu(i) + e (ij) SigmaA2 B-24 DRAFT - DO NOT CITE OR QUOTE ------- 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 Model A2 : Yij = Mud) + e(ij) Var{e(ij)} = Sigma Model A3: Yij = Mu(i) + e(ij) Var{e (ij ) } = SigmaA2 Model A3 uses any fixed variance parameters that were specified by the user Model R: Yi = Mu + e (i) Var{e (i) } = SigmaA2 Likelihoods of Interest Model Al A2 A3 fitted R Log (likelihood) -31 .113274 -28.050020 -31 .113274 -31.491356 -72.394938 # Param's 5 8 5 4 2 AIC 72 .226548 72 .100041 72 .226548 70. 982711 148.789876 Explanation of Tests 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 Test 1: Do responses and/or variances differ (A2 vs. R) Test 2: Are Variances Homogeneous? (Al vs A2) Test 3: Are variances adequately modeled? (A2 Test 4: Does the Model for the Mean Fit? (A3 among Dose levels? vs. A3) vs. fitted) (Note: When rho=0 the results of Test 3 and Test 2 will be the same.) Tests of Interest Test -2*log (Likelihood Ratio) Test df Test 1 88.6898 6 Test 2 6.12651 3 Test 3 6.12651 3 Test 4 0.756163 1 The p-value for Test 1 is less than .05. There p-value <.0001 0.1056 0.1056 0.3845 appears to be a difference between response and/or variances among the dose levels It seems appropriate to model the data The p-value for Test 2 is greater than .1. A homogeneous variance model appears to be appropriate here The p-value for Test 3 is greater than .1. The to be appropriate here The p-value for Test 4 is greater than .1. The to adequately describe the data Benchmark Dose Computation Specified effect = 1 modeled variance appears model chosen seems B-25 DRAFT - DO NOT CITE OR QUOTE ------- 1 Risk Type = Estimated standard deviations from the control mean 2 3 Confidence level = 0.95 4 5 HMD = 21.8955 6 7 8 BMDL = 14.7785 9 10 11 Serum ALT activity in male and female rats (Tables B-9 and B-10) 12 All doses were retained in the BMD modeling of serum ALT in males and females. The 13 assumption of constant variance was not upheld for either dataset, but in each case, the power 14 model for variance built into the BMDS provided adequate fit to the variance data. With the 15 variance model applied, adequate fit to the means was provided by the Hill, power, and 2- and 5- 16 degree polynomial models for the males, and by the Hill model alone for the females. For the 17 males, estimated BMDLs from the adequately fitting models differed by less than threefold. In 18 accordance with U.S. EPA (2000), the model with the lowest AIC (i.e., 2-degree polynomial) 19 was selected as the basis for the BMDiso and BMDLiso estimates of 41 and 26 mg/kg-day. For 20 the females, BMDiso and BMDLiso estimates of 82 and 69 mg/kg-day were based on the Hill 21 model. B-26 DRAFT - DO NOT CITE OR QUOTE ------- Table B-9. Summary of benchmark dose modeling results for serum ALT activity in male rats Model Test for significant difference />-valuea Variance />-valueb Means />-valueb Scaled residuals of interest0 AIC BMD1SD (mg/kg- day) BMDL1SD (mg/kg- day) All dose groups included Constant variance Linear O.0001 <0.0001 O.0001 -0.19/-1.55 486.88 43.91 37.37 Non-constant variance Hille Linear Polynomial (2-degree) ' Polynomial (3 -degree) Polynomial (4-degree)d Polynomial (5-degree)d Power6 O.0001 O.0001 <0.0001 O.0001 O.0001 <0.0001 <0.0001 0.72 0.72 0. 72 0.72 0.72 0.72 0.72 0.51 O.0001 0.84 <0.0001 <0.0001 0.47 0.73 0.10/0.77 >10 -0.21/1.00 >10 NA -0.14/1.06 -0.11/0.76 370.02 6.00 366.08 10.00 606.63 370.17 367.96 42.19 0.00 40.98 0.00 NA 40.62 41.97 34.33 NA 26.35 NA 28.22 26.19 32.24 Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; NA= not applicable (BMD/BMDL computation failed); SD = standard deviation aValues >0.05 fail to meet conventional goodness-of-fit criteria. Values <0.10 fail to meet conventional goodness-of-fit criteria. °Scaled residuals at doses immediately below and immediately above the benchmark dose. Coefficients restricted to be positive. ePower restricted to >1. fBest-fitting model is displayed in boldface type. BMDLs for models providing adequate fit differed by < threefold, so the model with the lowest AIC was selected. DRAFT - DO NOT CITE OR QUOTE ------- Polynomial Model with 0.95 Confidence Level 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 c o Q_ (/) 0 ce c ro 0 350 300 250 200 150 100 50 Polynomial BMDL 3MD 50 100 150 dose 200 250 300 09:5903/292010 Polynomial Model. (Version: 2.13; Date: 04/08/2008) input Data File: C:\USEPA\lRlS\TCE\NTP\ALT\male\ply_ALTM_poly_2.(d) Gnuplot Plotting File: C:\USEPA\IRIS\TCE\NTP\ALT\male\ply_ALTM_poly_2.pit Mon Mar 29 10:59:45 2010 HMDS Model Run The form of the response function is: Y[dose] = beta 0 + beta l*dose + beta 2*doseA2 + ... Dependent variable = mean Independent variable = dose The polynomial coefficients are restricted to be positive The variance is to be modeled as Var(i) = exp(lalpha + log(mean(i)) * rho) Total number of dose groups = 6 Total number of records with missing values = 0 Maximum number of iterations = 250 Relative Function Convergence has been set to: le-008 Parameter Convergence has been set to: le-008 B-28 DRAFT - DO NOT CITE OR QUOTE ------- 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 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 Default Initial Parameter Values lalpha = 6.52437 rho = 0 beta_0 = 48.8991 beta 1 = 0.00912505 beta_2 = 0.00233971 ! ! ! Warning: optimum may not have been found. ! ! ! !!! You may want to try choosing different initial values. !!! Asymptotic Correlation Matrix of Parameter Estimates ( *** The model parameter (s) -rho have been estimated at a boundary point, or have been specified and do not appear in the correlation matrix ) lalpha beta 0 beta 1 beta 2 lalpha 1 -0.0021 -0.015 0.027 beta_0 -0.0021 1 -0.71 0.49 beta_l -0.015 -0.71 1 -0.86 beta_2 0.027 0.49 -0.86 1 Parameter Estimates 95.0% Wald Confidence I Variable Estimate Std. Err. Lower Conf. Limit Upper lalpha -6.58334 0.182468 -6.94097 rho 2.62555 NA beta_0 47.7312 1.57297 44.6483 beta 1 0.05625 0.0541054 -0.0497946 beta_2 0.00216953 0.000281829 0.00161716 NA - Indicates that this parameter has hit a bound implied by some inequality constraint and thus has no standard error. Table of Data and Estimated Values of Interest Dose N Obs Mean Est Mean Obs Std Dev Est Std Dev Scaled Res. 0 10 48 47.7 6.3 5.95 0.143 20 10 49 49.7 6.3 6.28 -0.365 40 10 53 53.5 6.3 6.9 -0.207 80 10 69 66.1 9.5 9.12 1 170 10 115 120 25.3 19.9 -0.792 320 10 292 288 56.9 62.9 0.206 Model Descriptions for likelihoods calculated Model Al: Yij = Mu(i) + e(ij) Var{e (ij ) } = SigmaA2 by the user nterval Conf. Limit -6 .22571 50 .8142 0.162295 0 .0027219 B-29 DRAFT - DO NOT CITE OR QUOTE ------- 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 Model A2 : Yij = Mud) + e(ij) Var{e(ij)} = Sigma Model A3: Yij = Mu(i) + e(ij) Var{e(ij)} = expdalpha + rho*ln (Mu (i) ) ) Model A3 uses any fixed variance parameters that were specified by the user Model R: Yi = Mu + e (i) Var{e (i) } = SigmaA2 Likelihoods of Interest Model Al A2 A3 fitted R Log (likelihood) -222 .570247 -177 .293103 -178 .329731 -179.039110 -300.315008 # Param's 7 12 8 4 2 AIC 459. 140493 378.586206 372.659462 366.078220 604.630016 Explanation of Tests 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 Test 1: Do responses and/or variances differ among Dose levels? (A2 vs. R) Test 2: Are Variances Homogeneous? (Al vs A2) Test 3: Are variances adequately modeled? (A2 vs. A3) Test 4: Does the Model for the Mean Fit? (Note: When rho=0 the results of Test 3 Tests of Interest Test -2*log (Likelihood Ratio) Test Test 1 246.044 10 Test 2 90.5543 5 Test 3 2.07326 4 Test 4 1.41876 4 The p-value for Test 1 is less than .05. (A3 vs. fitted) and Test 2 will be the same.) df p-value <.0001 <.0001 0.7223 0.8409 There appears to be a difference between response and/or variances among the dose levels It seems appropriate to model the data The p-value for Test 2 is less than .1. A model appears to be appropriate The p-value for Test 3 is greater than .1. to be appropriate here The p-value for Test 4 is greater than .1. to adequately describe the data Benchmark Dose Computation Specified effect = 1 Risk Type = Estimated standard non-homogeneous variance The modeled variance appears The model chosen seems deviations from the control me. B-30 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 Confidence level = 0.95 3 4 HMD = 40.9754 5 6 7 BMDL = 26.3459 B-31 DRAFT - DO NOT CITE OR QUOTE ------- Table B-10. Summary of benchmark dose modeling results for serum ALT activity in female rats Model Test for significant difference />-valuea Variance />-valueb Means />-valueb Scaled residuals of interest0 AIC BMD1SD (mg/kg- day) BMDL1SD (mg/kg- day) All dose groups included Constant variance Lineard O.0001 <0.0001 O.0001 -0.12/2.54 512.92 45.04 38.30 Non-constant variance HUf Lineard Polynomial (2 -degree/ Polynomial (3 -degree) Polynomial (4-degree)d Polynomial (5-degree)d Power6 <0.0001 <0.0001 <0.0001 O.0001 <0.0001 <0.0001 <0.0001 0.23 0.23 0.23 0.23 0.23 0.23 0.23 0.16 <0.0001 <0.0001 O.0001 <0.0001 <0.0001 0.02 0.09/-0.29 0.79/3.84 -0.91/-0.16 -0.95/-0.20 -0.77/-0.40 -0.85/-0.14 -0.26/-1.58 351.50 444.14 413.32 415.39 392.73 432.77 355.84 82.49 142.23 65.95 71.30 71.75 79.16 64.07 68.61 12.12 19.55 15.90 22.50 13.16 55.45 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; SD = standard deviation aVallies >0.05 fail to meet conventional goodness-of-fit criteria. bValues <0.10 fail to meet conventional goodness-of-fit criteria. °Scaled residuals at doses immediately below and immediately above the benchmark dose. , J J Coefficients restricted to be positive. ePower restricted to >1. Best-fitting model is displayed in boldface type. In this case, Hill model was the only model that provided an adequate fit to the data. B-32 DRAFT - DO NOT CITE OR QUOTE ------- Hill Model with 0.95 Confidence Level 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 c o Q_ (/) 0 ce c ro 0 400 350 300 250 200 150 100 50 300 10:0803/292010 Hill Model. (Version: 2.14; Date: 06/26/2008) Input Data File: C:\USEPA\IRIS\TCE\NTP\ALT\female\hil_ALTF_Hill.(d) Gnuplot Plotting File: C:\USEPA\IRIS\TCE\NTP\ALT\female\hil_ALTF_Hill.pit Mon Mar 29 11:08:43 2010 HMDS Model Run The form of the response function is: Y [dose] = intercept + v*doseAn/(kAn + dose^n) Dependent variable = mean Independent variable = dose Power parameter restricted to be greater than 1 The variance is to be modeled as Var(i) = exp(lalpha + rho * In(mean(i))) Total number of dose groups = 6 Total number of records with missing values = 0 Maximum number of iterations = 250 Relative Function Convergence has been set to: le-008 Parameter Convergence has been set to: le-008 B-33 DRAFT - DO NOT CITE OR QUOTE ------- 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 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 Default Initial Parameter Values lalpha = rho = intercept = v = n = k = 6 .46604 0 46 293 2 . 07344 416.806 Asymptotic Correlation Matrix of Parameter Estimates lalpha rho intercept lalpha 1 -0 .99 -0 .12 0.1 -0 .0074 0. 051 rho -0 .99 1 0. 098 -0 .11 0 .0073 -0. 052 intercept -0 .12 0. 098 1 -0 .41 0 .49 -0 .42 V 0 .1 -0.11 -0.41 1 -0.9 0 .98 n -0. 0074 0. 0073 0.49 -0 .9 1 -0 .95 k 0 .051 -0 .052 -0.42 0. 98 -0. 95 1 Parameter Estimates Variable lalpha rho intercept V n k Estimate -5.48513 2.36002 43 .8372 440. 049 3.71466 266.476 Std. Err. 1.18231 0 .272384 1. 06856 121.144 0 .661842 45 .4588 95.0% Wald Confidence Interval Lower Conf. Limit Upper Conf. Limit -7.80242 -3.16783 1.82615 2.89388 41.7428 45.9315 202.612 677.486 2.41747 5.01185 177.378 355.573 Table of Data and Estimated Values of Interest Dose N Obs Mean Est Mean Obs Std Dev Est Std Dev Scaled Res. 0 20 40 80 170 320 10 10 10 10 10 10 46 42 44 49 112 339 43 .8 43 .9 44 .2 48 .8 114 336 6 .3 3.2 6.3 6.3 22 .1 56.9 5 .58 5 .58 5 .63 6 .33 17.1 61.6 1 .23 -1 .06 -0.124 0 .0904 -0 .29 0.159 Model Descriptions for likelihoods calculated Model Al: Yij = Mu(i) + e(ij) Var{e(ij) } = SigmaA2 Model A2: Yij = Mu(i) + e(ij) Var{e(ij) } = Sigma(i)A2 Model A3: Yij = Mu(i) + e(ij) Var{e(ij)} = exp(lalpha + rho*ln(Mu(i))) Model A3 uses any fixed variance parameters that B-34 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 6 7 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 we Model Test 1 Test 2 Test 3 Test 4 (Note: 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 were specified by the user Yi = Mu + e(i) Var{e(i)} = SigmaA2 Likelihoods of Interest Log (likelihood) -220.820465 -165 . 059425 -167.889045 -169.749216 -312 . 021870 # Param' s 7 12 8 6 2 AIC 455. 640931 354. 118851 351.778089 351.498431 628. 043741 Model Al A2 A3 fitted R Explanation of Tests Do responses and/or variances differ among Dose levels? (A2 vs. R) Are Variances Homogeneous? (Al vs A2) Are variances adequately modeled? (A2 vs. A3) Does the Model for the Mean Fit? (A3 vs. fitted) When rho=0 the results of Test 3 and Test 2 will be the same.) Tests of Interest -2*log(Likelihood Ratio) Test df p-value Test Test 1 Test 2 Test 3 Test 4 The p-value for Test 1 is less than .05. There appears to be a difference between response and/or variances among the dose levels It seems appropriate to model the data 293.925 111.522 5. 65924 3 . 72034 10 5 4 2 <.0001 <.0001 0.2261 0.1556 The p-value for Test 2 is less than .1. model appears to be appropriate A non-homogeneous variance The p-value for Test 3 is greater than .1. The modeled variance appears to be appropriate here The p-value for Test 4 is greater than .1. to adequately describe the data The model chosen seems B-35 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 Benchmark Dose Computation 3 4 Specified effect = 1 5 6 Risk Type = Estimated standard deviations from the control mean 7 8 Confidence level = 0.95 9 10 HMD = 82.493 11 12 BMDL = 68.6138 13 14 15 Serum SDH activity in male and female rats (Tables B-ll and B-12) 16 No model provided an adequate fit to the data for changes in serum SDH activity in male 17 rats. This was due to the difficulty in modeling the reported variances. As a result, these data 18 are considered unsuitable for BMD modeling. For females, only the power model provided an 19 adequate fit to the serum SDH activity data after the highest dose was dropped and the variance 20 was modeled using the non-constant variance model included in BMDS. This model served as 21 the basis for the BMDiso and BMDLiso estimates of 157 and 113 mg/kg-day for this endpoint. B-3 6 DRAFT - DO NOT CITE OR QUOTE ------- Table B-ll. Summary of benchmark dose modeling results for serum SDH activity in male rats Model Test for significant difference />-valuea Variance />-valueb Means />-valueb Scaled residuals of interest0 AIC BMD1SD (mg/kg- day) BMDL1SD (mg/kg- day) All dose groups included Constant variance Linear O.0001 O.0001 0.19 -0.75/-1.42 293.96 41.70 35.55 Non-constant variance Lineard O.0001 0.05 O.0001 -0.92/0.60 307.18 62.52 11.14 Highest dose group dropped Constant variance Lineard O.0001 0.02 0.08 1.33/-1.16 212.18 34.45 28.37 Non-constant variance Lineard O.0001 0.03 0.05 1.09/-1.28 212.07 32.47 19.12 Two Highest dose groups dropped Constant variance Lineard 0.0004 0.04 0.26 -0.92/0.15 159.19 45.73 31.69 Non-constant variance Linear 0.0004 0.03 0.17 -0.91/0.13 161.04 42.28 25.15 Three highest dose groups dropped Constant variance Linear 0.03 0.04 0.14 -0.60e 125.02 58.79 27.97 Non-constant variance Lineard 0.03 0.05 0.64 1.20/-0.82 122.10 27.88 13.75 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; SD = standard deviation aValues >0.05 fail to meet conventional goodness-of-fit criteria. Values <0.10 fail to meet conventional goodness-of-fit criteria. °Scaled residuals at doses immediately below and immediately above the benchmark dose. Coefficients restricted to be positive. eResidual reported for highest dose tested. B-37 DRAFT - DO NOT CITE OR QUOTE ------- Table B-12. Summary of benchmark dose modeling results for serum SDH activity in female rats Model Test for significant difference />-valuea Variance />-valueb Means />-valueb Scaled residuals of interest0 AIC BMD1SD (mg/kg- day) BMDL1SD (mg/kg- day) All dose groups included Constant variance Linear O.0001 <0.0001 O.0001 0.18/-3.60 321.64 47.70 40.47 Non-constant variance Lineard O.0001 0.04 O.0001 NA 432.91 NA 24.11 Highest dose group dropped Constant variance Lineard O.0001 0.0002 0.0001 -0.05/-3.48 244.99 63.45 48.93 Non-constant variance Hille Lineard Polynomial (2 -degree) Polynomial (3-degree)d Polynomial (4-degree)d Power*'* <0.0001 <0.0001 <0.0001 <0.0001 O.0001 <0.0001 0.18 0.18 0.18 0.18 0.18 0.18 0.05 0.00 0.00 0.01 0.04 0.10 -1.34/0.00 -0.09/-2.36 -2.77/1.04 -2.19/0.42 -1.78/0.17 -1.34/0.00 217.37 229.76 224.39 219.90 217.52 215.37 153.80 67.45 87.97 106.18 118.22 156.52 NA 38.00 66.87 87.33 102.34 113.49 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; NA = not applicable (BMD/BMDL computation failed); SD = standard deviation aValues >0.05 fail to meet conventional goodness-of-fit criteria. Values <0.10 fail to meet conventional goodness-of-fit criteria. °Scaled residuals at doses immediately below and immediately above the benchmark dose. Coefficients restricted to be positive. "Power restricted to >1. fBest-fitting model is displayed in boldface type. Power model was the only model that provided an adequate fit to the data. B-38 DRAFT - DO NOT CITE OR QUOTE ------- Power Model with 0.95 Confidence Level 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 c o Q_ (/) 0 ce c ro 0 50 45 40 35 30 25 20 L Power BMDL BMD 0 20 40 60 80 dose 100 120 140 160 14:2003/292010 Power Model. (Version: 2.15; Date: 04/07/2008) Input Data File: C:\USEPA\IRIS\TCE\NTP\SDH\female\pOW_SDHFHDD_pOwer.(d) Gnuplot Plotting File: C:\USEPA\IRIS\TCE\NTP\SDH\female\pOW_SDHFHDD_pOwer.plt Mon Mar 29 15:20:23 2010 HMDS Model Run The form of the response function is: Y[dose] = control + slope * dose^power Dependent variable = mean Independent variable = dose The power is restricted to be greater than or equal to 1 The variance is to be modeled as Var(i) = exp(lalpha + log(mean(i)) * rho) Total number of dose groups = 5 Total number of records with missing values = 0 Maximum number of iterations = 250 Relative Function Convergence has been set to: le-008 B-39 DRAFT - DO NOT CITE OR QUOTE ------- 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 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 Parameter Convergence has been set to: le-008 Default Initial Parameter Values lalpha = 3.46985 rho = 0 control = 25 Slope = 0.0617409 power = 1.1118 Asymptotic Correlation Matrix of Parameter Estimates ( *** The model parameter (s) -power have been estimated at a boundary point, or have been specified and do not appear in the correlation matrix ) lalpha rho control slope lalpha 1 -1 -0.15 0.37 rho -1 1 0.14 -0.37 control -0.15 0.14 1 -0.22 Slope 0.37 -0.37 -0.22 1 Parameter Estimates 95.0% Wald Confidence I Variable Estimate Std. Err. Lower Conf. Limit Upper lalpha -7.0365 3.52075 -13.937 rho 3.00361 1.03813 0.968917 control 26.75 0.652491 25.4711 slope 1.29772e-039 2.07902e-040 8.90244e-040 1. power 18 NA NA - Indicates that this parameter has hit a bound implied by some inequality constraint and thus has no standard error. Table of Data and Estimated Values of Interest Dose N Obs Mean Est Mean Obs Std Dev Est Std Dev Scaled Res. 0 10 27 26.7 3.2 4.13 0.192 20 10 27 26.7 3.2 4.13 0.192 40 10 28 26.7 6.3 4.13 0.958 80 10 25 26.8 3.2 4.13 -1.34 170 10 45 45 9.5 9.01 3.88e-006 Model Descriptions for likelihoods calculated Model Al: Yij = Mu(i) + e(ij) Var{e (ij) } = SigmaA2 by the user nterval Conf. Limit -0.135945 5 .0383 28.0289 7052e-039 B-40 DRAFT - DO NOT CITE OR QUOTE ------- 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 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 Model A2 : Yij = Mud) + e(ij) Var{e(ij)} = Sigma Model A3: Yij = Mu(i) + e(ij) Var{e(ij)} = expdalpha + rho*ln (Mu (i) ) ) Model A3 uses any fixed variance parameters that were specified by the user Model R: Yi = Mu + e (i) Var{e (i) } = SigmaA2 Likelihoods of Interest Model Log(likelihood) Al -109.112298 A2 -98.178926 A3 -100.610596 fitted -103.685379 R -135.518801 Explanation of Tests Test 1: Do responses and/or variances differ among Dose levels? (A2 vs. R) Test 2: Are Variances Homogeneous? (Al vs A2) Test 3: Are variances adequately modeled? (A2 vs. A3) Test 4: Does the Model for the Mean Fit? (A3 vs. fitted) (Note: When rho=0 the results of Test 3 and Test 2 will be the same.) # Param' s 6 10 7 4 2 AIC 230.224595 216.357851 215.221192 215.370759 275.037602 Test Test 1 Test 2 Test 3 Test 4 Tests of Interest -2*log (Likelihood Ratio) Test df 74.6798 21.8667 4.86334 6. 14957 p-value <.0001 0.000213 0.1821 0.1046 The p-value for Test 1 is less than .05. There appears to be a difference between response and/or variances among the dose levels It seems appropriate to model the data The p-value for Test 2 is less than .1. model appears to be appropriate A non-homogeneous variance The p-value for Test 3 is greater than .1. The modeled variance appears to be appropriate here The p-value for Test 4 is greater than .1. to adequately describe the data The model chosen seems Benchmark Dose Computation Specified effect = 1 Risk Type = Estimated standard deviations from the control mean Confidence level = 0.95 B-41 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 HMD = 156.523 3 4 5 BMDL = 113.491 6 7 Serum bile acids in male and female rats (Tables B-13 and B-14) 8 All doses were retained in the modeling of serum bile acids in males and females. The 9 assumption of constant variance was not upheld for either dataset, but in each case, the power 10 model for variance included in BMDS provided adequate fit to the variance data. With the 11 variance model applied, adequate fit to the mean data was provided by several models for each 12 sex, and for both datasets, BMDL estimates across models with adequate fit differed by less than 13 threefold. In accordance with U.S. EPA (2000), the models with the lowest AIC (power model 14 for males and 5-degree polynomial model for females) were selected as the basis for the BMDiso 15 and BMDLiso estimates for these endpoints (respectively, 72 and 57 mg/kg-day for males and 16 188 and 170 mg/kg-day for females). 17 B-42 DRAFT - DO NOT CITE OR QUOTE ------- Table B-13. Summary of benchmark dose modeling results for serum bile acid levels in male rats Model Test for significant difference />-valuea Variance />-valueb Means />-valueb Scaled residuals of interest0 AIC BMD1SD (mg/kg- day) BMDL1SD (mg/kg- day) All dose groups included Constant variance Linear O.0001 <0.0001 0.002 -0.10/-1.38 578.68 76.00 62.75 Non-constant variance Hille Linear Polynomial (2-degree)d Polynomial (3-degree)d Polynomial (4-degree) Polynomial (5-degree)d Power'* O.0001 O.0001 O.0001 <0.0001 <0.0001 <0.0001 <0.0001 0.77 0.77 0.77 0.77 0.77 0.77 0.77 0.69 O.0001 0.21 0.32 0.32 O.0001 0.46 0.17/-0.74 0.48/2.69 -0.88/-1.16 -0.65/-0.56 -0.65/-0.56 -1.08/0.17 -0.56/-0.43 427.84 454.67 428.95 428.58 428.58 449.32 427.70 82.84 115.63 58.37 69.21 69.21 76.72 72.45 66.69 36.05 50.80 54.31 54.31 25.65 57.77 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; SD = standard deviation aValues >0.05 fail to meet conventional goodness-of-fit criteria. Values <0.10 fail to meet conventional goodness-of-fit criteria. °Scaled residuals at doses immediately below and immediately above the benchmark dose. Coefficients restricted to be positive. "Power restricted to >1. fBest-fitting model is displayed in boldface type. BMDLs for models providing adequate fit differed by < threefold, so the model with the lowest AIC was selected. B-43 DRAFT - DO NOT CITE OR QUOTE ------- Power Model with 0.95 Confidence Level 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 c o Q_ (/) 0 ce c ro 0 400 300 200 100 300 14:3903/292010 Power Model. (Version: 2.15; Date: 04/07/2008) input Data File: C:\USEPA\lRlS\TCE\NTP\bile\male\pow_BileM_power.(d) Gnuplot Plotting File: C:\USEPA\lRlS\TCE\NTP\bile\male\pow_BileM_power.plt Mon Mar 29 15:39:39 2010 HMDS Model Run The form of the response function is: Y [dose] = control + slope * dose^power Dependent variable = mean Independent variable = dose The power is restricted to be greater than or equal to 1 The variance is to be modeled as Var(i) = exp(lalpha + log(mean(i)) * rho) Total number of dose groups = 6 Total number of records with missing values = 0 Maximum number of iterations = 250 Relative Function Convergence has been set to: le-008 Parameter Convergence has been set to: le-008 B-44 DRAFT - DO NOT CITE OR QUOTE ------- 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 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 Default Initial Parameter Values lalpha = rho = control = slope = power = 8.35885 0 27.2 0.000160062 2.50584 Asymptotic Correlation Matrix of Parameter Estimates lalpha lalpha rho control slope power -0. -0. -0. 0. 1 .98 .31 .17 .22 rho -0. 0. 0. -0. .98 1 .25 .18 .23 control -0.31 0.25 1 -0.3 0.28 Slope -0.17 0.18 -0.3 1 -1 power 0 .22 -0.23 0.28 -1 1 Variable lalpha rho control slope power Estimate -3.601 2.39924 26.8064 0.000289806 2.40282 Parameter Estimates 95.0% Wald Confidence Interval Std. Err. Lower Conf. Limit Upper Conf. Limit 1.08576 -5.72905 -1.47295 0.272426 1.86529 2.93318 1.58205 23.7056 29.9071 0.000360688 -0.00041713 0.000996743 0.233505 1.94515 2.86048 Table of Data and Estimated Values of Interest Dose N Obs Mean Est Mean Obs Std Dev Est Std Dev Scaled Res. 0 20 40 80 170 320 10 10 10 10 10 10 29.2 27 .5 27 .2 35 .9 92 332 26 .8 27 .2 28 .9 37 .6 93 .1 330 9.2 8 .5 8 .5 12 .3 52 .5 150 8.54 8.69 9.33 12 .8 38 173 0 .886 0 .111 -0 .561 -0 .429 -0. 0914 0. 0463 Model Descriptions for likelihoods calculated Model Al: Yij var{e(ij) } Model A2: Yij var{e(ij)} Model A3: Yij var{e(ij)} Mu(i) + e (ij) SigmaA2 Mu(i) + e (ij) Sigma(i)A2 Mu(i) + e (ij) exp(lalpha + rho*ln(Mu(i) B-45 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 6 7 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 MO we Model Test 1 Test 2 Test 3 Test 4 (Note: 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 Model A3 uses any fixed variance parameters that were specified by the user R: Yi = Mu + e(i) Var{e(i)} = SigmaA2 Likelihoods of Interest Log (likelihood) -277.604668 -206.636351 -207.553828 -208.851786 -320.497188 # Param' s 7 12 8 5 2 AIC 569.209336 437.272702 431. 107657 427. 703572 644. 994376 Model Al A2 A3 fitted R Explanation of Tests Do responses and/or variances differ among Dose levels? (A2 vs. R) Are Variances Homogeneous? (Al vs A2) Are variances adequately modeled? (A2 vs. A3) Does the Model for the Mean Fit? (A3 vs. fitted) When rho=0 the results of Test 3 and Test 2 will be the same.) Test Test 1 Test 2 Test 3 Test 4 Tests of Interest -2*log (Likelihood Ratio) Test df 227.722 141. 937 1.83495 2 . 59591 10 5 4 3 p-value <.0001 <.0001 0.7661 0.4582 The p-value for Test 1 is less than .05. There appears to be a difference between response and/or variances among the dose levels It seems appropriate to model the data The p-value for Test 2 is less than .1. model appears to be appropriate A non-homogeneous variance The p-value for Test 3 is greater than .1. The modeled variance appears to be appropriate here The p-value for Test 4 is greater than .1. to adequately describe the data The model chosen seems B-46 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 Benchmark Dose Computation Specified effect = 1 Risk Type = Estimated standard deviations from the control mean Confidence level = 0.95 HMD = 72.4471 BMDL = 57.1682 Table B-14. Summary of benchmark dose modeling results for serum bile acid levels in female rats Model Test for significant difference p- valuea Variance />-valueb Means />-valueb Scaled residuals of interest0 AIC BMD1SD (mg/kg- day) BMDL1SD (mg/kg- day) All dose groups included Constant variance Lineard O.0001 O.0001 O.0001 -1.13/-3.83 596.57 101.36 81.28 Non-constant variance Hill6 Lineard Polynomial (2 -degree/ Polynomial (3 -degree) Polynomial (4-degree)d Polynomial (5-degree) 'g Powere O.0001 O.0001 O.0001 O.0001 O.0001 <0.0001 O.0001 0.47 0.47 0.47 0.47 0.47 0.47 0.47 0.38 O.0001 <0.0001 0.003 0.08 0.33 0.38 -0.51/0.02 3.70f 3.09f -0.71/-2.18 -0.42/-1.95 -1.34/0.34 -0.50/0.02 466.68 505.52 485.36 477.39 469.90 466.14 466.68 186.94 343.48 344.76 149.70 168.35 187.71 216.74 177.64 139.12 145.95 129.07 152.78 169.55 177.00 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; SD = standard deviation aValues >0.05 fail to meet conventional goodness-of-fit criteria. Values <0.10 fail to meet conventional goodness-of-fit criteria. °Scaled residuals at doses immediately below and immediately above the benchmark dose. Coefficients restricted to be positive. "Power restricted to >1. fResidual at highest dose tested. 8Best-fitting model is displayed in boldface type. BMDLs for models providing adequate fit differed by < threefold, so the model with the lowest AIC was selected. 16 B-47 DRAFT - DO NOT CITE OR QUOTE ------- Polynomial Model with 0.95 Confidence Level 1 2 ^ ^J 4 5 450 400 350 300 8 c EL 250 0 ce i 200 0 150 100 50 0 '• rl_i,.___l:li Polynomial - - - - . - • - ' T T -r- ^- 1 I : BMDL 0 50 100 150 dose 14:4703/292010 1 ' ' / / ^/ - - - - - - - BMD : 200 250 300 Polynomial Model. (Version: 2.13; Date: 04/08/2008) Input Data File: 6 C:\USEPA\lRlS\TCE\NTP\bile\female\ply BileF Poly 5.(d) 1 8 9 10 _L *_* 11 12 13 _L —* 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 Gnuplot Plotting File: C:\USEPA\lRlS\TCE\NTP\bile\female\ply BileF Poly 5. pit Mon Mar 29 15:47:49 2010 BMDS Model Run The form of the response function is: Y[dose] = beta 0 + beta l*dose + beta 2*doseA2 + ... Dependent variable = mean Independent variable = dose The polynomial coefficients are restricted to be positive The variance is to be modeled as Var(i) = exp(lalpha + log (mean (i)) Total number of dose groups = 6 Total number of records with missing values Maximum number of iterations = 250 = 0 * rho) Relative Function Convergence has been set to: le-008 Parameter Convergence has been set to: le-008 B-48 DRAFT - DO NOT CITE OR QUOTE ------- 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 43 44 45 46 47 48 49 50 Default Initial Parameter Values lalpha = 8.43454 rho = 0 beta_0 = 37 beta_l = 0 beta_2 = 0 beta_3 = 0 beta_4 = 0 beta 5 = 0 Asymptotic Correlation Matrix of Parameter Estimates ( *** The model parameter(s) -beta_l -beta_2 -beta_3 -beta_4 have been estimated at a boundary point, or have been specified by the user, and do not appear in the correlation matrix ) lalpha lalpha 1 rho -0.98 beta_0 -0.049 beta 5 0.16 rho beta_0 -0.98 -0.049 1 0.049 0.049 1 -0.16 -0.15 beta_5 0. -0. -0. .16 .16 .15 1 Parameter Estimates Variable lalpha rho beta_0 beta 1 beta_2 beta 3 beta_4 beta 5 Estimate -1.58198 2. 03725 38 .2101 1.251286-026 0 0 0 7. 95519e-011 Std. Err. 1. 00675 0 .245366 2.76802 NA NA NA NA 1.432946-011 95.0% Wald Confidence Interval Lower Conf. Limit Upper Conf. Limit -3.55517 0.391218 1.55634 2.51816 32.7849 43.6353 5.14667e-011 1.076376-010 NA - Indicates that this parameter has hit a bound implied by some inequality constraint and thus has no standard error. B-49 DRAFT - DO NOT CITE OR QUOTE ------- 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 Table of Data and Estimated Values of Interest Dose N Obs Mean Est Mean Obs Std Dev Est Std Dev 0 10 20 10 40 10 80 10 170 10 320 10 37 46 .6 39.1 36 .3 39.3 322 Model Descriptions for Model Al: Model A2 : Model A3 : Yij = var{e(ij)} = Yij = var{e(ij)} = Yij = var{e(ij)} = 38.2 22.5 18.5 38.2 20.6 18.5 38.2 17.7 18.5 38.5 12.3 18.7 49.5 25 24.1 305 160 154 likelihoods calculated Mu (i ) + e (i j ) SigmaA2 Mu (i ) + e (i j ) Sigma (i) A2 Mu (i ) + e (i j ) expdalpha + rho*ln (Mu (i) ) ) Scaled Res -0 .206 1 .43 0 .15 -0 .368 -1 .34 0.336 26 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 Model R Test 1: Test 2: Test 3: Test 4: (Note: i Test Test 1 Test 2 Test 3 Test 4 The p-vali difference Model A3 uses any fixed variance parameters that were specified by the user Yi = Mu + e(i) Var{e(i)} = SigmaA2 Likelihoods of Interest Log (likelihood) -279 .875470 -224.999384 -226.787639 -229.071113 -318.845182 # Param' s 7 12 8 4 2 AIC 573 . 750939 473 . 998768 469. 575277 466. 142225 641.690364 Model Al A2 A3 fitted R Explanation of Tests Do responses and/or variances differ among Dose levels? (A2 vs. R) Are Variances Homogeneous? (Al vs A2) Are variances adequately modeled? (A2 vs. A3) Does the Model for the Mean Fit? (A3 vs. fitted) When rho=0 the results of Test 3 and Test 2 will be the same.) Tests of Interest -2*log(Likelihood Ratio) Test df p-value 187.692 109.752 3.57651 4.56695 10 5 4 4 <.0001 <.0001 0.4663 0.3347 The p-value for Test 1 is less than .05. There appears to be a difference between response and/or variances among the dose levels B-50 DRAFT - DO NOT CITE OR QUOTE ------- 1 It seems appropriate to model the data 2 3 The p-value for Test 2 is less than . 1. A non-homogeneous variance 4 model appears to be appropriate 5 6 The p-value for Test 3 is greater than .1. The modeled variance appears 7 to be appropriate here 8 9 The p-value for Test 4 is greater than .1. The model chosen seems 10 to adequately describe the data 11 12 13 Benchmark Dose Computation 14 15 Specified effect = 1 16 17 Risk Type = Estimated standard deviations from the control mean 18 19 Confidence level = 0.95 20 21 HMD = 187.713 22 23 24 BMDL = 169.553 25 26 27 Fetal body weights in Sprague-Dawley rats (Tables B-15 and B-16) 28 Fetal body weight data from Gulati et al. (1991) in Sprague-Dawley rats administered 29 1,1,2,2-tetrachloroethane in the diet on GD 4 - 20 are shown in Table B-15. BMD modeling 30 results based on these data are shown in Table B-l6. Adequate model fit was achieved for the 31 fetal body weight data only after the highest two dose groups were dropped. This was due to 32 difficulty in modeling the reported variances. After dropping the two highest dose groups, the 33 remaining dose groups satisfied the assumption of constant variance. Assuming constant 34 variance, the linear model provided adequate fit to the mean fetal body weight data. The higher 35 order models either did not fit (p < 0.1: higher order polynomial, power) or failed due to too 36 many parameters for the available data points (Hill). The linear model is the basis for the 37 BMDiso and BMDLiso estimates of 83 and 60 mg/kg-day, respectively, for this endpoint shown 38 in Table B-l6. B-51 DRAFT - DO NOT CITE OR QUOTE ------- Table B-15. Fetal body weight in Sprague-Dawley rats administered 1,1^2,2-tetrachloroethane in the diet on gestation days 4-20 Dose (mg/kg-day) 0 34 98 180 278 330 Number of animals 9 8 8 9 9 5 Mean (g) 2.28 2.17 2.19 1.99 2.04 1.81 Standard error 0.04 0.04 0.03 0.05 0.14 0.12 Source: Gulati et al. (1991). B-52 DRAFT - DO NOT CITE OR QUOTE ------- Table B-16. Summary of benchmark dose modeling results for fetal body weight following exposure of pregnant Sprague-Dawley rats on gestational days 4—20 Model Test for significant difference />-valuea Variance />-valueb Means />-valueb Scaled residuals of interest0 AIC BMD1SD (mg/kg- day) BMDL1SD (mg/kg- day) All dose groups included Constant variance Lineard O.0001 O.0001 0.40 -0.92/1.23 -91.54 201.09 139.17 Non constant variance Lineard O.0001 0.07 0.20 -1.25/0.88 -112.47 84.64 56.25 Highest dose group dropped Constant variance Lineard O.0001 O.0001 0.40 -1.24/0.70 -83.65 238.24 147.87 Non constant variance Linear O.0001 0.05 0.18 -1.27/0.83 -105.40 84.31 53.36 Two highest dose groups dropped Constant variance Hille T • d,f Linear Polynomial (2-degree)d Polynomial (3 -degree) Powere 0.0002 0.0002 0.0002 0.0002 0.0002 0.35 0.35 0.35 0.35 0.35 NA 0.12 0.06 0.08 0.06 0.38/-0.06 -1.19/1.46 0.87/-0.20 0.65/-0.09 0.38/-0.06 -101.33 -104.84 -103.53 -103.98 -103.33 129.74 83.10 110.21 118.06 129.71 61.35 59.73 62.16 64.06 61.40 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; SD = standard deviation aValues >0.05 fail to meet conventional goodness-of-fit criteria. bValues <0.10 fail to meet conventional goodness-of-fit criteria. °Scaled residuals at doses immediately below and immediately above the benchmark dose. Coefficients restricted to be negative. "Power restricted to >1. Best-fitting model is displayed in boldface type. The linear model is the only model providing an adequate fit to the data. 2 B-53 DRAFT - DO NOT CITE OR QUOTE ------- Linear Model with 0.95 Confidence Level c o Q_ (/) 0 ce c ro 0 2.4 2.3 2.2 2.1 1.9 Linear BMDL BMD 20 40 60 80 100 120 140 160 dose 180 1 2 ^ ^J 4 5 6 1 8 9 10 _L *_* 11 12 13 _L —* 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 15:0203/292010 Polynomial Model. (Version: 2.13; Date: 04/08/2008) Input Data File: C:\USEPA\lRlS\TCE\gulati\fetalbdwt\lin f etalbdwt2HDD linear. Gnuplot Plotting File: C:\USEPA\lRlS\TCE\gulati\fetalbdwt\lin f etalbdwt2HDD linear. Mon Mar 29 16 : 02 :5 HMDS Model Run The form of the response function is: Y[dose] = beta 0 + beta l*dose + beta 2*doseA2 + ... Dependent variable = mean Independent variable = dose rho is set to 0 The polynomial coefficients are restricted to be negative A constant variance model is fit Total number of dose groups = 4 Total number of records with missing values = 0 Maximum number of iterations = 250 Relative Function Convergence has been set to: le-008 (d) pit 7 2010 B-54 DRAFT - DO NOT CITE OR QUOTE ------- 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 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 Parameter Convergence has been set to: le-008 Default Initial Parameter Values alpha = 0.0141567 rho = 0 Specified beta_0 = 2.26747 beta 1 = -0.0014099 Asymptotic Correlation Matrix of Parameter Estimates ( *** The model parameter(s) -rho have been estimated at a boundary point, or have been specified by the user, and do not appear in the correlation matrix ) alpha beta_0 beta 1 alpha 1 -l.Se-OlO 2e-010 beta_0 -l.Se-OlO 1 -0. 75 beta_l 2e-010 -0 .75 1 Variable alpha beta_0 beta 1 Parameter Estimates 95.0% Wald Confidence Interval Estimate Std. Err. Lower Conf. Limit Upper Conf. Limit 0.0141234 0.00342543 0.00740968 0.0208371 2.26874 0.0306445 2.20868 2.3288 -0.00143017 0.000290756 -0.00200004 -0.000860296 2.28 2.17 2.19 1. 99 2.27 2.22 2.13 2. 01 0.12 0.11 0. 08 0.15 0 .119 0 .119 0 .119 0 .119 0.284 -1 .19 1 .46 -0.538 Table of Data and Estimated Values of Interest Dose N Obs Mean Est Mean Obs Std Dev Est Std Dev Scaled Res. 0 34 98 180 Model Descriptions for likelihoods calculated Model Al: Yij = Mu(i) + e(ij) Var{e(ij) } = SigmaA2 Model A2: Yij = Mu(i) + e(ij) Var{e(ij) } = Sigma(i)A2 Model A3: Yij = Mu(i) + e(ij) Var{e(ij)} = SigmaA2 Model A3 uses any fixed variance parameters that were specified by the user Model R: Yi = Mu B-55 DRAFT - DO NOT CITE OR QUOTE ------- 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 Test 1 Test 2 Test 3 Test 4 (Note: Test Var{e(i)} = SigmaA2 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 Likelihoods of Interest Log (likelihood) 57 .506457 59 .148779 57 .506457 55 .418685 46.282389 # Param' s 5 8 5 3 2 AIC -105. 012914 -102 .297557 -105. 012914 -104.837369 -88. 564779 Model Al A2 A3 fitted R Explanation of Tests Do responses and/or variances differ among Dose levels? (A2 vs. R) Are Variances Homogeneous? (Al vs A2) Are variances adequately modeled? (A2 vs. A3) Does the Model for the Mean Fit? (A3 vs. fitted) When rho=0 the results of Test 3 and Test 2 will be the same.) Tests of Interest -2*log (Likelihood Ratio) Test df p-value 25 .7328 3.28464 3.28464 4. 17554 6 3 3 2 0.0002497 0.3498 0.3498 0.124 Test 1 Test 2 Test 3 Test 4 The p-value for Test 1 is less than .05. There appears to be a difference between response and/or variances among the dose levels It seems appropriate to model the data The p-value for Test 2 is greater than .1. model appears to be appropriate here The p-value for Test 3 is greater than .1. to be appropriate here The p-value for Test 4 is greater than .1. to adequately describe the data A homogeneous variance The modeled variance appears The model chosen seems B-56 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 Benchmark Dose Computation 3 4 Specified effect = 1 5 6 Risk Type = Estimated standard deviations from the control mean 7 8 Confidence level = 0.95 9 10 HMD = 83.0965 11 12 13 BMDL = 59.7345 14 15 B-57 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 APPENDIX C. BENCHMARK DOSE MODELING RESULTS FOR THE DERIVATION OF THE ORAL SLOPE FACTOR Hepatocellular carcinomas in male and female B6C3F] mice (Tables C-l and C-2) The incidence data for hepatocellular carcinomas in male and female B6C3Fi mice exposed via gavage to 1,1,2,2-tetrachloroethane 5 days/week for 78 weeks are shown in Table C- 1 (NCI, 1978). Table C-l. Incidence of hepatocellular carcinomas in B6C3Fi mice administered 1,1^2,2-tetrachloroethane by gavage for 78 weeks Endpoint Hepatocellular carcinomas Sex M F Dose (mg/kg-day)a 0" 3/36 1/40 8.22 13/50 30/48 16.5 44/49 43/47 10 11 12 13 14 15 16 17 18 aHED as calculated in Section 5.4.3 and shown in Table 5-5. bPooled vehicle controls Source: NCI (1978). The BMD modeling results from the data in Table C-l are summarized in Tables C-2 (for males) and C-3 (for females) followed by the standard BMDS output for the selected models from version 2.1.1 of the software. The multistage cancer model did not provide an adequate fit to the incidence data for hepatocellular carcinomas in male mice; these data are considered unsuitable for BMD modeling. The one-stage multistage model provided the best fit to the incidence data for hepatocellular carcinomas in females, and this model was used as the basis for the BMDio and BMDLio estimates (0.81 and 0.65 mg/kg-day, respectively, as HEDs) for this endpoint. C-l DRAFT - DO NOT CITE OR QUOTE ------- Table C-2. Summary of benchmark dose modeling results for the incidence of hepatocellular carcinomas in male mice Model Multistage (1 -degree polynomial)0 Multistage (2 -degree polynomial)0 DF 1 1 x2 18.30 5.24 % Goodness of fit />-valuea O.001 0.02 Scaled residuals of interest1" 0.51/-3.27 0.53/-1.83 AIC 134.58 119.87 BMD10[HED] (mg/kg-day) 1.42 4.10 BMDL10[HED] (mg/kg-day) 1.11 3.08 2 3 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; DF = degrees of freedom "Values < 0.1 fail to meet conventional goodness-of-fit criteria. Scaled residuals at doses immediately below and immediately above the benchmark dose. °Betas restricted to > 0. Table C-3. Summary of benchmark dose modeling results for the incidence of hepatocellular carcinomas in female mice Model Multistage (1-degree polynomial)*'* Multistage (2 -degree polynomial)0 DF 1 0 x2 0.74 0.00 X2 Goodness of fit />-valuea 0.39 NA Scaled residual of interest 0.04/-0.61 0.00/0.00 AIC 104.99 106.22 BMD10[HED] (mg/kg-day) 0.81 1.18 BMDL10[HED] (mg/kg-day) 0.65 0.67 AIC = Akaike Information Criterion; BMD = maximum likelihood estimate of the dose associated with the selected benchmark response; BMDL = 95% lower confidence limit on the BMD; DF = degrees of freedom; NA= not applicable (/>value was not generated due to insufficient DF) "Values < 0.1 fail to meet conventional goodness-of-fit criteria. bScaled residuals at doses immediately below and immediately above the benchmark dose. °Betas restricted to > 0. dSelected model is displayed in boldface type. C-2 DRAFT - DO NOT CITE OR QUOTE ------- Multistage Cancer Model with 0.95 Confidence Level 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 C o • 0.8 0.6 0.4 0.2 Multistage Cancer Linear extrapolation BMDL BMD 8 dose 10 12 14 16 15:11 03/292010 Multistage Cancer Model. (Version: 1.7; Date: 05/16/2008) Input Data File: C:\USEPA\lRlS\TCE\NCl\hepcarc\female\msc_hepcarcF_MS_l.(d) Gnuplot Plotting File: C:\USEPA\lRlS\TCE\NCl\hepcarc\female\msc_hepcarcF_MS_l.pit Mon Mar 29 16:11:43 2010 HMDS Model Run The form of the probability function is: P [response] = background + (1-background)*[1-EXP( -betal*doseAl)] The parameter betas are restricted to be positive Dependent variable = incidence Independent variable = dose Total number of observations = 3 Total number of records with missing values Total number of parameters in model = 2 Total number of specified parameters = 0 Degree of polynomial = 1 = 0 C-3 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 3 4 5 6 7 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 43 44 45 46 47 48 T-O 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 Maximum number of iterations = 250 Relative Function Convergence has been set to: le-008 Parameter Convergence has been set to: le-008 Default Initial Parameter Values Background = 0 Beta(l) = 0.147828 Asymptotic Correlation Matrix of Parameter Estimates Background Beta ( 1 ) Background 1 -0.54 Beta(l) -0.54 1 Parameter Estimates 95.0% Wald Confidence Variable Estimate Std. Err. Lower Conf. Limit Upper Background 0.0240983 * * Beta(l) 0.130589 * * * - Indicates that this value is not calculated. Analysis of Deviance Table Model Log (likelihood) # Param's Deviance Test d.f. P- value Full model -50.1115 3 Fitted model -50.4931 2 0.763231 1 0.3823 Reduced model -92.948 1 85.673 2 <.0001 AIC: 104.986 Goodness of Fit Dose Est. Prob. Expected Observed Size 0.0000 0.0241 0.964 1.000 40 8.2200 0.6664 31.988 30.000 48 16.5000 0.8869 41.682 43.000 47 ChiA2 = 0.74 d.f. = 1 P-value = 0.3897 Benchmark Dose Computation Specified effect = 0.1 Risk Type = Extra risk Confidence level = 0.95 Interval Conf. Limit * * Scaled Residual 0.037 -0.608 0.607 C-4 DRAFT - DO NOT CITE OR QUOTE ------- 1 2 HMD = 0.806812 3 4 BMDL = 0.648049 5 6 BMDU = 1.01577 7 8 Taken together, (0.648049, 1.01577) is a 90 % two-sided confidence 9 interval for the HMD 10 11 Multistage Cancer Slope Factor = 0.154309 12 13 14 15 C-5 DRAFT - DO NOT CITE OR QUOTE ------- |