Pilot Study On International Information Exchange On Dioxins and Related Compounds Scientific Basis for the Development of the International Toxicity Equivalency Factor (I-TEF) Method of Risk Assessment for Complex Mixtures of Dioxins and Related Compounds Report Number 178 December 1988 North Atlantic Treaty Organization Committee on the Challenges of Modern Society EPA 600 6 90 015 ------- ------- fell-1X qc-IO- 0003 ACKNOWLEDGMENTS This report of the NATO/CCMS Pilot Study on International Information Exchange on Dioxins and Related Compounds was prepared by the U.S. Envi- ronmental Protection Agency (EPA), Office of Research and Development, and Versar Inc., under Contract No. 68-02-4254. Mr. Erich W. Bretthauer of the U.S. EPA was the Study Director for the Exposure and Hazard Assessment Working Group, and Dr. Donald G. Barnes of the U.S. EPA was the Chairman of the TEF Subgroup. Dr. Stephen H. Safe of Texas A&M University was the principal author of this report. Contributing authors also included Dr. Frederick W. Kutz of the U.S. EPA and Mr. David P. Bottimore of Versar Inc. Peer reviewers of this report are listed below as are the lead dele- gates and TEF Subgroup members that reviewed and concurred with the publi- cation of this report. Dr. Linda S. Birnbaum Dr. Brendan Birmingham Ms. Sigrid Louise Bjornstad Dr. Martin J. Boddington Mr. E.A. Cox Dr. Alessandro di Domenico Dr. Donald L. Grant Prof. Dr. med. Helmut Greim Dr. Arne Grove Dr. G.K. Matthew Ms. Christa Morawa Dr. James R. Olson Ms. Frances Pollitt Dr. Ellen Silbergeld Dr. C.A. van der Hiejden Dr. Job A van Zorge Dr. James Wilson National Institute of Environmental Health Sciences (United States) Ontario Ministry of the Environment (Canada) State Pollution Control Authority (Norway) Environment Canada (Canada) Inspectorate of Pollution, Department of the Environment (United Kingdom) Istituto Superiore di Sanita (Italy) Health and Welfare Canada (Canada) GSF Muenchen Institute fur Toxikologie (Federal Republic of Germany) Kemiteknik, Teknologisk Institut (Denmark) Department of Health and Social Security (United Kingdom) Umweltbundesamt (Federal Republic of Germany) State University of New York - Buffalo (United States) Department of Health and Social Security (United Kingdom) Environmental Defense Fund (United States) National Institute of Public Health and Environmental Hygiene (The Netherlands) Ministry of Housing, Physical Planning and Environment (The Netherlands) Monsanto Chemical Company (United States) HEADQUARTERS U««Y ENVIRONMENTAL PR0TBCT10H WASHINGTON, D.C. 20460 ------- ------- SCIENTIFIC BASIS FOR THE DEVELOPMENT OF THE INTERNATIONAL TOXICITY EQUIVALENCY FACTOR (I-TEF) METHOD OF RISK ASSESSMENT FOR COMPLEX MIXTURES OF DIOXINS AND RELATED COMPOUNDS Table of Contents Page No. List of Tables . ii List of Figures i ii 1. INTRODUCTION 1 2. PCDDs AND PCDFs - ENVIRONMENTAL IMPACT 3 3. PCDDs AND PCDFs - PROBLEMS IN RISK ASSESSMENT 6 4. 2,3,7,8-TCDD AND RELATED COMPOUNDS - MECHANISM OF ACTION .... 9 5. DEVELOPMENT OF TEFs FOR PCDDs AND PCDFs 17 5.1 Lethal i ty 18 5.2 In Vivo Biologic and Toxic Responses 18 5.3 In Vitro Potencies 24 5.4 Toxicity Equivalency Factors 32 6. BIOASSAYS FOR HAZARD ASSESSMENT OF PCDD AND PCDF MIXTURES ... 41 REFERENCES 46 ------- LIST OF TABLES Multiplicity of PCDD and PCDF Isomers and Congeners Page No. 7 Table 1. Table 2. Toxicity of 2,3,7,8-TCDD and Related Compounds: Species Differences 10 Table 3. Cooperative LDc0 Values for Several PCDD and PCDF Congeners in the Guinea Pig, Mouse, and Rat 19 Table 4. A Summary of the Dose-Response In Vivo Biologic and Toxic Effects of Several Halogenated Aromatics in the Immature Male Rat and Guinea Pig 21 Table 5. £055 Values for PCDF Congeners in the Mouse: Immunotoxicity and Teratogenicity 23 Table 6. PCDDs and PCDFs as Competitive Ligands for the Rat Hepatic Cytosol Receptor SARs 26 Table 7. PCDDs and PCDFs as Inducers of AHH and EROD Activities in Rat Hepatoma H-4-II E Cells in Culture: SARs 28 Table 8. International Toxicity Equivalency Factors (I-TEFs): Comparison of Relative Potency Data for the 2,3,7,8- Substituted PCDDs and PCDFs 35 Table 9. Brominated Aromatic Flame Retardant Pyrolysis and Municipal Fly Ash Extracts: In Vitro and In Vivo Determination of 2,3,7,8-Equivalents 42 Table 10. Calculated 2,3,7,8-TCDD Equivalent Concentrations of the Binghamton Soot for Various Dose-Related Endpoints Following Subchronic Exposure 45 11 ------- LIST OF FIGURES Page No. Figure 1. Figure 2. Figure 3. Figure 4, Figure 5. Figure 6. Figure 7, Chemical Sturucture of PCDDs and PCDFs Proposed Mechanism of Action of 2,3,7,8-TCDD and Related Compounds Relative Potencies of Three Isomeric TetraCDDs (Rat) . Correlation Between the -log ECgQ Values (m) for AHH Induction Activity in Ret Hepatoma H-4-II E Cell vs. the -log ED5Q Values (mol/kg) for Hepatic Microsomal AHH Induction Activity (top panel), Inhibition of Body Weight Gain (middle panel), and Thymic Atrophy (bottom pane') in Male Wistar Rats for Several PCDD and PCDF Congeners Correlation Between the -log EC5Q Values (m) for AHH Induction Activity in Ret Hepatoma H-4-II E Cell vs. the -log £050 Values (mol/kg) for Hepatic Microsomal AHH Induction Activity (top panel), Inhibition of Body Weight Gain (middle panel), and Thymic Atrophy (bottom panel) in Male Wistar Rats for Several PCDD, PCDF, PCB, and PBDD Congeners Correlation Between -Log EC^n (m)AHH Induction (in Rat Hepatoma H-4-II-E cells) vs. -Log £050 (mol/kg) Inhibition of Body Weight Gain (Guinea Pig) .. Correlation Between -Log ECcQ (m)AHH Induction (In Rat Hepatoma H-4-II-E Cells) vs. -Log ED50 (mol/kg) Immunotoxicity (C57BL/6 Mice) 12 14 30 Figure 8. 2378-Substituted PeCDFs 31 33 34 40 in ------- ------- 1. INTRODUCTION The Pilot Study on International Information Exchange on Dioxins and Related Compounds was initiated to apply the cooperative efforts of sever- al nations to address issues associated with polychlorinated dibenzo- p-dioxins (PCDDs), dibenzofurans (PCDFs), and related compounds. The three-year study was conducted under the aegis of the Committee on the Challenges of Modern Society (CCMS) of the North Atlantic Treaty Organiza- tion (NATO). Participating nations included Canada, Denmark, the Federal Republic of Germany, Italy, the Netherlands, Norway, the United Kingdom, and the United States. In addition, the governments of Sweden and Austria, although not members of NATO, requested to be informed of the progress of the study. International organizations that participated as observers included the World Health Organization, the United Nations Environmental Programme, the Organization for Economic Cooperation and Development, and the Commission of European Communities. Numerous information exchange activities were undertaken to promote cooperative research and to identify duplicative efforts and knowledge voids so that better informed decisions can be made concerning future activities. When the project was initiated in 1985, it was divided into three areas of study: exposure and hazard assessment, technology assess- ment, and management of accidents. The Exposure and Hazard Assessment Working Group, chaired by the United States, was charged with several tasks concerning research and risk assessment. The working group devel- oped the International Toxicity Equivalency Factor (I-TEF) method to assess the risks posed by exposures to complex mixtures of PCDDs and PCDFs. The objective was to develop an updated TEF scheme based on exist- ing methods and the most current data available (Bellin and Barnes, 1987). The I-TEFs were reported in NATO/CCMS Report Number 176 and were presented in the Special Session on Prospective Dioxin Research and Regulatory Issues sponsored by the United States Environmental Protection Agency and the CCMS at the Eighth International Symposium on Chlorinated Dioxins and Related Compounds (Dioxin '88) in Umea, Sweden. ------- The I-TEFs were presented as an interim procedure that should be of practical assistance in addressing a number of questions associated with PCDOs and PCDFs. The thrust of Report Number 176 was to endorse the toxicity equivalency factor (TEF) concept as an interim measure of pru- dent science policy and to recommend specific I-TEF values to facilitate consistency among participating nations in communicating analytical re- sults and performing risk assessments. That report also included examples of how the procedure is applied to analytical data and how the various TEF schemes differ. This report presents a detailed discussion of the scientific basis for the development of toxicity equivalency factors and provides the toxicity data and methodology used to determine the I-TEFs. Section 2 of this report discusses the sources and environmental im- pact of PCDDs and PCDFs and provides a background for the utility of risk assessment techniques for these chemicals with regard to their presence in humans and the environment. Section 3 presents some of the problems encountered in performing risk assessments for complex mixtures of PCDDs and PCDFs and the need for the development of toxicity equivalency fac- tors. Section 4 presents the mechanisms of toxicity for these compounds and the structure-activity relationships of the various isomers/congeners of PCDDs and PCDFs. Section 5 provides a discussion of the development of TEFs from experimental toxicity data. In addition, this section pre- sents the data and methodology used to develop the I-TEFs by the NATO/CCMS Pilot Study on International Information Exchange on Dioxins and Related Compounds. Section 6 presents bioassay techniques that can be used to estimate and/or verify the toxic equivalents calculated from analytical data for a variety of mixtures including PCDDs and PCDFs as well as poly- brominated dibenzo-p-dioxins (PBDDs) and polybrominated dibenzofurans (PBDFs). ------- 2. PCDDs AND PCDFs - ENVIRONMENTAL IMPACT Polychlorinated dibenzo-p-dioxins (PCDDs) and dibenzofurans (PCDFs) (Figure 1) are members of a class of organic pollutants that include the polychlorinated biphenyls (PCBs), naphthalenes, azobenzenes, and ter- phenyls; the polybrominated biphenyls (PBBs); and other mixed polychloro/ bromo aromatics. PCBs were widely used as industrial compounds, whereas the PCDDs and PCDFs are not industrial or commercial chemicals but are formed as by-products of diverse processes. For example, PCDDs and PCDFs are formed during the synthesis of chlorinated phenols and their derived products (Rappe et al. 1978, 1979; Nilsson et al. 1978). PCDDs and PCDFs are also formed during the incomplete combustion or incineration of organohalogen-cortaining wastes (Buser et al. 1978; Rappe et al. 1979, 1983; Buser, 1979; Lustenhouwer et al. 1980; Ballschmiter et al. 1983; 01ie et al. 1977) and in automobile combustion processes (Marklund et al. 1987). Polybrominated dibenzo-p-dioxins (PBDDs), dibenzofurans (PBDFs), and halogenated (mixed bromo/chloro) dibenzo-p-dioxins and dibenzo- furans have recertly been identified as by-products of waste incineration in which there are sources of chlorine and bromine (Thoma et al. 1986, Schafer and Ballschmiter 1986). In addition, PCDFs have also been reported as contaminants in commercial PCBs (Vos et al. 1970, Bowes et al. 1975, Kunita et al. 1984). PCDDs, PCDFs, PCBs, and related halogenated aryl hydrocarbons are highly stable, lipophilic chemicals and this is paralleled by their envi- ronmental persistence and preferential bioaccumulation in higher trophic levels of the food chain. Recent studies have identified PCDDs and PCDFs in river and lake sediments (Czuczwa et al. 1984a, 1984b) and at subparts- per-billion levels in fish, wildlife, human adipose tissue, and milk (Van den Berg et al. 1985; Rappe et al. 1981, 1987; Ryan et al. 1985; Ryan 1986, Ono et al. 1986; Stalling et al. 1985; Kahn et al. 1988). High resolution gas chromatographic-mass spectrometric (GC-MS) analysis has demonstrated that the PCDDs and PCDFs present in environmental matrices ------- Cl, Polychlorinated dibenzo-p-dioxins (PCDDs) Cl, Polychlorinated dibenzofurans (PCDFs) FIGURE 1. Chemical Structure of PCDDs and PCDFs ------- are highly complex mixtures of isomers and congeners, and this correlates with the composition of PCDDs and PCDFs identified in industrial or combustion by-products. Several studies have reported the high resolution GC-MS analysis of PCDDs and PCDFs in human samples (Ryan et al. 1985, Van den Berg et al. 1985) and it was evident that humans are also primarily exposed to complex mixtures of these compounds. In contrast, human exposure to 2,4,5-tri- chlorophenol and its derived products (e.g., 2,4,5-T, Agent Orange) can result primarily in exposure to a single PCDD congener, namely the highly toxic 2,3,7,8-tetrachlorodibenzo-p-dioxin (2,3,7,8-TCDD). In situa- tions where there is excess exposure to 2,3,7,8-TCDD, the mixture of PCDDs and PCDFs will contain higher levels of this compound. For example, "Vietnam veterans who were heavily exposed to Agent Orange exceeded matched control subjects in both blood and adipose tissue levels of 2,3,7,8-TCDD" (Kahn et al. 1988). Nevertheless, even in this study, 2,3,7,8-TCDD was only one of 13 PCDD and PCDF congeners identified in these human samples. ------- 3. PCODs AND PCDFs - PROBLEMS IN RISK ASSESSMENT It is apparent that risk assessment of PCDD or PCDF mixtures require prior information on the identities of the specific congeners and their concentration in any analyte. An analytical approach to this problem requires the availability of all the appropriate PCDF and PCDD congeners (see Table 1} and methods that can both separate and quantitate each indi- vidual compound in a mixture. Several laboratories have made significant progress in this field, using sophisticated cleanup procedures followed by high resolution capillary GC and/or GC-MS analysis, which can separate, identify, and quantitate individual PCDD and PCDF congeners {Buser and Rappe 1980, Crummett 1983). In addition to the general lack of availabil- ity of all the PCDD and PCDF standards, these analytical techniques require highly sophisticated equipment and training and are therefore very costly. Moreover, the use of analytical data for risk assessment requires prior information on the relative potencies of the individual components in the mixture, and it must also be assumed that the interactive effects of the components in the mixture are not significant. This assumption may not always be correct since several studies have reported antagonistic interactions between 2,3,7,8-TCDD and the commercial PCB, Aroclor 1254, and several 1,3,6,8-substituted dibenzofurans (Keyes et al. 1986, Haake et al. 1987, Bannister et al. 1987, Davis and Safe, 1988). Antagonistic interactions were observed only when the relative concentrations of the antagonists were high. Regulatory risk assessments for individual compounds routinely use the results of long-term cancer bioassays for generating various "effect levels" that can be used in standard setting. Rodent carcinogenicity studies for 2,3,7,8-TCDD (Kociba et al. 1978) and a hexachlorodibenzo- p-dioxin mixture (NTP 1980) have been reported, and the data obtained for 2,3,7,8-TCDD have been used extensively by numerous regulatory agencies (USEPA 1985, Kimbrough et al. 1984, Van der Heijden et al. 1982, Bell in and Barnes 1987, Ontario, 1982, OME, 1985) to calculate virtually safe doses (VSDs) or allowable daily intakes (ADIs). Carcinogenicity ------- 8366H Table 1. Multiplicity of PCOO and PCOF Isomers and Congeners PCDDs PCDFs Toxic PCOOsa Toxic PCDFs8 Total 2.3.7.8-CDDs/CDFs Total non -2.3.7.8-CDDs/CDFs TOTAL Number of i softie rs C1L C12 C13 C14 C15 C16 Clj C18 Total 2 10 14 22 14 10 2 1 75 4 16 28 38 28 16 4 1 135 11311 7 1 2 4 2 1 10 17 193 210 2,3,7,8 substituted congeners (Mason et al. 1985. Safe 1986). ------- studies on the other environmentally relevant individual PCODs and PCDFs or mixtures of these compounds have not been determined. Thus, it is not possible to calculate carcinogenicity-derived VSD or ADI values for PCDD or PCDF isomers and congeners (other than 2,3,7,8-TCDD), and this has necessitated the development of toxicity equivalency factors (TEFs) for this class of compounds (Bellin and Barnes, 1987; Safe 1987). This approach utilizes the available in vivo and in vitro data on the toxic potencies of individual PCDDs and PCDFs and assigns a fractional number comparing their toxic potency to that of the most toxic member of this class of compounds, namely 2,3,7,8-TCDD (fractional number = 1.0). The rationale for the use of TEFs is based on extensive research on the mech- anism of action of 2,3,7,8-TCDD and related halogenated aryl hydrocarbons, The following section of this report briefly highlights some of the results that provide strong support for the use of TEFs in the risk assessment process for PCDDs and PCDFs. ------- 4. 2,3,7,8-TCDO AND RELATED COMPOUNDS - MECHANISM OF ACTION PCDDs, PCDFs.. and related aryl hydrocarbons elicit a broad spectrum of biologic and toxic responses in diverse animal species and mammalian cells in culture (reviewed in Poland et al. 1979; Poland and Knutson 1982; Safe 1986; Whitlock 1986, 1987). Some of these effects include a wasting syn- drome, chloracne and related dermal lesions, hepatotoxicity and porphyria, reproductive toxicity, immunologic alterations, and tumor promotion. In addition, treatment of laboratory animals and mammalian cells with halo- genated aromatics causes a number of responses including (a) the induction of diverse Phase I and Phase II drug-metabolizing enzymes, ornithine decarboxylase, and enzymes associated with steroid metabolism; (b) modula- tion of diverse cellular hormone receptor levels; and (c) the depletion of hepatic vitamin A levels. 2,3,7,8-TCDD, the most potent halogenated aryl hydrocarbon, has served as a prototype for investigating the effects and mechanism of action of this important class of chemicals. Utilizing 2,3,7,8-TCDD as a probe, it has been shown that the effects and relative potency of this compound and related halogenated aromatics are dependent on several factors, including (a) the age and sex of the animal, (b) the species and strain used, and (c) the structure of the chemical used. Although the wasting syndrome and lymphoid involution are routinely observed in most laboratory animals exposed to 2,3,7,8-TCDD and related compounds, many other responses are highly species-specific (Table 2). For example, chloracne and related dermal lesions are observed in humans, monkeys, some genetically inbred strains of mice e.g., hairless (HRS/J) mice, cattle and rabbits, but not in other animal species. Some of the early research on 2,3,7,8-TCDD illustrated the differen- tial species sensitivity to the lethality of 2,3,7,8-TCDD as evidenced by the following LD^g values: guinea pig (0.6-2.0 ^g/kg), rat (22-45 M9/kg), chicken (25-50 ^g/kg), monkey (70 /^g/ kg), rabbit (115 /^g/kg), dog (100-200 M9/kg), mouse (114-284 //g/kg), bull frog (> 1000 M9/kg), and hamster (1157-5000 /ig/kg) (Kociba and Schwetz 1982). More recent studies ------- 8366H Table 2. Toxicity of 2.3.7,8-TCDD and Related Compounds: Species Differences Animal species Effect Chloracne and related dermal lesions Body weight loss Developmental toxic ity Liver damage Edema Thymic atrophy Guinea Human Monkey Pig J J 0 J 4 J J J 0 J 0 J J Mouse Chicken Jb o 4 J J J J 4 J J J 4 Rat 0 4 4 4 0 4 4 Effect observed in animal species. 0 Effect not observed. a Poland and Knutson (1982). Observed only in hairless HRS/J mice. 10 ------- have also demonstrated that the mink are also susceptible to 2,3,7,8-TCDD induced lethality (Hochstein et al, 1988) (LD5Q 4.2 /*g/kg) and that there are remarkable intraspecies strain differences in the toxicity of this compound (Chapman and Schiller 1985; Walden and Schiller 1985; Pohjanvirta et a". 1988). These results illustrate the greater than 5000- fold difference ~n the sensitivity of the guinea pig and hamster to the lethal effects of 2,3,7,8-TCDD; however, it is not clear whether this species sensitivity is observed for other effects caused by 2,3,7,8-TCDD and related compounds. The mechanisn of action of 2,3,7,8-TCDD and related compounds has been extensively investigated using a number of different in vivo and in vitro models. Unlike nany other toxic and genotoxic hydrocarbons, 2,3,7,8-TCDD does not act via metabolic activation processes or by covalent modifica- tion of cellular protein, RNA, and DNA {reviewed in Shu et al. 1987). However, a multitude of studies strongly support a receptor-mediated mech- anism of action for 2,3,7,8-TCDD and related halogenated aryl hydrocarbons (Figure 2). The following lines of evidence support this mechanism. Studies with genetically inbred "responsive" and "nonresponsive" strains of mice (e.g., C57BL/6 and DBA/2 mice, respectively) and their backcrosses showed that many of the biochemical (e.g., induction of aryl hydrocarbon hydroxylase (AHH) (Poland and Glover 1973, 1974, 1975; Poland et al. 1974) and toxic (porphyria, immunotoxicity, thymic atrophy, body weight loss, epidermal hyperplasia and hyperkeratosis, and lethality) responses caused by 2,3,7,8-TCDD segregate with a specific genetic locus, namely, the aryl hydrocarbon (Ah) locus (Poland and Glover 1980; Vecchi et al. 1980, 1983; Nagarkatti et al. 1984; Jones and Sweeney 1980; Luster et al. 1986). The initial hypothesis for this locus, which codes for the Ah receptor protein, was proposed to explain the differences in AHH inducibility observed in genetically inbred strains of mice by aryl hydrocarbons and 2,3,7,8-TCDD. Poland and coworkers (1976) first identified a saturable, high-affini- ty, low-capacity Ah receptor protein in murine hepatic cytosol. Subse- quent studies in several laboratories have extensively characterized the 11 ------- Nuclear Binding Sites Cytoplasm Cytochrome P-4501A1 mRNA Cytochrome P-4501A1 Induction (AHH ind other monooxygenases) . "Induced Proteins" . Pleiotroplc Response* ' -2,3,7,8-TCDD and related isostereomert -PAH FIGURE 2. Proposed Mechanism of Action of 2,3,7,8-TCDD amd Related Compounds. 12 ------- molecular properties of this intracellular protein in several animal species including humans (reviewed in Safe 1988). Furthermore, the direct binding of the Ah receptor with other aryl hydrocarbons (Okey et al. 1984), including radiolabeled PCDF congeners has also been reported (Parrel 1 et al. 1987). Endogenous receptor ligands such as steroid hormones and neurotrans- mitters interact with receptors that are located within specific tissues or cells. Tissue specificity has also been demonstrated for the 2,3,7,8- TCDD receptor ir rats and mice (Okey et al. 1983, 1984); Carlstedt-Duke 1979; Denison et al. 1986; Gasiewicz et al. 1984). C57BL/6J mice and Sprague-Dawley rats, which are highly responsive to 2,3,7,8-TCOD, exhibit tissue-dependent concentrations of the receptor that vary from 0 to 54 fmol/mg cytosolic protein. Relatively high levels of receptor have been identified in tissues (e.g., liver, thymus) that are responsive to the effects of 2,3,7,8-TCDD and related compounds. In contrast, nondetectable levels of the receptor protein are observed in cytosol from DBA/2J mice, which are relatively nonresponsive to the effects of 2,3,7,8-TCDD and related toxic halogenated aryl hydrocarbons. However, the correlation between tissue susceptibility and the receptor levels requires further study. Stereoselective interactions between receptors and their respective hormone agonists are one of the important characteristics of the receptor- mediated processes. The effects of structure on the biologic and toxic potencies of PCDDs and PCDFs have been extensively investigated (reviewed in Poland et al. 1975, 1979; Poland and Knutson 1982; Safe 1986). Figures 3 and 4 depict the effects of variable lateral 2,3,7, and 8 chlo- rine substituents on the toxicity of a series of PCDD and PCDF congeners (Safe 1986, 1987; Mason et al. 1985, 1986). Figure 3 summarizes the relative potencies of three tetrachlorodibenzo-p-dioxin (tetraCDD) isomers, namely 2,3,7,8-, 1,3,7,8-, and 2,3,6,8-tetraCDD, on body weight loss and thymic atrophy in the rat and their relative activities as induc- ers of AHH in rat hepatoma H-4-II E cells and rat hepatic microsomes. The 13 ------- 2,3,7,8-TCDD- 1,3,7,8-TCDD_~ 2,3,6,8-TCDD <5.0 <4.0 I I in vitro in vivo (rat) 8 -log ED 50 10 12 2,3,7,8-TCDD _ 1,3,7,8-TCDD- 2,3,6,8-TCDD_ <3.0 <3.0 body wt. loss thymic atrophy I 5 I 7 -log ED50 FIGURE 3. Relative Potencies of 3 Isomeric TetraCDDs (Rat) [Upper Panel; Induction of AHH in Rat Hepatoma H-4-NE Cells (In Vitro) and Rat Hepatic Microsomes fin Vivo): Lower Panel; Toxicities Observed in Male Rats (Safe 1986, 1987)]. The In Vivo ED50 Values Are mol/kg and the jn Vitro Values for Induction Are Concentrations (M). 14 ------- major structural diversity in these tetraCDD isomers is the number of lateral chlorine substituents (4, 3, and 2, respectively). The EDgQ values for 2,3,7,8-TCDO are approximately 100-fold lower than the values for 1,3,7,8-tetraCDD, and the only structural difference in the two isomers involves the shift of the C-2 lateral Cl group to a nonlateral (C-l) position. The third isomer, 1,3,6,8-tetraCDD, is not toxic at doses up to 10 mol/kg. Comparable SARs have been observed for PCDF congeners (Bandiera et al. 1984; Mason et al. 1985). Structure-activity relationships (SARs) for PCDDs and PCDFs have been observed for a number of Ah receptor-Tiediated responses (e.g., teratogenicity, immunotoxicity, lethality, enzyne induction, body weight loss, thymic atrophy, Ah receptor binding, and various dermal lesions) in several animal species (guinea pig, rat, and mouse) and mammalian cells in culture (see Section 5). These data strongly support the role of the Ah receptor in mediating the biologic and toxic responses elicited by 2,3,7,8-TCDD and related PCDDs and PCDFs and provide the scientific basis for the development of TEFs for this class of compounds. It should also be noted that the devel- opment of toxic responses after exposure to PCDDs and PCDFs is a complex multicomponent process. For example, cellular models have been developed for investigating 2,3,7,8-TCDD-mediated epidermal hyperplasia, hyperkera- tosis, sebaceous gland metaplasia, and tumor promotion in HRS/J hairless (hr/hr) and haired (hr/+) mice (Knutson and Poland 1980, 1982; Poland et al. 1982, 1933, 1984). Their results indicate that a second genetic locus (the hr lacus) is involved in the development of these epidermal responses and indicate that the Ah locus is necessary but not sufficient for mediating the effects caused by 2,3,7,8-TCDD and related compounds. However, despite these complicating factors, the structure-toxicity rela- tionships were also observed for these responses. It is likely that the mechanisms involved in other effects elicited by PCDDs and PCDFs also re- quire other genetic loci and other factors; however, this should not alter the structure-dapendent potencies of these compounds. It should be noted, however, that information questions the unique validity of extending the Ah receptor mechanism for TCOD toxicity to 15 ------- species other than the two strains of mice (C57BL/6J and DBA/2J) in which the correlations are well established. For example, acute lethality (Pohjanvirta et al. 1988), microsomal enzyme induction (Rozman et al. 1985a, 1985b, Henry and Gasiewicz 1986), thymic involution {Gorski et al. 1988), induction of cleft palate (Lamb et al. 1986), induction of hyper- keratosis (Puhvel and Sakamotot 1987), suppression of antibody response to sheep red blood cells (Pazdernik and Rozman 1985), as a consequence of 2,3,7,8-TCDD administration have been shown not to correlate with binding affinity to the Ah-high affinity protein in other strains/species. Also, {Gasiewicz and Rucci 1984) indicate that the correlation between sensitiv- ity to 2,3,7,8-TCDD toxicity and Ah receptor binding affinity seen in mice is not necessarily applicable to other species. Therefore, whenever possible, TEFs are also to be based upon whole animal data, rather than solely "Ah-receptor" binding affinities or enzyme induction. Such data may be developed from other methods such as the early life stage bioassay being developed in the Netherlands (Helder and Seinen 1985). 16 ------- 5. DEVELOPMENT OF TEFs FOR PCDDs AND PCDFs The basic premise of this approach is that the TEF for a given com- pound is assigned based upon its toxic potency relative to 2,3,7,8-TCDD {which is assigned a value of 1). The development and validation of TEFs for individual PCDDs and PCDFs require a detailed examination of all the quantitative SARs (QSARs) that have been reported for this class of compounds. These data can then be used to derive TEFs for all those compounds that have been tested. Moreover, if an individual PCDD or PCDF can be compared to 2,3,7,8-TCDD for more than one Ah receptor-mediated response or in more than one animal species, the TEF values will undoubt- edly encompass a range of values. Regulatory agencies can either develop TEF ranges for individual PCDDs and PCDFs or use a graded system that selects specific TEF values derived from criteria-based toxic or biologic endpoints. For example, Health and Welfare Canada and Ontario Ministry of the Environment (communication from D.L. Grant) have developed an evaluation procedure in which the following responses are weighted in the following descending order of rating priority: 1. Evidence for carcinogencity based on long-term animal studies. 2. Where evidence from long-term studies is absent, data from studies of reproductive effects. Toxicological evidence indicates that doses or exposure levels resulting in reproductive effects overlap the range of doses or exposure levels that result in carcinogenic effects, 3. Other subchronic toxic effects, e.g., thymic atrophy, body weight loss, general toxicity. 4, Acute toxicity studies, e.g., LD50. 5. In vivo or in vitro biological effects, e.g., receptor binding, enzyme induction. It is clear that because of the general lack of carcinogencity data for PCDDs and PCDFs, the effects noted in 2 through 5 will have to be utilized to generate most of the TEFs. This section of the report will briefly summarize the major studies that have investigated in vivo or in vitro QSARs for PCDDs and PCDFs. 17 ------- 5.1 Lethality Table 3 summarizes the acute LD5Q values for 2,3,7,8-TCDD and related PCDD isomers and congeners. The SARs are comparable to those observed for other Ah receptor-mediated biologic and toxic responses (Safe 1986), and these studies provided some of the first experimental evidence demonstrating SARs for PCDDs. Although there are considerable interspe- cies differences with respect to the absolute values for the acute LDcn values, the SARs for these compounds are comparable in all three species. For example, in the guinea pig, the relative potency ratios for 2,3,7,8- tetraCDD/l,2,3,7,8-pentaCDD/ 1,2,3,4,7,8-hexaCDD were 1, 0.2, and 0.008, respectively (using the LDcQ of 0.6 pg/kg). The absolute potency ratios derived from the mouse LDrQ values were different from those observed in the guinea pig, however, the order of relative potencies of the three com- pounds were the same in both animal species. 5.2 In Vivo Biologic and Toxic Responses Several studies have reported the toxic effects of PCDDs, PCDFs and related toxic halogenated aryl hydrocarbons for several receptor-mediated responses (Mason et al. 1985, 1986, 1987; Bandiera et al. 1984; Nagayama et al. 1985; Poland and Knutson 1982; Birnbaum et al. 1987a, 1987b; Davis and Safe 1988; Pluess et al. 1988a, 1988b). Most of these studies are limited with respect to the number of congeners used; however, in all cases the qualitative SARs are comparable to those observed for other receptor-mediated responses. Safe (1987) and coworkers have carried out QSAR studies for a series of PCDD, PCDF, PCB, polybrominated dibenzo- p-dioxins (PBDDs), and halogenated (Br-Cl) dibenzo-p-dioxins in male Wistar rats (inhibition of body weight gain, thymic atrophy), male Hartley strain guinea pigs (inhibition of body weight gain) and male C57BL/6 mice (immunotoxicity) (Safe 1986, 1987; Bandiera et al. 1984, Mason et al. 1985, 1986, 1987; Leece et al. 1985; Holcomb et al. 1988). In addition, the structure-dependent effects of PCDDs, PCDFs, and related compounds on the induction of rat hepatic microsomal AHH and related 18 ------- 8366H Table 3. Comparative LD50 Values for Several PCDD and PCDF Congeners in the Guinea Pig, Mouse, and Rata Congener 2.3.7.8 TetraCOD 2.3-DiCDD 2.7-OiCOO 2.8-OiCDD 1.3.7-TriCDO 2,3,7-TriCDD 1.2.3.4-letraCDD 1.3,6.8-TetraCDD 1.2,3,7,8 PentaCOD 1,2,4,7.8-PentaCOO 1,2,3,4,7,8-HexaCDD l,2,3,6,7,8-Hexa:DD 1,2.3.7.8.9 HexaCOO 1,2.3,4.6,7.8-Hepta COD OCDD 2.8-DiCDF 2,4,8-TriCDF 2,3.7.8-TetraCDF 2,3.4,7,8-PtmtaCOF 2,3.4,6.7.8 HexaCDF Guinea pig Ug/kg) 0.6 2 - - >300.000 29.444 - >15.000.00Q 3.1 1.125 72.5 70-100 60 100 >600 - - 5-10 3-10 120 LD values Mouse Ug/kg) 114-284; 182; 2,570 - >2. 000. 000 8.470,000 >15.000,000 >3.000 - >2. 987. 000 337.5 >5,000 825 1,250 >1,440 >4. 000. 000 >1 5. 000. 000 >15,000.000 >6,000 - - Rat Ug/kg) 22-45; 164-409; >1400 >1. 000, 000 >1. 000, 000 >5, 000, 000 >5, 000, 000 >1. 000, 000 >1, 000, 000 >10,000,000 - - _ - _ > 1,000, 000 >15.000.000 >1 5, 000, 000 >1.000 916 ~ a Kociba and Schwetz 1982. Schwetz et al. 1973. McConnell et al. 1978, Kociba and Cabey 1985, Moore et al. 1979, McKinney et al. 1981; Brewster et al 1988; Chapman and Schiller 1985; Walden and Schiller 1985; Pohjanvirta et al. 1987. 19 ------- monooxygenases have also been determined. The results of all these studies are summarized in Tables 4 and 5. The SARs were comparable for the responses in the three animal spe- cies. In all cases the most toxic compounds were the 2,3,7,8-substituted PCDDs and PCDFs; the removal of lateral Cl groups or the addition of non- lateral Cl substituents resulted in decreased toxic potencies. However, a comparison of the relative toxic potencies of some of the more toxic 2,3,7,8-substituted compounds illustrates some important species and response-dependent differences in relative toxicities. For example, in the rat, 2,3,4,7,8-pentaCDF is consistently more toxic than 1,2,3,7,8- pentaCDF, and the 2,3,4,7,8-pentaCDF/l,2,3,7,8-pentaCDF relative potency ratios were 2.5, 8.4, and 39.7 for inhibition of body weight gain, thymic atrophy, and the induction of hepatic microsomal AHH. Thus, based on studies on the rat, 2,3,4,7,8-pentaCDF exhibited 2.5 to 39.7 times greater potency than 1,2,3,7,8-pentaCDF. These values are well reflected in the relative potency ratio of 10:1 (0.5/0.05), which was assigned for these two isomers in the International-TEF (I-TEF) method. Recent in vivo studies on the subchronic toxicities of 2,3,7,8-TCDD, 1,2,3,7,8-pentaCDD, 1,2,3,6,7,8-hexaCOF, 2,3,4,7,8-pentaCDF and 1,2,3,7,8-pentaCDF in rats gave TEFs of 1.0, 0.4, 0.1, 0.4, and 0.01, respectively (Pluess et al. 1988a, 1988b). The 2,3,4,7,8-pentaCDF/l,2,3,7,8-pentaCDF ratio (i.e., 40:1} was closer to the I-TEF value. A closer examination of the relative activity of 1,2,3,7,8- and 2,3,4,7,8-pentaCDFs in the guinea pig (Table 4) shows that the former isomer is more potent in this species (i.e., for inhibition of body weight gain). The reasons for these species-dependent differences in relative potencies are unknown. Table 5 summarizes the relative toxicities of 2,3,7,8-TCDD and several PCDF congeners in the mouse (Birnbaum et al. 1987a, 1987b; Davis and Safe, 1988). These results further illustrate some species and response- dependent variations in TEFs. For example, 2,3,4,7,8-pentaCDF and 2,3,7,8-TCDD are equipotent as immunotoxins (i.e., inhibition of the 20 ------- 8366H Table 4. A Sunnary of the Dose-Response _!_£ Vivo Biologic and Toxic Effects of Several Halogenated Anomalies in the Immature Male Rat and Guinea Piga Compounds A. PCDDs. PBDDs. and Br-PCDDs 2,3,7,8 tetraCDD 1,2,3,7,8-pentaCDO 1,2,3,4,7,8-hexaCDD 1,3, 7, 8- tetraCDD 1,2,4,7.8-pentaCDD 2,3,7,8-tetraBDD 2,3-dibroitio-7,8-diCDD 2,bromo-3,7,8-triCDD 1.2.3.7.8-pentaBOD 1.2.4.7.8-pentaBDO 1.3,7,8-tetraBDO B. PCDFs 2,3,4,7.8-pentaCDF 1,2,3,4,7,8-hexaCOF 1,2,3,7,8-pentaCOF 2.3,4,6,7,8-hexaCDF 1.2,3,6,7.9-hexaCDF 2,3,7,8-tetraCOF 1.3.4.7.8-pentaCDF 2.3.4.7.9-pentaCDF 2.3.4.7-tetraCDF 1,2,3,7,9-pentaCDF 1.2.4.7,8-pentaCOF 1.2.3,7-tetraCDF 2,3.4,8-tetraCOF 1,2,4,6.7-pentaCDF 1.2.3.6-tetraCOF PCOF mixtureb Inhibition of body weight gain Gu i nea Rat pig 0.05 0.0056 0.62 1.63 132 0.66 34.0 0.061 0.068 0.012 0.12 0.87 12.9 252 1.04 0.012 1.30 2.64 0.0059 2.80 3.20 3.20 26.1 22.0 0.040 34.0 49.3 0.160 49.3 86.9 137 >150 >250 0.52 In Vivo ED (jimol/kq) 50 Thvmiu atroohv Guinea Rat pig 0.09 ND 0.17 1.07 100 ND 11.0 ND 0.034 0.0073 0.035 0.39 6.17 35.5 0.21 ND 0.5 1.76 ND 0.93 0.93 3.60 0.70 5.5 ND 7.84 23.0 ND 46.4 110 >150 >150 >250 0.65 Hepatic AHH Rat 0.004 0.031 0.03 31.2 2.82 0.00076 0.00049 0.0025 0.025 0.195 6.50 0.037 0.293 1.47 0.265 0.347 0.652 3.49 6.96 46.1 14.7 7.80 110 >150 >250 0.016 Induction Guinea pig 0.00028 1.6 0.049 0.0012 0.0059 0.026 0.14 21 ------- r 8366H Table 4. (continued) In Vivo ED 5() Compounds Inhibition of body weiqht qain Gu i nea Rat pig Thymic atrophy Guinea Rat pig Hepatic AHH Induction Guinea Rat pig C. PCBs 2.3.4,4',5-pentaCBP 2.3,3',4.4'.S'-hexaCBP 2,3,3't4,4'.5-nexaCBP 2,3,3'.4.4'-pentaCBP 2,3',4,4',5-pentaCBP 2.3,4,4'.5.-pentaCBP S.S'^^'.S.S'-hexaCBP 3.3'.4.4'.5-pentaCBP 180 220 180 750 1.120 370 15 3.3 200 225 180 1.030 1,550 2.790 8.9 0.95 30 6 25 65 165 130 0.50 1.10 a Mason et al. 1985, 1986; Leece et al. 1985; Hoicomb et al. 1988). This mixture contained the following congeners: 2,3,7,8-tetra-, 1,2,4,7,8-penta- 2,3.4,7.8-penta-, and 1,2,3,4.7.8-hexachlorodiberuofuran. KD = non detect. 1.2.3.7.8-penta-, 22 ------- B3S6H Table 5. ED50 Values for PCDF Congeners in the House: Iirmunotoxicily and Teratogenicily3 ED5Q values (pmol/kg) Compound 2,3,7,8-TCOD 2.3,4,7.8-pentaCDF 1,2,3,7,8-pentaCOF 2,3,7,8-tetraCDF 1.2,3.7,9-pentaCDF 1.2.3.4.7.8-hexaCDF Teratogenicity 0.011 0.09 0.35 0.22 -- 0.84 Imnunotoxicity 0.0024 0.0030 - 0.014 0.710 _ 1,3,6,8-tetraCDF 35.7 "Davis and Safe 1988. Birnbaum et al. 1987a. 1987b. 23 ------- splenic plaque-forming cell response to sheep red blood cells) but the teratogenic activity of 2,3,7,8-TCDD is eight-fold greater than 2,3,4,7,8- pentaCDF. For the teratogenic response (i.e., cleft palate) (Birnbaum et al. 1987b), the 2,3,4,7,8/1,2,3,7,8-pentaCDF potency ratio was less than 4, which is significantly lower than the 10:1 ratio derived from the I-TEF values (i.e., 0.5/0.05). Examination of the data in Tables 4 and 5 illu- strates that several other relative potency ratios are different from the I-TEF values. The species- and response dependent differences in toxic potencies are due to several factors including pharmacokinetic and meta- bolic differences (Brewster and Birnbaum 1987, 1988), route, and duration of administration (Couture et al. 1988). This latter factor is particu- larly important for higher chlorinated PCDDs and PCDFs (Couture et al. 1988) and is discussed later in this document. 5.3 In Vitro Potencies Several groups have developed the use of bioassays for detecting and quantitating toxic halogenated arotnatics (Bradlaw and Casterline 1979; Bradlaw et al. 1980; Eadon et al. 1986; Gierthy and Crane 1985; Knutson and Poland 1980, 1982; Gierthy et al. 1984; Jansing and Shain 1985; Safe 1987). These assay systems primarily utilize mammalian cells in culture, which measure a specific Ah receptor-mediated response (e.g., keratiniza- tion, changes in cell morphology, receptor binding, or enzyme induction) associated primarily with the toxic PCB, PCDF, and PCDD congeners. Bradlaw and coworkers (1979, 1980) first reported that PCBs, PCDDs, and PCDFs readily induce AHH activity in rat hepatoma H-4-II E cells in cul- ture, and they demonstrated the utility of this assay system for detecting toxic halogenated aromatics in diverse matrices including fish extracts, PCB/PCDF-contaminated rice oil, and diverse food extracts including gelatin samples containing pentachlorophenol and trace levels of higher chlorinated PCDDs. In addition, relative Ah receptor binding assays for halogenated aryl hydrocarbons also constitute a potential in vitro assay system (Safe 1987). 24 ------- Table 6 summarizes the competitive Ah receptor binding affinities of several PCODs and PCDFs using rat hepatic cytosol and [3H]-2,3,7,8-TCDD as a radioligand. The qualitative SARs for these responses were compar- able to those observed for the structure-toxicity relationships (Table 4); however, it was apparent that there were major differences in their QSAR data (Bandiera et al. 1984; Mason et al. 1985, 1986). This assay system is limited by solubility problems and the narrow window of relative activ- _p _C ities {i.e., 10 to 10 M using the sucrose density gradient assay system), and recent studies show that relative binding affinity EC50 values using the competitive assay system are not appropriate for quanti- tatively determining structure-dependent differences (Mason et al. 1985, 1986; Bunce et al. 1988). Table 7 summarizes the in vitro induction of AHH and ethoxyresorutin 0-deethylase (EROD) activities by several PCDD and PCDF congeners in rat hepatoma H-4-II E cells in culture. Again, the SARs for this highly char- acteristic Ah receptor-mediated response were comparable to those observed in other studies (see Tables 4 and 6). The monooxygenases are readily induced by low levels of the more toxic halogenated aryl hydrocarbons (10" to 10" M), and differences in congener potencies of > 10 can be determined. Figure 4 illustrates the correlation between the -log EC50 value for AHH induction (in vitro) vs. the -log ED^Q values for in vivo body weight loss, thymic atrophy, and AHH induction (in the rat) for the PCDDs and PCDFs combined (see Table 4). The linear correlation coefficients were r = 0.89, 0.83, and 0.82, respectively. The correlations observed (Figure 5) between the in vitro induction bioassay results and the complete list of halogenated aryl hydrocarbons shown in Table 4 gave even higher r values (0.91, 0.92, and 0.84). It was also reported that the only individual compounds that did not fit in these correlations were compounds that contained two adjacent unsubstituted positions. These compounds tend to be more rapidly metabolized in vivo (Safe 1980) and are less active in the in vivo assays (note: the results for these congeners 25 ------- r 6366H Table 6. PCDDs and PCDFs as Competitive Ligands for the Rat Hepatic Cytosol Receptor SARs Congener Dibenzofuran 2-monoCDF 3-monoCDF 4-raonoCOF 2.3-diCDF 2,6-dlCDF 2.8-diCDF 1,3.6-triCDF 1,3,8-triCDF 2.3,4-trlCDF 2,3,8-triCOF 2,6.7-triCDF 2.3.4,6-tetraCDF 2,3,4.8-tetraCDF 2,3,6.8-tetraCDF 2,3,7.8-tetraCDF 1,2.4.8-tetraCOF 1.2.3.6-tetraCOF 1.2.3,7-tetraCOF 1.3,4,7,8-pentaCDF 2.3,4,7,9-pentaCDF 1,2,3.7.9-pentaCDF 1.2.4.6.7-pentaCDF 1,2.4.7.9-pentaCOF 1.2,3.4.8-pentaCDF 1.2,3,7.8-pentaCDF 1,2,4.7.8-pentaCDF 2,3,4.7.8-pentaCDF 1,2.3 4 7 8-hexaCOF 1.2,3.6.7,8-hexaCDF 1.2.4,6,7.8-hexaCDF 2.3,4,6.7.8-hexaCOF 2.3,7,8-TCDO 2,3.7-triCDD 2,8-diCDD 1,2,3,7.8-pentaCDD 2.3.6.7-tetraCDD 2.3.6-triCDD 1,2.3.4,7,8-hexaCOO 1.3.7.8-tetraCDD 1,2.4,7.8-pentaCOO Receptor binding affinities (EDSQ) (M) <10'3 2.8 x 10~4 4.2 ± 0.6 x 10"5 < 10~3 4.72 x 10"6 2.46 x 10~4 2.57 x 10~4 4.40 x l(f6 8.50 x 10~5 1.9 x 10"5 1.0 ± 0.1 x 10"G 4.5 x 10~7 3.5 x 10~7 2.0 x 10~7 2.2 x 10"7 4.1 ± 0.6 x 10"8 >io-5 3.5 x 10"7 1.1 x 10~7 2.0 x 10"7 2.0 x 10"7 4.0 x 1(T7 6.77 x 10~8 2.0 x 10"5 1.2 x 10"7 7.45 t 2.04 x 10"8b 1.3 x 10"6 flK 1.5 ± 0.1 x 10 HD 2.3 x 10~7 2.7 ± 1.0 x 10~7b 8.3 x 10~6 4.7 ± 0.4 x 10"8b 1.0 x 10"8 7.1 x 10"8 3.2 x 10"6 7.9 x 10"8 1.6 x 10"7 2.2 x 10"7 2.8 x 10~7 7.9 x 10~7 1.1 x 10"6 26 ------- 8366H Table 6. (continued) Congener Receptor binding affinities (E05Q) (M) 1.2,3,4-tetraCDD 1.2.3.4./-pentaCDD 1.2.4-t.riCDD OCDD 1 monoCDD 1.3 x 10 6.4 x 10 1.3 x 10 -6 -5 >1.0 x 10 >1.0 x 10~ -5 Competitive EDrn values were determined using the sucrose density gradient assay system. Dose-response experiments carried out in triplicate and expressed as means + SD. 27 ------- 8366H Table 7. PCDDs and PCDFs as Inducers of AHH and EROD Activities in Rat Hepatoma H-4-II E Cells in Culture: SARs Congeners A. PCDFs Oibenzofuran 2-monoCDF 3-monoCDF 4-monoCDF 2.3-diCOF 2,6-diCDF 2.8-diCDF 1.3.6-triCDF 1.3.8-triCDF 2,3,4-triCDF 2,3,8-triCDF 2.6.7-triCDF 2,3,4,6-tetraCDF 2.3,4,8-tetraCDF 2.3,6,8-tetraCDF 2,3,7,8-tetraCDF 1,2,4,8-tetraCDF 1,2,3,6-tetraCDF 1.2.3.7-tetraCOF 1.3,4,7,8-pentaCDF 2.3,4,7,9-pentaCDF 1,2.3.7,9-pentaCDF 1.2.4,6.7-pentaCDF 1,2,4.7,9-pentaCDF 1.2.3.4.8-pentaCDF 1,2.3,7.8-pentaCDF 1,2,4,7.8-pentaCDF 2.3.4,7.8-pentaCDF 1,2,3.4.7.8-hexaCOF 1,2.3.6,7.8-hexaCDF 1,2,4.6,7,8-hexaCDF 2,3,4.6.7.8-hexaCDF B. PCOOs 2,3,7,8-tetraCDO 1,2,3,7,8-pentaCDD 2,3,6,7-tetraCDO 2,3,6-triCOD 1,2,3,4,7,8-hexaCDD AHH (H) ND ND ND 1.0 x 10"5 2.19 x 10"B 6.17 x 10"5 3.95 x 10~5 2.53 x 10~6 1.94 x 10"5 1.51 x 10"7 2.49 x 10"6 2.80 x 10"6 1.32 x 10~6 4.14 x 10"8 1.04 x 10~6 3.91 x 10"9 1.20 x 10"5 >10~* 2.70 x 10"5 1.60 x 10"9 7.90 x 10~9 8.60 x 10~8 3.25 x 10~7 3.77 x 10"8 2.09 x 10~7 2.54 x 10~9 1.06 x 10~7 2.56 x 10~10 3.56 x 10~10 1.47 x 10"9 4.24 x 10~8 6.87 x 10'10 7.2 x 10"11 1.1 x 10~8 6.1 x 10'8 -_ 2.1 x 10"9 EROD (M) ND ND ND 1.71 x 10"5 4.84 x 10"6 6.31 x 10"5 4.0 x 10"5 3.37 x 10"6 3.02 x 1C"5 2.48 x 10"7 1.56 x 10"6 3.13 x 10"6 1.13 x 10~6 3.76 x 10~8 7.79 x 10"7 2.02 x 10"9 9.26 x 10"5 >10^4 6.30 x 10"5 5.80 x 10~9 5.80 x 10~9 8.60 x 10"8 3.48 x 10~7 3.84 x 10~8 1.63 x 10~7 3.06 x 10"9 1.48 x 10"7 1.34 x 10"10 3.79 x 10~10 1.24 x 10"9 2.93 x 10"8 5.75 x 10"10 1.9 x 10"10 1.7 x 10~8 1.1 x 10"8 __ 4.1 x 10"9 28 ------- 8366H Table 7. (continued) Congeners AHH (M) EROD (M) B. PCOOs (continued) 1.3.7,8-tetraCOO 1,2,4,7,8-pentaCDD 1.2.3.4-tetraCDD 2.3.7-triCOO 2,8-diCDO 1.2,3,4.7-pentaCDD 1.2.4-triCDO OCDD 1-monoCDD 5.9 x 10 2.1 x 10 3.7 x 10 3.6 x 10 -8 -6 -7 >1.0 x 10 -4 -7 6.6 x 10 4.8 x 10" >1.0 x 10 >1.0 x 10" -4 3.2 x 10 1.1 x 10 2.4 x 10" 1.4 x 10 r7 -8 -7 >1.0 x 10 8.2 x 10 2.2 x 10 -7 -6 >1.0 x 10 >1.0 x 10 -4 -4 29 ------- AHH INDUCTION: in vitro vs. In vivo PCDDs and PCDFs 11 10 - 9 - e - 7- R a 0.82 5678 •toy HMO AHH INDUCTION (BAT) *« !* - - « AHH INDUCTION vs. BODY WEIGHT LOSS PCDDs and PCDFs e *> e 3 11 10- 9 - e 7H 6 Re 0.89 3 4 5 6 7 C -log ED-50 BODY WEJGHT LOSS (RAT) AHH INDUCTION vs. THYMIC ATROPHY PCDDs and PCDFs e •S I s ti 4567 •Jos EO-50 THYUIC ATROPHY (RAT) 30 c E o^T ^ ^* « Q> LL. z «2 io . 0> C- O - ag§ Sf=> Sa — LU Q. Sli E o I X — » «g i c S •r 5* 2. *i°i o&Iil SxSic £«2^§ *; OC u -5 « <3<=!i<5E UJ DC O ------- AHH INDUCTION: In vitro vs. In vivo PCBs, PCDDs, PCDFs, PBDDs o I s 11 10 - 9- 8- 7- 6- 5- 4 R 00.84 56789 •log ED-50 AHH INDUCTION (RAT) 10 AHH I '> c 5 INDUCTION vs. BODY WEIGHT LOSS PCBs, PCDDs, PCDFs, PBDDs 34567 -log ED-50 BODY WBQHT LOSS (RAT) AHH INDUCTION vs. THYMIC ATROPHY PCBs, PCDDs, PCDFs, PBDDs | X 3: S? ti = cc in UJ DC O 345678 -log ED-50 THYUIC ATROPHY (RAT) 31 ------- are not included in the Figures 4-7). It should also be noted that there are good correlations between the in vitro AHH induction potencies for PCDDs and PCDFs and their toxic potencies (body weight loss and immunotox- icity) in other animals (guinea pigs and mice), as illustrated in Figures 6 and 7. The excellent correlation between the in vivo and in vitro potencies for PCDDs and PCDFs is consistent with the proposed receptor-mediated mechanism of action of PCDDs, PCDFs, and related compounds. Moreover, the correlation observed between the in vitro AHH induction bioassay results and the in vivo toxicity data {Figures 4 through 7) confirms the utility of both in vivo and in vitro assay systems for estimating the TEF for mixtures of PCDDs, PCDFs, and related compounds. It should also be noted that other in vitro bioassays that measure an Ah receptor-mediated response could also be used to estimate TEFs for toxic halogenated aryl hydrocarbons. 5.4 Toxicity Equivalency Factors Several groups have reported TEF values for the 2,3,7,8-substituted PCDDs and PCDFs, and these have been summarized (Bellin and Barnes 1987, NATO/CCMS 1988). Prior to the development of the I-TEF values by the Pilot Study on International Information Exchange on Dioxins and Related Compounds, numerous slightly-different TEF schemes were used throughout the world. As a result of the existence of so many methods, the communi- cation of the toxicological significance of a set of analytical data was often hindered by the absence of a standardized approach. One of the goals of the Pilot Study was to achieve consensus on a specific TEF meth- odology. In addition to the addition of the most recent toxicological data, the I-TEFs were developed with simplicity as one of the underlying principles. Therefore, in developing the I-TEFs, order of magnitude values were generally used rather than the precise numeric values. Table 8 summarizes the I-TEF values for the 2,3,7,8-substituted PCDDs and PCDFs and the ranges of TEF values that have been derived from the data presented in Tables 3 through 7. The choice of a single TEF for an indi- vidual PCDD or PCDF was based on several factors, including those noted by Health and Welfare Canada and Ontario Ministry of the Environment. Two 32 ------- o ' O O S a CB O 11 10- 9- 4 8- 7- 2,3,7.8-TCDD • 2.3,4,7,8-PeCDF ^ 2.3,4.7,9-PeCDF \ 1.2.4,7,8-PeCDD 1,2,3,7,8-PeCDF 1,2,3,7,9-PeCDF 1,3.7,8-TCDD R = 0.93 789 •log ED-50 BODY WEIGHT LOSS - GUINEA PIG FIGURE 6. Correlation Between -log ECM (M) for AHH Induction in Rat Hepatoma H-4-H E Cells vs. -log ED^ (moI/kg) Inhibition of Bodyweight Gain (Guinea Pig) 33 ------- LU s* 2,3.7.8-TCDD 2,3,4.7.8-TCDD - 56789 -tog ED-50 {mol/kg) IMMUNOTOXICJTY - C57BL/6 MICE FIGURE 7. Correlation Between -log ECM (M) for AHH Induction in Rat Hepatoma H-4-II E Cells vs. -log EDW (mol/kg) Immuno- toxicity (C57BL/6 Mice) 34 ------- 8366H Table 8. International Toxicity Equivalency Factors (I-TEFs):* Comparison of Relative Potency Data for the 2,3,7,8-Substituted PCDDs and PCOFs Congener 2.3.7.8-tetraCDD 1,2,3,7,8-pentaCDD 1.2.3.4.7.8-hexaCOO 1.2.3.7.8.9-hexaCDDNTP l,2,3,6,7,8-hexaCDDNTP 1,2,3,4,6,7,8-heptaCOO OCDO 2.3.7,8-tetraCOf 2.3.4.7,8-pentaCDF 1.2.3,7.8-pentaCDF 1,2,3,4,7,8-hexaCOF 1,2,3,6,7.8-hexaCOF Observed I-TEF in vivo toxic it ies 1 1 0.5 0.59 - 0.053 (0.59m1. 0.42gl. 0.081r, 0.053r) 0.1 0.24 - 0.018 (0.24m1. 0.084r. 0.018gl. 0.13r) 0.1 0.14 - 0.016 (0.016g1. 0.14m1) 0.1 0.16 - 0.015 (0.16m1. 0.015g1) O.Cll 0.001 0.1 0.17 - 0.016 (0.017gl. 0.17m1. 0.05mt. 0.025r. 0.016r) O.S 0.8 - 0.048 (0.8m1, 0.479, 0.43r, 0.13g1 (0.12"*. 0.0481") 0.05 0.95 - 0.019 (0.95s. 0.05r. 0.031mt. 0.019r) 0.1 0.18 - 0.013 (0.18r. 0.0381", 0.013mt) 0.1 0.097 - 0.016 (0.097r. 0.016r) TEF ranges AHH in vivo 1 0.13 (0.13r) 0.13 (0.13r) - 0.0002C 0.006 (0.006r) 0.23 - 0.11 (0.239 - O.ll1") 0.047 - 0.003 (0.0479, 0.003r) 0.014 (0.014r) 0.012 (0.0121") induction in vitro 1 0.011 - 0.0065 (0. 011/0. 0065h) 0.046 - 0.034 (0. 034/0. 046h) 0.008 (0.008hB) 0.012 (0.012hB) 0.003 (0.003hB) 0.0006 , till' (0.0006nB 0.09 - 0.018 (0. 018/0. 09h) 1.41 - 0.28 (0.28/1.41h) 0.06 - 0.028 (0.06/0,028h) 0.50 - 0.20 (O^O/O.SO*1) 0.153 - 0.04 (0. 049/0. 153h) 35 ------- 8366H Table 8. (continued) Observed TEF ranges Congener I-TEF In vivq toxic Hies AHH induction in vivo in vitro 1.2.3.7.8,9-hexaCDF 0.1 2,3.4.6.7,8-hexaCDF 0.1 1.2,3,4,6,7.8-heptaCDF 0.01 1,2.3,4,7.8.9-heptaCOF 0.01 OCDF 0.001 0.097 - 0.011 (0.097r. 0.018r. 0.0119) 0.015 (0.015r) 0.33 - 0.11 (0.11/0.33h) ^Guinea pig and rrat data (Table 4). 91 Guinea pig and mouse lethalities {Table 3). Mouse teratogenicity and ""mouse imnunotoxicity (Table 5). W hepatoma data (AHH/EROD) (Table 7). hBRat hepatoma data (AHH) (Bradlaw and Casterline 1979). °Couture et al. 1988. MTP, NTP (1980) carcinogenic potency values - 0.04. Taken from NATO/CCNS 1988. 36 ------- conservative approaches would be (a) to utilize the highest observed TEF for each compound or (b) to use the TEF values for in vitro AHH (or EROO) induction since these values would minimize the species-dependent pharma- cokinetic differences in congener potencies. Another option would utilize a TEF range for each compound. These are some of the factors that were considered in finalizing the I-TEF values given in Table 8. One of the most noteworthy deviations from previous TEF methods is the assignment of values to OCDD and OCDF. In previous schemes, these congeners received values of zero on the basis of limited short-term in vivo and in vitro data. In a recently published study (Couture et al., 1988), however, male rats were exposed to low levels of OCDD for 13 weeks. At the end of the experiment, the animals were beginning to show signs of toxicity that were reminiscent of "dioxin toxicity." Detectable levels of OCDD had accumulated in the organism. These data suggest that OCDD exhibits minimal toxicity in short-term studies simply because so little of the compound is absorbed in a short time. However, after multiple exposures for longer periods, the animals appear to absorb and bio- accumulate sufficient amounts of OCDD to manifest "dioxin-like" effects. Based on these new data a value of 0.001 has been assigned to both OCDD and OCDF in the I-TEF scheme. Another modification made in the I-TEF scheme is the elimination of a factor for non-2378 substituted dioxins and furans. In several past methods, including the EPA method (Bellin and Barnes 1987), the non-2378- substituted congeners were assigned a value of 1% of the value of the re- spective 2378-substituted compound. During the past two years, scientists have gathered additional data indicating that nearly all of the 210 PCDDs/ PCDFs can be found at very low levels in many parts of the environment. However, it appears that the 2378-substituted congeners are selectively absorbed and/or retained in higher animals; e.g., fish, humans, and other mammals. That is, of the PCDDs/PCDFs detected in a variety of samples of biological tissues (e.g., human, mammalian animals, and fish) the 2378- PCDDs/PCDFs congeners clearly predominate over the non-2378-substituted 37 ------- congeners. This is true even when the source of the PCDDs/PCDFs is rela- tively low in the concentration of 2378-substituted congeners. For example, flyash from municipal waste combustors (MWCs) generally contains detectable amounts of PCDDs/PCDFs. In most instances, the amount of non-2378-substituted congeners vastly outweighs the amount of 2378- substituted PCDDs and PCDFs in such samples. However, when mice or fish are exposed to MWC flyash and their tissues are subsequently analyzed for the presence of PCDDs/PCDFs, essentially only the 2378-substituted are detected (Kuehl et al., 1986; Van den Berg et al., 1985). Similarly, the "background levels" of CDDs/CDFs routinely found in human tissues (fat, blood, and milk) contain almost exclusively the 2378-substituted compounds (Rappe et al., 1987). The environmental concern rests primarily with long-term exposures. Since the non-2378-substituted congeners appear to be either not absorbed or quickly eliminated by biological systems, it is the 2378-substituted congeners that seem to pose the greatest long-term potential. .Therefore, in the interest of keeping the TEF system as simple as possible, attention is focused exclusively on 2378-congeners in the I-TEF scheme. The I-TEF scheme introduces an additional complexity that was not a part of the previous schemes. In the I-TEF scheme, the 2,3,4,7,8-PeCDF is assigned a value of 0.5, while the 1,2,3,7,8-PeCDF is assigned a value of 0.05. This is the only instance in which the I-TEFs depart from the guiding principle of "simplicity" in which TEFs are expressed as rounded orders of magnitude. This departure is prompted by a growing body of data that indicate that 2,3,4,7,8-PeCDF is notably more active than originally thought. Based upon the data in Table 8, it can be seen that the 0.5 value for 2,3,4,7,8-PeCDF gains support from the in vivo thymic atrophy data (0.43) and the mouse immunotoxicity data (0.8). The 0.05 value for 1,2,3,7,8-PeCDF gains support from the in vivo investigations of thymic atrophy (0.05) and the in vivo and in vitro investigations of enzyme induction (0.003-0.06). Note that there is one outlier in the 38 ------- eight data points reported for 1,2,3,7,8-PCDF in Table 8. Specifically, there is a 0.95 value recorded for reduction in body weight gain seen in guinea pigs. This one experiment in one laboratory should be investigated further to determine its possible significance. At the present time, how- ever, the weight of the evidence argues for the lower TEF. The fact that the two 2378-substituted congeners can elicit such dif- ferent biological responses can be rationalized by examining the stereo- chemistry of the two chemicals (Bandiera et al., 1984). When superimposed on the molecular structure of 2,3,7,8-TCDD, the C-4 of the "bent" PeCDF is more stereochemically a "lateral position" (i.e., closer to C-3 on the 2,3,7,8-TCDD skeleton), while the C-l is even less stereochemically a "lateral position" (i.e., farther away from C-2) (see Figure 8). There- fore, the 2,3,4,7,8-PeCDF would theoretically be expected to be more active than the 1,2,3,7,8-PeCDF since it has more chlorine substituents in the lateral positions. 39 ------- Cl Cl 1, 2, 3, 7, 8 - PeCDF on 2, 3, 7, 8 - TCDD 2, 3, 4, 7, 8 - PeCDF on 2, 3, 7, 8 - TCDD Figure 8. 2378 - Substituted Pentachlorodibenzofurans (Adapted from Bandlera et al., 1984) 40 ------- 6. BIOASSAVS FOR HAZARD ASSESSMENT OF PCDD AND PCDF MIXTURES The previous sections of this report have confirmed that the SARs for PCDDs and PCDFs are similar for most bioassays (in vivo and in vitro). It is also evident that relative potencies for individual compounds can be variable and are both assay- and species-dependent. This is not sur- prising since there are undoubtedly species-dependent differences in the pharmacokinetics of individual congeners and this affects the delivered dose to the target organ/cells. Despite these variables, the correlations between the in vitro and in vivo QSARs are remarkably consistent (Figures 4 through 7). These findings provide further support for the proposed Ah receptor mediated mechanism of action for 2,3,7,8-TCDD and related compounds. Thus, in vitro systems which measure a specific Ah receptor mediated response such as the in vitro AHH induction assay (Bradlaw and Casterline, 1979) or the keratinization/flat cell assay (Gierthy and Crane, 1985) should be useful for determining the TEF values for complex mixtures. Two recent studies by Safe and coworkers have utilized in vitro and in vivo bioassays to estimate the relative toxicities of two extracts con- taining complex irixtures of halogenated dibenzo-p-dioxins and dibenzo- furans (Table 9) (Safe et al. 1987, Zacharewski et al. 1988). The dose- response in vivo and in vitro effects of these extracts were directly compared to results obtained for 2,3,7,8-TCDD. Using sample dilution factors, one can determine the relative amount or concentration of analyte required to elicit one-half maximal response (ECcQ or EDj-Q). This amount/concentration is equivalent to the known amount/concentration of 2,3,7,8-TCDD required to cause this same magnitude of response. Using this approach, one can readily calculate the "2,3,7,8-TCDD equivalents" in an extract. GC-MS analysis of the PCDDs and PCDFs in a fly ash extract from a municipal incinerator indicated a total of 3,830 and 5,520 ng/g of these compounds, respectively. Using the in vitro enzyme induction bioassay procedure (i.e., rat hepatoma H-4-II E cells), the estimated "2,3,7,8-TCDD 41 ------- 8366H Table 9. Braninated Aromatic Flame Retardant Pyrolysis and Municipal Fly Ash Extracts: In Vitro and In Vivo Determination of "2,3,7.8-TCDD Equivalents"3 Sample "2.3.7.8-TCDD equivalents" loam) FireHasterb Bioassay 300 BA AHH induction 174 (in vitro) EROD induction 194 (in vitro) AHH induction ( in vivo) EROD induction (in vivo) Body weight loss (in vivo) Thymic atrophy (in vivo) PBDDs plus PBDFs 10.935 (total ppm)c PCODs plus PCOFs (total. ppm}c FireMasterb Bromka1b Bromkalb Bromkalb Fly ash BP-6 70-5-OE 70-DE GI extract 1,400 2.140 8.780 3.920 0.105 480 4.680 6,740 5,260 540 - 5,200 - 0.075 520 - 3,860 760 - 6.260 1,680 - 8,960 2,070 610.390 268.480 547.700 9.35 a Safe et al. 1987. Zacharewski et al. 1988. Brominated flame retardants (FireMaster BP-6 is a polybrom mated biphenyl and the remainder of flame retardants are pnlybrominated diphenylethcrs). c Determined by GC-MS analysis. 42 ------- equivalents" in this extract were 105 ng/g fly ash. The dose-response in vivo induction of AHH in the rat was also determined, and the estimated "2,3,7,8-TCDD equivalents" in this extract were 75 ng/g fly ash. This confirms the correlation between the in vivo and in vitro bio-assays and illustrates that the "2,3,7,8-TCDD equivalents" of these PCDD, and PCDF- containing extracts are significantly lower than the total concentration of these compounds in the sample extract as determined by GC-MS analysis. Using a comparable approach, the "2,3,7,8-TCDD equivalents" (in vitro) were determined for several extracts of pyrolyzed brominated (biphenyl and diphenylether) flame retardants. The GC-MS analysis of the extracts revealed a complex mixture of PBDDs and PBDFs for which very few analyti- cal standards are available. The ranges of "2,3,7,8-TCDD equivalent" levels (ug/g or ppm) derived from the AHH and EROD bioassays for each of the pyrolyzed flame retardant samples were 174-194, 480-1,400, 2,140- 4,680, 6,740-8,780, and 3,920-5,260 ppm for FireMaster 300 BA, FireMaster BP-6, Bromkal 70-5 DE, Bromkal 70-DE, and Bromkal Gl, respectively. The in vivo dose response effects of two pyrolyzed flame retardant extracts were determined in immature male Wistar rats and compared to the dose- response activities of 2,3,7,8-TCDD. The in vivo responses measured included hepatic microsomal AHH and EROD induction, body weight loss, and thymic atrophy in the rat (Zacharewski et al. 1988). For the pyrolyzed FireMaster BP-6 and Bromkal 70 DE samples, the in vivo "2,3,7,8-TCDD equivalents" (ppm in sample) for the four in vivo bioassays were 520- 1,680 ppm and 3,860-8,960 ppm, respectively, and the in vitro "2,3,7,8- TCDD equivalents" were 480-1,400 and 6,740-8,780 ppm, respectively. The excellent overlap between the in vivo and in vitro "2,3,7,8-TCDD equiva- lents" data for the two flame retardant pyrolysate extracts supports the utility of the in vitro induction bioassay for quantitatively determining the relative toxic potencies associated with mixtures containing toxic halogenated aryl hydrocarbons. 43 ------- Eadon and coworkers (1986) have also summarized studies that compare the calculated "2,3,7,8-TCDD equivalents" with various dose-related end- points. Table 10 summarizes the "2,3,7,8-TCDD equivalents" determined in the guinea pig for several responses using soot from the Binghamton office building (De Caprio et al. 1983, 1986). The soot was contaminated with PCDDs, PCDFs, and polychlorinated biphenylenes; the estimated TEF, based on analytical chemical data, was 22 ppm (i.e., 2,3,7,8-TCDD equivalents). The experimentally determined "2,3,7,8-TCDD equivalents" from the in vivo studies in guinea pigs varied from 2 to 21 ppm. This observed range was response-dependent and correlated well with previous studies in the rat and with the TEF ranges summarized in Table 8. These results clearly demonstrate that hazard assessments of toxic mixtures of PCDDs and PCDFs and related compounds are readily determined using both in vivo and in vitro bioassays. Invariably, for either indi- vidual PCDD and PCDF congeners or mixtures of these compounds, multiple bioassays will give a range of TEFs that exhibit overlap with TEFs esti- mated by conventional analytical (chemical) approaches. The range of TEFs obtained from multiple in vivo bioassays will also generally overlap with TEFs derived from in vitro bioassays. This suggests that the short- term in vitro bioassays are useful and validated methods for estimating "2,3,7,8-TCDD equivalents" in complex mixtures of PCDDs, PCDFs, and related compounds. 44 ------- 8366H Table 10. Calculated 2,3,7,8-TCDD-Equivalent Concentrations of the Binghampton Soot for Various Dose-Related Endpoints Following Subchronic Exposure3 Endpoint Sex Hethodc 2,3.7,8-TCDD- equivalent concentration in soot (ppm) Relative thymus weight (decrease) Linear 0.951 19" Percent of initial body weight (decrease) Linear 0.994 Serum triglycerides (increase) Linear 0.998 Serum ALT (decrease) Log 0.960 1BU Hepatocellular cyto- plasmic inclusion bodies Mortality F M ED50 LD50 10 (4-28)9 2 (1-3)9 * Eadon ct al.. 1986. Response data from the previously reported Binghamton soot (DeCaprio et al. 1983). and B.3,7,8 TCDD subchronic studies, (DeCaprio et al. 1986) were used to calculate 2,3,7,8-TCDO-equivalent concentrations for the soot. c Method by which 2,3,7.8-TCDD dose-response standard curve was constructed: Linear = linear regression analysis of dose level (ppt) vs. response; LOG = linear regression analysis of log dose level (ppt) vs. response; ED™ = calculation of ED^g dose level by method of Carmines et al. (1980); LD^Q for subchronic exposure by method of Carmines et al. (1980). Correlation coefficient from linear regression analysis. e Calculated using response data from 1.9-ppm Binghamton soot dose level. Calculated using response data from 3.9-ppm Binghamton soot dose level. ^ 95 percent confidence limits. 45 ------- REFERENCES Ballschmiter K, Zoller W, Scholz CH, Nottrodt A. 1983. Occurrence and absence of polychlorodibenzofurans and polychlorodibenzodioxins in fly ash from municipal incinerators. Chemosphere 12:585-594. Bandiera S, Sawyer T, Romkes M, Zmudzka B, Safe L, Mason G, Keyes B, Safe S. 1984. Polychlorinated dibenzofurans (PCDFs): effects of structure on binding to the 2,3,7,8-TCDD cytosolic receptor protein, AHH induction and toxicity. Toxicology 32:131-144. Bannister RD, Davis D, Zacharewski T, lizard I, Safe S. 1987. Aroclor 1254 as a 2,3,7,8-tetrachlorodibenzo-p-dioxin antagonist: effects on enzyme induction and immunotoxicity. Toxicology 46:29-42. Bell in JS, Barnes DG. 1987. Interim procedures for estimating risks associated with exposures to mixtures of chlorinated dibenzo-p-dioxins and dibenzofurans (CDDs and CDFs). Risk Assessment Forum. USEPA 625/3-87/012. Birnbaum LS, Harris MW, Barnhart ER, Morrissey RE. 1987a. Teratogenicity of three polychlorinated dibenzofurans in C57BL/6N mice. Toxicol. Appl. Pharmacol. 90:206-216. Birnbaum LS, Harris MW, Crawford DO, and Morrissey RE. 1987b. Tetratogenic effects of polychlorinated dibenzofurans in combination in C5713L/6N mice. Toxicol. Appl. Pharmacol. 91, 246-255 Bowes GW, Mulvihill MJ, Simoneti BRT, Burlingame AL, Risebrough RW. 1975. Identification of chlorinated dibenzofurans in American polychlorinated biphenyls. Nature 256:305-307. Bradlaw JA, Casterline, JR. 1979. Induction of enzyme activity in cell culture: a rapid screen for detection of planar polychlorinated organic compounds. J. Assoc. Off. Anal. Chem. 65:904-906. Bradlaw JA, Garthoff LL, Hurley NE, Firestone D. 1980. Comparative induction of aryl hydrocarbon hydroxylase activity in vitro by analogues of dibenzo-p-dioxin. Food Cosmet. Toxicol. 18:627-635. Brewster DW, Birnbaum LS. 1987. Disposition and excretion of 2,3,4,7,8- pentachlorodibenzofuran in the rat. Toxicol. Appl. Pharmacol. 90:243-252. Brewster DW, Birnbaum LS. 1988. Disposition of 1,2,3,7,8- pentachlorodibenzofuran in the rat. Toxicol. Appl. Pharmacol. 95:490-498. Bunce NJ, Landers JP, Safe S. 1988. Kinetic models for association of 2,3,7,8-tetrachlorodibenzo-p-dioxin with the AH receptor. Arch. Biochem. Biophys. 267:384-397. 46 ------- Buser HR, Bosshardt HP, Rappe C. 1978. Identification of polychlori- nated dibenzo-p-dioxin isomers found in fly ash. Chemosphere 7:165-172. Buser HR, Rappe C. 1980. High resolution gas chromatography of the 2,2- tetrachlorodibenzo-p-dioxin isomers. Anal. Chem. 52:2257-2262. Buser HR. 1979. Formation of PCDFs and PCDDs from the pyrolysis of chlorobenzenes. Chemosphere 8:415-424. Carlstedt-Duke JMB. 1979. Tissue distribution of the receptor for 2,3,7,8-tetrachlorodibenzo-p-dioxin in the rat. Cancer Res. 39:3172-3176. Carmines EL, Carchman RA, Borzelleca JF. 1980. A method for the evaluation of dose-effective data utilizing a programmable calculator. J. Environ. Pathol. Toxicol. 4:23-30. Chapman DE, Schiller CM. 1985. Dose-related effects of 2,3,7,8- tetrachlorodibenzo-p-dioxin (TCDD) in C57BL/6J and DBA/2J mice. Toxicol. Appl. Pharmacol. 78:147-157. Couture LA, Elwell MR, Birnbaum LS. 1988. Dioxin-like effects observed in male rats following exposure to octachlorodibenzo-p-dioxin (OCDD) during a 13 week study. Toxicol. Appl. Pharmacol. 93:31-36. Crummett WB. 1983. Status of analytical systems for the determination of PCDDs and PCDFs. Chemosphere 12:429-446. Czuczwa JM, McVeety BD, Hites RA. 1984a. Environmental fate of combustion-generated polychlorinated dioxins and furans. Environ. Sci. Technol. 18:444-450. Czuczwa JM, McVeety BD, Hites RA. 1984b. Polychlorinated dibenzo-p- dioxins and dibenzofurans in sediments from Siskiwit Lake, Isle Royal. Science 226:568-569. Davis D, Safe S. 1988. Immunosuppressive activities of polychlorinated dibenzofuran congeners: quantitative structure activity relationships and interactive effects. Toxicol. Appl. Pharmacol. 94:141-149. De Caprio AP, McMartin DN, O'Keefe PW, Ref R, Silkworth JB, Kaminsky LS. 1986. Subchronic oral toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in the guinea pig: comparisons with a PCB-containing transformer fluid pyrolysate. Fundam. Appl. Toxicol. 6:454-463. De Caprio AP, McMartin DN, Silkworth JB, Ref R, Pause R, Kaminsky L. 1983. Toxicity in guinea pigs of soot from a polychlorinated biphenyl- containing transformer fire. Toxicol. Appl. Pharmacol. 6:308-322. 47 ------- Denison MS, Wilkinson CF, Okey AB. 1986. Ah receptor for 2,3,7,8- tetrachlorodibenzo-p-dioxin: comparative studies in mammalian and nonmammalian species. Chemosphere 15:1675-1682. Eadon G, Kaminsky L, Silkworth J, Aldous K, Hilker D, O'Keefe P, Smith R, Gierthy J, Hawley J, Kim N, De Caprio A. 1986. Calculation of 2,3,7,8- TCDD equivalent concentrations of complex environmental contaminant mixtures. Environ. Health Persp. 70:221-227. Farrell K, Safe L, Safe S. 1987. Synthesis and Ah receptor binding properties of radiolabeled polychlorinated dibenzofuran congeners. Arch. Biochem. Biophys. 259:185-195. Gasiewicz TA, Rucci G. 1984. Cytosolic receptor for 2,3,7,8- tetrachlorodibenzo-p-dioxin: evidence for a homologous nature among various mammalian species. Mol. Pharmacol. 26:90-98. Gierthy JF, Crane D, Frenkel GD. 1984. Application of an in vitro keratinization assay to extracts of soot from a fire in PCB-containing transformer. Fund. Appl. Toxicol. 4:1036-1041. Gierthy JF, Crane D. 1985. In vitro bioassay for dioxin-like activity based on alterations in epithelial cell proliferation and morphology. Fund. Appl. Toxicol. 5:754-759. Gorski, et al. 1988. Elevated plasma corticosterone levels and histopathology of the adrenals and thymuses in 2,3,7,8-terachlordibenzo-p- dioxin (TCDD)-treated rats. Toxicology, in press. Greig JB, et al. 1984. Incomplete correlation of 2,3,7,8- tetrachlorodibenzo-p-dioxin hepatoxicity with Ah phenotype in mice. Toxicol. Appl. Pharmacol. 74:17-25. Haake JM, Safe S, Mayura K, Phillips TD. 1987. Aroclor 1254 as an antagonist of the teratogenicity of 2,3,7,8-tetrachlorodibenzo-p- dioxin. Toxicol. Lett. 38:299-305. Helder T, Seinen W. 1985. Standardization and application of an E.L.S-bioassay for PCDDs and PCDFs. Chemosphere 14:183-193. Henry EC, Gasiewicz TA. 1986. Effects of thyroidectomy on the Ah- receptor and enzyme inducibility by 2,3,7,8-tetrachlorodibenzo-p-dioxin in the rat liver. Chem-Biol Interactions 59:29-42. Hochstein JR, Aulerich RJ, Bursian SJ. 1988. Acute toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin {TCDD) to mink. Arch. Environ. Contam. Toxicol. 17:33-37. 48 ------- Holcomb M, Yao C, Safe S. 1988. The biologic and toxic effects of polychlorinated dibenzo-p-dioxins and dibenzofurans in the guinea pig: quantitative structure-activity relationships. Biochem. Pharmacol 37:1535-1539. Jansing RL, Shain W. 1985. Aryl hydrocarbon hydroxylase induction in rat hepatocytes in primary culture by several chlorinated aromatic hydrocarbons including 2,3,7,8-tetrachlorodibenzo-p-dioxin. Fund. Appl. Toxicol. 5:713-720. Jones KG, Sweeney GD. 1980. Dependence of the porphyrogenic effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin upon inheritance of aryl hydrocarbon hydroxylase responsiveness. Toxicol. Appl. Pharmacol. 53:42-49. Kahn PC, Gochfield M, Hygren M, Hansson M, Rappe C, Velez H, Ghent- Guenther T, Wilson WP. 1988. Dioxins and dibenzofurans in blood and adipose tissue of Agent Orange-exposed Vietnam veterans and matched controls. J. Amer. Med. Assoc. 259:1661-1667. Keyes B, Piskorska-Pliszczynska J, Safe S. dibenzofurans as 2,3,7,8-TCDD antagonists: monooxygenase induction. Toxicol. Lett. 31 1986. Polychlorinated in vitro inhibition of 151-158. Kimbrough RD, Falk H, Stehr P, Fries G. 1984. Health implications of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) contamination of residential soil. J. Toxicol. Environ. Health 14:47-93. Knutson JC, Poland A. 1980. Keratinization of mouse teratoma cell line XB produced by 2,3,7,8-tetrachlorodibenzo-p-dioxin: An in vitro model of toxicity. Cell 22:27-36. Knutson JC, Poland A. 1982. 2,3,7,8-Tetrachlorodibenzo-p-dioxin: examination of biochemical effects involved in the proliferation and differentiation of XB cells. J. Cell Physiol. 21:243-151. Kociba RJ, Cabey 0. 1985. Comparative toxicity and biological activity of chlorinated dibenzo-p-dioxins and furans relative to 2,3,7,8- tetrachlorodibenzo-p-dioxins (TCDD). Chemosphere 15:649-660. Kociba RJ, Keyes DG, Beyer JE, Carreon RM, Wade CE, Dittenber DA, Kalnins RP, Franson LE, Park CN, Bernard SD, Hummel RA, Humiston CG. 1978. Results of a two-year chronic toxicity and oncogenicity study of 2,3,7,8- tetrachlorodibenzo-p-dioxin in rats. Toxicol. Appl. Pharmacol. 46:279-303. Kociba RJ, Schwetz BA. 1982. Toxicity of 2,3,7,8-tetrachlorodibenzo- p-dioxin (TCDD). Drug Metab. Rev. 13:387-406. 49 ------- Kuehl DW, Cook PM, Batterman AR. 1986. Update and depuration studies of PCDDs and PCDFs in fresh water fish. Chemosphere 15:2023-2026. Kunita N, Kashimoto T, Miyata H, Fukushima S, Hori S, Organa H. Causal agents of Yusho. Am. J. Ind. Med. 5:45-58. 1984. Lamb JL, et al. 1986. Effect of thyroid hormones on the induction of cleft palate by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in C57BL/6N mice. Toxicol. Appl. Pharmacol. 84:115-124. Leece B, Denomme MA, Towner R, Li SMA, Safe S. 1985. Polychlorinated biphenyls: correlation between in vivo and in vitro quantitative structure-activity relationships (QSARs). J. Toxicol. Environ. Health. 16:379-388. Lustenhouwer JWA, 01ie K, Hutzinger 0. 1980. Chlorinated dibenzo-p- dioxins and related compounds in incinerator effluents. Chemosphere 9:501-522. Luster MI, Hong LI, Osborne R, Blank JA, Clark G, Silver MT, Boorman GA, Greenlee WF. 1986. l-Amino-3,7,8-trichlorodibenzo-p-dioxin: a specific antagonist for TCDD-induced myelogoxicity. Biochem. Biophys. Res. Commun. 139:747-756. Marklund S, Rappe C, Tysklind M, Edgeback KE. 1987. Identification of polychlorinated dibenzofurans and dioxins in exhausts from cars run on leaded gasolines. Chemosphere 16:29-36. Mason G, Farrell K, Keys B, Piskorska-Pliszczynska J, Safe L, Safe S. 1986. Polychlorinated dibenzo-p-dioxins: quantitative in vitro and in vivo structure-activity relationships. Toxicol. 41:21-31. Mason G, Sawyer T, Keys B, Bandiera S, Romkes M, Piskorska-Pliszczynska J, Zmudzka B, Safe S. 1985. Polychlorinated dibenzofurans (PCDFs): correlation between in vivo and in vitro structure-activity relationships. Toxicol. 37:1-12. Mason G, Zacharewski T, Denomme MA, Safe L, Safe S. 1987. Polybrominated dibenzo-p-dioxins and related compounds: quantitative in vivo and in vitro structure-activity relationships. Toxicol. 44:245-255. McConnell EE, Moore JA, Haseman JK, Harris MW. 1978. The comparative toxicity of chlorinated dibenzo-p-dioxins in mice and guinea pigs. Toxicol. Appl. Pharmacol. 44:335-356. McKinney J, McConnell EE. 1981. Chlorinated dioxin and related compounds. (0. Hutzinger, R.W. Frei, E. Merian, and F. Pocciara, eds). New York: Pergamon Press. 50 ------- Moore JA, McConnell EE, Dalgard DW, Harris MW. 1979. Comparative toxicity of three halogenated dibenzofurans in guinea pigs, mice, and rhesus monkeys. Ann. N.Y. Acad. Sci. 320:151-163. NATO/CCMS. 1988. North Atlantic Treaty Organization/Committee on the Challenges of Modern Society. International toxicity equivalency factor method of risk assessment for complex mixtures of dioxins and related compounds. Report Number 176. NTP. 1980. National Toxicology Program. Carcinogenesis bioassay of 1,2,3,6,7,8- and 1,2,3,7,8,9-hexachlorodibenzo-p-dioxin for possible carcinogenicity (gavage study). DHHS Publication No. (NIH) 80-1754. Available from: National Technical Information Service, Springfield, VA. Nagarkatti P, Sweeney GO, Gauldie J, Clark DA. 1984. Sensitivity to suppression of cytotoxic T cell generation by 2,3,7,8-tetrachlorodibenzo- p-dioxin (TCDD) is dependent on the Ah genotype of the murine host. Toxicol. Appl. Pharmacol. 72:169-176. Nagayama J, Kiyohara C, Masuda Y, Kuratsune M. 1985. Genetically mediated induction of aryl hydrocarbon hydroxylase in human lymphoblastoid cells by polychlorinated dibenzofuran isomers and 2,3,7,8- tetrachlorodibenzo-p-dioxin. Arch. Toxicol, 56:230-235. Nilsson CA, Norstrom A, Anderson K, Rappe C. 1978. Impurities in commercial products related to PCP. In: Pentachlorophenol. Environ. Sci. Res. Series, 12 {K.R. Rao, ed.). New York: Plenum Press, pp. 313-323. OME. 1985. Ontario Ministry of the Environment. Scientific criteria document for standard development. Polychlorinated dibenzo-p- dioxins (PCDDs) and polychlorinated dibenzofurans (PCDFs). Toronto, Canada: No. 4-84. Ministry of the Environment. Okey AB, Duke AQ, Vella LM. 1984. Binding of benzo[a]pyrene and dibenzo[a,h]arithracene to the Ah receptor in mouse and rat hepatic cytosols. Cancer Res. 44:1426. Okey AB, Mason ME, Vella LM. 1983. The Ah receptor: species and tissue variation in binding of 2,3,7,8-tetrachlorodibenzo-p-dioxin and carcinogenic aromatic hydrocarbons. In: Extrahepatic Drug Metabolism and Chemical Carcinogenesis (J. Rydstrom, M. and M. Bengtsson, eds.), Elsevier, Amsterdam, pp. 389-393. Olie K, Vermeulen PL, Hutzinger 0. 1977. Chlorodibenzo-p-dioxins and chlorodibenzofurans are trace components of fly ash and flue gas of some municipal incinerators in the Netherlands. Chemosphere 6:455-459. 51 ------- Ono M, Wakimoto T, Tatsukawa R, Masuda Y. 1986. Polychlorinated dibenzo-p-dioxins and dibenzofurans in human adipose tissues of Japan. Chemosphere 15:1629-1634. Ontario Government. 1982. Chlorinated dioxins and chlorinated furans. Ambient air guideline. Health Studies Service. Ministry of Labor. Pazdernik T, Rozman K. 1985. Effect of thyroidectomy and thyroxine on 2,3,7,8-TCDD-tetrachlorodibenzo-p-dioxin-induced immunotoxicity. Life Sci. 36:695-703. Pluess N, Poiger H, Hohbach C, Suter M, Schlatter C. 1988a. toxicity of 2,3,7,8-pentachlorodibenzofuran (PeCDF) in rats. 17:1099-1110. Subchronic Chemosphere Pluess N, Poiger H, Hohbach C, Suter M, Schlatter C. 1988b. Subchronic toxicity of some chlorinated dibenzofurans (PCDFs) and a mixture of PCDFs and chlorinated dibenzodioxins (PCDDs) in rats. Chemosphere 17:937-984. Pohjanvirta R, Tuomisto J, Vartiainan T, Roxman K. 1987. Han/Wister rats are exceptionally resistant to TCOD.I. Pharacol. Toxicol. 60:145-150. Pohjanvitra R, et al. 1988. Hepatic Ah-receptor levels and the effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCOD) on hepatic monooxygenase activities in a TCDD-susceptible and -resistant rat strain. Toxicol. App. Pharmacol. 92:131-140. Poland A, Glover E, Kende AS. 1976. Stereospecific, high affinity binding of 2,3,7,8-tetrachlorodibenzo-p-dioxin by hepatic cytosol. J. Biol. Chem. 251:4936-4946. Poland A, Glover E. 1973. Chlorinated dibenzo-p-dioxins: potent inducers of d-aminolevulinic acid synthetase and aryl hydrocarbon hydroxylase. Mol. Pharmacol. 9:736-747. Poland A, Glover E. 1974. Comparison of 2,3,7,8-tetrachlorodibenzo- p-dioxin, a potent inducer of aryl hydrocarbon hydroxylase, with 3-methyl-cholanthrene. Mol. Pharmacol. 10:349-359. Poland A, Glover E. 1975. Genetic expression of aryl hydrocarbon hydroxylase by 2,3,7,8-tetrachlorodibenzo-p-dioxin: Evidence for a receptor mutation in genetically nonresponsive mice. Mol. Pharmacol. 11:389-398. Poland A, Glover E. 1980. 2,3,7,8-Tetrachlorodibenzo-p-dioxin: Segregation of toxicity with the Ah locus. Mol. Pharmacol. 17:86-94. Poland A, Greenlee WF, Kende AS. 1979. Studies on the mechanism of action of the chlorinated dibenzo-p-dioxins and related compounds. Ann. N.Y. Acad. Sci. 320:214-230. 52 ------- Poland A, Knutson J, Glover E, Kende A. 1983. Tumor promotion in the skin of hairless mice by halogenated aromatic hydrocarbons. In: Genes and proteins in oncogenesis (I.B. Weinstein, H.J. Vogel, eds.). New York: Academic Press, Inc., pp. 143-161. Poland A, Knutson JC, Glover E. 1984. Histologic changes produced by 2,3,7,8-tetrachlorodibenzo-p-dioxin in the skin of mice carrying mutations that affect integument. J. Invest. Dermatol. 83:454-459. Poland A, Knutson JC. 1982. 2,3,7,8-Tetrachlorodibenzo-p-dioxin and related halogenated aromatic hydrocarbons: examination of the mechanism of toxicity. Annu. Rev. Pharmacol. Toxicol. 22:517-554. Poland A, Palen 0, Glover E. 1982. Tumor promotion by TCDD in skin of HRS/J hairless mice. Nature (London) 300:271-273. Poland AP, Glover E, Robinson JR, Nebert DW. 1974. Genetic expression of aryl hydrocarbon hydroxylase activity. Induction of monooxygenase activities and cytochrome Pj-450 formation by 2,3,7,8-tetrachloro- dibenzo-p-dioxln in mice genetically "nonresponsive" to other aromatic hydrocarbons. J. Biol. Chem. 245:5599-5606. Puhvel SM, Sakamoto M. 1987. Response of murine epidermal keratinocyte cultures to 2,3,7,8-ttrachlorodibenzo-p-dioxin (TCDD): Comparison of haired and hairless genotypes. Toxicol. Appl. Pharmacol. 89:29-36. Rappe C, Buser HR, Bosshardt HP. 1979. Dioxins, dibenzofurans and other polyhalogenated aromatics: production, use, formation, and destruction. Ann. N.Y. Acad. Sci. 320:1-18. Rappe C, Buser HR, Stalling DL, Smith LM, Dougherty RC. 1981. Identification of polychlorinated dibenzofurans in environmental samples. Nature (London) 292:524-526. Rappe C, Gara S, Buser HR. chlorophenol formulations. 1978. Identification of PCDFs in commercial Chemosphere 7:981-991. Rappe C, Marklund S, Bergquist PA, Hansson M. 1983. PCDDs, PCDFs and other polynuclear aromatics formed during incineration and PCB fires. In: Chlorinated dioxins and dibenzofurans in the total environment (G. Choudhry et al. eds.). Boston: Butterworth Publications, pp. 99-124. Rappe E, Anderson R, Bergquist PA, Brohede C, Hansson M, Keller LO, Lindstrom G, Marklund S, Nygren M, Swanson SE, Tysklind M, Wiberr K. 1987. Overview on environmental fate of chlorinated dioxins and dibenzofurans: sources, levels and isoraeric patterns in various matrices, Chemosphere 16:1603-1618. 53 ------- Rozman K, et al. 1984. Effect of thyroidectomy and thyroxine on 2,3,7,8- tetrachlorodibenzo-p-dioxin (TCDD)-induced toxicity. Appl. Pharmacol. 72:372-376. Rozman K, et al. 1985a. Thyroid hormones modulate the toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin. J. Toxicol. Environ. Health 16:481-491. Ryan JJ, Lizotte R, Lau BP-Y. 1985. Chlorinated dibenzo-p-dioxins and chlorinated dibenzofurans in Canadian human adipose tissue. Chemosphere 14:697-706. Ryan JJ. 1986. Variation of dioxins and furans in human tissues. Chemosphere 15:1585-1593. Safe S, Mason G, Parrel 1 K, Keys B, Piskorska-Pliszczynska J, Madge JA, Chittim B. 1987. Validation of in vitro bioassays for 2,3,7,8-TCDD equivalents. Proceedings, Dioxin '86 Symposium, Fukuoka, Japan. Chemosphere 16:1723-1728. Safe S. 1980. Metabolism, uptake, storage and bioaccumulation of halogenated aromatic pollutants. In: Halogenated biphenyls, naphthalenes, dibenzodioxins and related products (R.D. Kimbrough, ed.), Elsevier, Amsterdam, pp. 81-107. Safe S. 1986. Comparative toxicology and mechanism of action of polychlorinated dibenzo-p-dioxins and dibenzofurans. Annu. Rev. Pharmacol. Toxicol. 26:371-399. Safe S. 1987. Determination of the 2,3,7,8-TCDD toxic equivalent factors: support for the use of the in vitro AHH induction assay. Chemosphere 16:791-802. Safe SH. 1988. The aryl hydrocarbon receptor. Pharmacology 2:78-83. ISI Atlas of Science: Schafer W, Ballschmiter K. 1986. Monobromo-polychloro-derivatives of benzene, biphenyl dibenzofuran and dioxin in chemical waste burning. Chemosphere 15:755-763. Schwetz BA, Norris JM, Sparschu GL. 1973. Toxicology of chlorinated dibenzo-p-dioxins. Environ. Health Perspect. 5:87-99. Shu HP, Paustenbach DJ, Murray FJ. 1987. A critical evaluation of the use of mutagenesis, carcinogenesis, and tumor promotion data in a cancer risk assessment of 2,3,7,8-tetrachlorodibenzo-p-dioxin. Regul. Toxicol. Pharmacol. 57-88. 54 ------- Stalling DL, Norstrom RJ, Smith LM, Simon M. 1985. Pattern of PCDD, PCDF, and PCB contamination in Great Lakes fish and birds and their characterization by principal component analysis. Chemosphere 14:627-643. Thoma H, Rist S, Hausschulz G, Hutzinger 0. 1986. Polybrominated dibenzodioxins and furans from the pyrolysis of some flame retardants. Chemosphere 15:649-652. USEPA. 1985. U. S. Environmental Protection Agency. Health assessment document for polychlorinated dibenzo-p-dioxins. EPA-600/8-84-014F. NTIS No. PB86-122546/AS. Van den Berg M, 01ie K, Hutzinger 0. (PCDFs): environnmetal occurrences, properties. Toxicol. Environ. Chem. 1985. Polychlorinated dibenzofurans physical, chemical and biological 9:171-217. Van den Berg M, de Broom E, van Grrvenbroek M, 01 ie K, Hutzinger 0. 1985. Bioavailab-ility of PCDDs and PCDFs absorbed on fly ash in rat, guinea pig, and Syrian golden hamster. Chemosphere 14:865-869. Van der Heijden CA, Knaap AGAC, Kramers PGN, Van Logten MJ. 1982. Evaluation of the carcinogenicity and mutagenicity of 2,3,7,8-tetra- chlorodibenzo-l,4-dioxin (TCDD): classification and no-effect level. Bilthoven, The Netherlands: State Institute of National Health. Report DOC/LCM 300/292. Vecchi A, Mantovani A, Sironi M, Luini W, Spreafico F, Garattini S. 1980. The effect of acute administration of 2,3,7,8-tetrachlorodibenzo-p- dioxin (TCDD) or humoral antibody production and cell-mediated activities in mice. Arch. Toxicol. 4:163-165. Vecchi A, Sironi M, Canegrati MA, Recchia M, Garattini S. 1983. Immunosuppressive effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin in strains of mice with different susceptibility to induction of aryl hydrocarbon hydroxylase. Toxicol. Appl. Pharmacol. 68:434-441. Vos JG, Koeman JH, Van der Mass HL, Ten Noever de Brauw MC, and Oe Vos RH. 1970. Identification and toxicological evaluation of chlorinated dibenzofuran and chlorinated naphthalene in two commercial polychlorinated biphenyls. Food Cosmet. Toxicol. 8:625-633. Walden R, Schiller CM. 1985. Comparitive toxicity of 2,3,7,8-tetra- chlorodibenzo-p-dioxin (TCDD) in four (sub)strains of adult male rat. Toxicol. Appl. Pharmacol. 77:490-495. Whitlock JP. 1986. The regulation of cytochrome P-450 gene expression. Annu. Rev. Pharmacol. Toxicol. 26:333-369. Whitlock JP. 1987. The regulation of gene expression by 2,3,7,8- tetrachlorodibenzo-p-dioxin. Pharmacol. Rev. 29:147-161. 55 ------- Zacharewski T, Harris M, Safe S, Thoma H, Hutzinger 0. 1988. Applications of the in vitro aryl hydrocarbon hydroxylase induction assay for determining "2,3,7,8-TCDD equivalents": pyrolyzed brominated flame retardants. Toxicology 51:177-189. 56 ------- |