TOXICOLOGICAL SCIENCES 91(1), 275–285 (2006) doi:10.1093/toxsci/kfj125 Advance Access publication February 2, 2006 Recommended Relative Potency Factors for 2,3,4,7,8-Pentachlorodibenzofuran: The Impact of Different Dose Metrics Robert A. Budinsky,*,1 Dennis Paustenbach,† Donald Fontaine,* Bryce Landenberger,* and Thomas B. Starr‡ *The Dow Chemical Company, Midland, Michigan 48674; †ChemRisk, Inc., San Francisco, California 94105; and ‡TBS Associates, Raleigh North Carolina 27615–3700 Received April 6, 2005; accepted January 18, 2006 The recent National Toxicology Program (NTP) cancer bioassays for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and 2,3,4,7,8-pentachlorodibenzofuran (4-PeCDF) permit a reevaluation of the current TEF value of 4-PeCDF. The data also allow for the derivation of relative potency factors (RPFs) for cancer, which are based not only on administered dose but also on potentially more informative dose metrics, such as liver concentration, area under the liver concentration curve, and lifetime average body burden. Our analyses of these data indicate that chi-squared tests of observed versus predicted liver tumor incidence for 4-PeCDF reject the current TEF value of 0.5 value as too high. 4-PeCDF RPFs were derived using estimation methods that either did or did not assume parallelism of the 4-PeCDF and TCDD dose–response curves. The resulting parallelism-based RPFs for administered dose, liver concentration at terminal sacrifice, liver concentration AUC, and lifetime average body burden are 0.26, 0.014, 0.021, and 0.036, respectively. The administered dose RPF estimate is approximately one-half the current TEF value of 0.5. However, the use of administered dose fails to take into account pharmacokinetic differences between congeners and the generally acknowledged belief that body burden or some other measure of cumulative dose is more appropriate for estimating the health risk posed by persistent chemicals. The other three dose metrics do account for these important factors, and the corresponding RPFs are at least 10-fold lower than the current TEF for 4-PeCDF. In summary, our analyses support an administered dose TEF no greater than 0.25 and one in the 0.05–0.1 range for internal dose metrics such as lifetime average liver concentration or body burden. INTRODUCTION 2,3,4,7,8-Pentachlorodibenzofuran (4-PeCDF) is an important dioxin-like compound for a number of reasons. It has a relatively high toxic equivalency factor (TEF) of 0.5 (Van den Berg et al., 1998), and it contributes substantially to the toxic 1 To whom correspondence should be addressed at The Dow Chemical Company, 1803 Building, Midland, MI 48674; Fax: 989-638-9863. E-mail: [email protected]. equivalents (TEQ) found in many environmental and biomonitoring samples (Kreuzer et al., 1997; NHANES, 2005; Patterson et al., 1994; Petreas et al., 2000; USEPA 2003a; Wittsiepe et al., 2000, 2004). It also exhibits a high degree of dose-dependent liver sequestration because of its ability to induce as well as bind to CYP1A2 (Brewster and Birnbaum, 1987; NTP, 2003a; Van den Berg et al., 1989), and it is one of the few dioxins for which 2-year carcinogenicity bioassay data are available (NTP, 2003a). Because of the significant role that 4-PeCDF can play in environmental remediation initiatives, an accurate estimate of its carcinogenic potency relative to that of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is of considerable value. Toxic equivalency factors for dioxin-like compounds were first introduced in the late 1980s. Pluss et al. (1988) recommended a potency of 0.4 for 4-PeCDF compared to TCDD based on clinical findings in a 13-week dietary rat study. Relative potency factors (RPFs) ranging from 0.05 to 0.5 (in vivo endpoints) and 0.7 to 0.9 (in vitro endpoints) were suggested for 4-PeCDF by Olson et al. (1989). Safe’s (1998) summary of 4-PeCDF RPFs from a number of earlier in vivo and in vitro toxicity studies ranged from 0.12 to 0.8 and 0.11 to 0.67, respectively, based on acute lethality, body weight loss, thymic atrophy, immunotoxicity, and teratogenicity endpoints. The current WHO TEF value of 0.5 is based on either 73 (Finley et al., 2003) or 60 (USEPA, 2003a) RPFs, all for non-cancer endpoints. The U.S. Environmental Protection Agency (EPA) Draft Dioxin Reassessment identified enzyme induction, organ and body weight changes, immune changes, developmental toxicity, thyroid hormone changes, hepatic retinoid changes, hepatic porphyria, and tumor promotion dose–response data sets as supporting a TEF of 0.5 for 4-PeCDF (USEPA, 2003a). Before completion of the recent 4-PeCDF cancer bioassay (NTP, 2003a), the Waern et al. (1991) initiation-promotion study came the closest to providing data that are directly relevant to cancer risk assessment. Waern et al. (1991) estimated RPFs of 0.1, based on administered dose, and 0.007, based on liver concentration. In contrast, the Harper et al. (1993) plaqueforming immunotoxicity study yielded a much higher range, The Author 2006. Published by Oxford University Press on behalf of the Society of Toxicology. All rights reserved. For Permissions, please email: [email protected] 276 BUDINSKY ET AL. with RPFs varying from 0.58 to 4.0. Finley et al. (2003) evaluated the World Health Organization (WHO) RPF database with a weighting scheme based on relevance to human cancer risk assessment. They obtained a weighted mean TEF for 4-PeCDF of 0.27. Haws et al. (2005) have recently updated this database. The minimum RPF estimate from in vivo data was 0.0065, with a maximum of 3.7 and a 50th percentile RPF estimate of 0.2. Based on the 101 available in vitro and in vivo RPFs, Haws and co-workers derived an average RPF value of 0.22. All of these estimates are for application with administered doses. The National Toxicology Program (NTP) recently reported findings from its cancer bioassays of TCDD, 4-PeCDF, PCB126, and a 1:1:1 mixture (based on current WHO TEF values) of all three compounds (first introduced by van Birgelen et al., 1997; NTP, 2003a, 2003b, 2003c, and 2003d). The TCDD and the TEF-adjusted doses for 4-PeCDF and PCB126 were selected using tumor incidence rates from the original TCDD bioassay conducted by Kociba et al. (1978). These NTP studies provide the first opportunity (1) to evaluate the hypotheses of parallel dose responses and dose additivity upon which the validity of the TEF concept relies and (2) to derive RPF sampling distributions for 4-PeCDF based on carcinogenicity. The liver and adipose tissue concentration data that were collected for TCDD and 4-PeCDF during the course of these bioassays also allow for assessments of relative carcinogenic potency based on dose metrics other than administered dose. For example, an RPF appropriate for use with lifetime average body burden would be consistent with the EPA dose–response modeling for cancer and non-cancer effects and for extrapolation of estimated risks from rodents to humans. The parallel dose–response and dose-additivity assumptions underlying TEFs have recently been evaluated from data gathered during the NTP bioassays (Toyoshiba et al., 2004; Walker et al., 2005). Toyoshiba et al. (2004) used Hill model dose–response relationships to assess the parallelism of the congener-specific and mixture dose–response curves, dose additivity, as well as to derive RPF estimates for 4-PeCDF and PCB126 from data for CYP1A1 and CYP1A2 activity. Interestingly, the assumption of parallelism was unilaterally rejected for CYP1A1 and CYP1A2 at all measured time points (14, 31, and 53 weeks on study). The hypothesis of dose additivity was also rejected except for CYP1A1 at 14 weeks and possibly for CYP1A2 at 53 weeks ( p ¼ 0.065). Relative potency factor estimates for 4-PeCDF from the full (unconstrained) dose–response models for CYP1A1 ranged from 0.04 to 0.19, whereas those for CYP1A2 ranged from 0.07 to 0.25. Walker et al. (2005) investigated parallelism, dose additivity, and the adequacy of the WHO TEF value of 0.5 as an RPF estimate for 4-PeCDF using the carcinogenicity data from the NTP bioassays. There was no significant evidence of nonparallelism, but significant departures from dose additivity (presuming parallel dose responses) were obtained for two of the four elevated tumor types: cystic keratinizing epithelioma (CKE) of the lung ( p ¼ 0.033) and gingival squamous cell carcinoma ( p ¼ 0.047). It was noted that the statistical power to detect significant departures from these fundamental assumptions was low, with the quantal tumor data ranging across the four tumor endpoints from 0.1 to 0.50. Using similar Hill model analyses of the bioassay tumor data, Walker et al. (2005) also estimated lower RPFs for 4-PeCDF than the current TEF value of 0.5. For cholangiocarcinoma and hepatocellular adenoma (HA) of the liver, CKE of the lung, and gingival squamous cell carcinoma, their ‘‘optimal’’ potency estimates for 4-PeCDF were 0.16, 0.34, 0.30, and 0.26, respectively. Although only the RPFs for cholangiocarcinoma of the liver and CKE of the lung were significantly smaller than the WHO TEF of 0.5 (p < 0.0001), the authors called attention to the consistency between their lower potency estimates and those reported by Waern et al. (1991). Further, they suggested that ‘‘the current TEF value for 4-PeCDF ought to be reevaluated for its application in quantitative cancer risk assessments.’’ Other preliminary RPF estimates for 4-PeCDF derived specifically for internal dose metrics have been reported (Budinsky et al., 2004; Fontaine et al., 2004). These estimates, appropriate for use with total body burden and liver concentration, are an order of magnitude lower than either the current WHO TEF or the administered dose RPF estimates reported by Walker et al. (2005) for 4-PeCDF. In the present article we expand on the preliminary RPF estimates reported by Budinsky et al. (2004) and Fontaine et al. (2004). Making use of a simple statistical goodness-of-fit test, we first determine whether or not the current TEF value of 0.5 accurately predicts liver and lung tumor responses to 4-PeCDF exposure using the observed dose responses for these endpoints in the TCDD bioassay. Then our analyses generate separate and distinct RPFs appropriate for use with the following dose metrics: daily administered dose per unit of body weight, time-averaged body burden, liver concentration, or area under the liver concentration curve (AUC), with the latter two dose metrics determined at terminal sacrifice. We also derive empirical sampling distributions for the estimated RPFs using Monte Carlo simulation. MATERIALS AND METHODS Tumor incidence. Tumor incidence data from the NTP bioassay studies of TCDD and 4-PeCDF (NTP, 2003a and 2003b) were used in dose–response modeling and in evaluating the adequacy of 0.5 as a RPF in predicting the observed 4-PeCDF tumor response from the modeled TCDD tumor response. The data were also used to develop the sampling distributions of 4-PeCDF RPFs appropriate for use with different internal dose metrics. The Weibull dose–response model (with independent background) was fit to the TCDD data for liver tumors or combined liver or lung tumors as a function of (1) administered dose (ng/kg per day),(2) liver concentration at terminal sacrifice (pg/gm), (3) area under the liver concentration curve at terminal sacrifice (AUC, pg/g 3 weeks), or (4) lifetime time average body concentration (pg/g) using the EPA Benchmark Dose software (version 1.3.2) (USEPA, 2001). 277 RELATIVE POTENCY FACTORS FOR 4-PeCDF Animals with multiple tumors were counted only once in incidence numerators, and incidence denominators were adjusted for the competing risks of intercurrent mortality using the poly-3 procedure (Bailer and Portier, 1988). For purposes of this analysis, liver tumors included both hepatocellular adenomas and cholangiolar carcinomas; lung tumors were predominantly cystic keratinizing epitheliomas, with occasional alveolar or bronchiolar adenomas. We chose to analyze combined liver or lung tumors in addition to liver tumors alone because the USEPA had used this combination in developing its original carcinogenic potency factor for TCDD of 156,000 (mg/kg/day)1 (USEPA, 1988). Dose metrics. A summary of the specific dose metric values that were utilized in the dose-response modeling is provided in Table 1. Liver concentrations of TCDD and 4-PeCDF at terminal sacrifice were taken directly from Tables 13 (TR 521) and 8 (TR 525) in the Board Draft NTP Technical Reports for these substances (NTP, 2003a, 2003b). Liver AUC at the terminal sacrifice was generated from the liver concentrations reported at weeks 14, 31, 53, and 104 using simple linear interpolation, the trapezoidal rule, and the assumption that the liver concentration at study onset, and other times it was reported to be below the limit of quantification (LOQ), equaled 1/2 LOQ. The presumed baseline AUC value of 104 * (1/2 LOQ) was then subtracted from each treatment group AUC at terminal sacrifice. To illustrate, in the 3 ng/kg/day TCDD dose group: Liver AUC @ 104 weeks ¼ 1=2½ð14 0Þ * ð1=2 * 50 þ 676:2Þ þ ð31 14Þ * ð676:2 þ 598:9Þ þ ð53 31Þ * ð598:90 þ 499:38Þ þ ð104 53Þ * ð499:38 þ 680:71Þ 104 *ð1=2 * 50Þ ¼ 55320:13 ðpg=gÞ * weeks: To obtain lifetime average body concentrations, weekly body concentrations during the course of the bioassays were first estimated with a simple pharmacokinetic model (Edmund Crouch, Cambridge Environmental, personal communication). Briefly, TCDD or 4-PeCDF body burden (the product of body concentration Cb and body weight, w) was assumed to reside entirely in liver and adipose tissue and have a time evolution governed by the differential equation: dðCb * wÞ=dt ¼ ðg k * f * Cb Þ * w; ð1Þ where g is the product of the administered dose and an oral gavage absorption fraction (assumed equal to 0.9), k is a constant metabolic elimination rate, and f is the fraction of the total body burden present in liver. Following Carrier et al. (1995), the liver fraction f was presumed to have a minimum value of 0.01 and Michaelis-Menten dependence upon body concentration: f ¼ 0:01 þ ðfmax 0:01Þ * Cb =ðCb þ Km Þ: ð2Þ To obtain estimates of the parameters fmax and Km in Equation 2, mean adipose and liver tissue concentrations (Ca and Cl) reported for the various dose groups at 14, 31, and 53 weeks were first combined with corresponding mean body weights, mean liver weights, and estimated adipose tissue weights (w, wl, and wa) to generate estimates of the body concentration Cb and liver fraction f at each of the above-noted time points for each nonzero dose group: Cb ¼ ðCa * wa þ Cl * wl Þ=w; ð3Þ f ¼ ðCl * wl Þ=ðCb * wÞ: ð4Þ Adipose tissue weight (wa) was not measured during the bioassays, so it was estimated from total body weight using a relationship reported by Brown et al. (1997): wa =w ¼ ð1:664 þ 0:0199 * wÞ=100: ð5Þ The parameters fmax and Km in Equation 2 were then optimized by minimizing the sum of squared differences between the right hand sides of Equations 2 and 4. To obtain an estimate of the metabolic elimination parameter k, Equation 1 was integrated numerically using a 1-week time step, reported body weights (linearly interpolated as necessary to obtain weekly values), and interatively estimated values of k: ðCb * wÞtþ1 ¼ g * wt þ ðCb * wÞt * ð1 k * ft Þ: ð6Þ The predicted body burdens were then divided by body weight to provide predicted body concentrations for comparison with those given by Equation 3 using the data reported at 14, 31, and 53 weeks. Assuming that the prediction errors were log-normally distributed, the parameter k was optimized by minimizing the sum of squared differences between the logarithms of the body concentrations predicted by Equation 6 and those given by Equation 3. With k optimized, the predicted TCDD and 4-PeCDF body concentrations were averaged over 104 weeks to obtain lifetime average body concentrations suitable for use in the dose–response modeling. Relative potency hypothesis tests, RPF estimates, and RPF sampling distributions. The predicted numbers of tumor-bearing animals for each 4-PeCDF bioassay dose group were generated by (1) applying the current TEF of 0.5 to the 4-PeCDF dose metrics to obtain TCDD-equivalent dose metrics; (2) using the fitted dose–response model for TCDD to estimate the probability of being tumor-bearing at each 4-PeCDF dose; and (3) multiplying each such probability by the poly-3 adjusted number of animals at risk in the corresponding 4-PeCDF dose group. Under the null hypothesis that the true relative potency (RPF) for 4-PeCDF equals 0.5, the sum of [(Observed – Predicted)2/ Predicted] over the different dose groups should be approximately distributed as a chi-squared random variate with degrees of freedom equal to the number of included dose groups. Two different methods were used to estimate RPFs and their sampling distributions from the 4-PeCDF and TCDD tumor data. The first method employed separate, possibly nonparallel Weibull model fits to each of the TCDD and 4-PeCDF tumor data sets. One thousand synthetic data sets for each compound were generated using the observed poly-3-adjusted numbers of TABLE 1 Dose Metrics for TCDD and 4-PeCDF Used in Dose–Response Modeling and Relative Potency Factor (RPF) Estimation Administered dose 5 days/week ng/kg/day TCDD 0 3 10 22 46 100 Liver concentration at terminal sacrifice pg/g Liver AUC at terminal sacrifice (pg/g) * weeks Body concentration lifetime average pg/g 4-PeCDF TCDD 4-PeCDF TCDD 4-PeCDF TCDD 4-PeCDF 0 6 20 44 92 200 25 681 2,213 4,364 6,413 9,325 1,615 17,615 62,457 125,110 263,557 499,565 8,176 55,320 203,100 434,792 786,705 1,507,044 36,401 1,339,162 4,589,442 10,136,372 21,445,272 41,065,441 0 44 123 255 518 1,110 0 491 1,610 3,570 7,410 16,300 278 BUDINSKY ET AL. animals at risk and the observed binomial probabilities of response at each dose. Weibull model parameters specific to each simulated data set were then obtained with the EPA Benchmark Dose software (USEPA, 2001). Then TCDD and 4-PeCDF doses were determined that gave extra risks of 5%, 10%, 25%, 50%, 75%, 90%, and 95% (referred to as EDx). Empirical sampling distributions for each RPFx (defined as the ratio of the TCDD EDx to the corresponding 4-PeCDF EDx) were obtained either by direct calculation of all 1 million possible ratios or from a randomly drawn subset of 1000 ratios. The second estimation method employed fitting the TCDD and 4-PeCDF tumor incidence data simultaneously, assuming parallel dose responses. A single Weibull model was fit to both data sets by maximum likelihood with the RPF for 4-PeCDF as an additional adjustable parameter (estimation routines kindly provided by Edmund Crouch, Cambridge Environmental). As with the first method, 1000 synthetic data sets for TCDD and 4-PeCDF tumor incidence were generated using the poly-3 adjusted numbers of animals at risk and the experimental binomial probabilities of response at each dose. The simultaneous estimation procedure was then implemented with each simulated data set in combination with each dose metric. Finally, the empirical sampling distributions for the 4-PeCDF RPF were constructed from the results, and their means, variances, and other characteristics were determined. TABLE 2 Observed and Predicted Numbers of Liver Tumor-Bearing Animals in the 4-PeCDF Bioassay Using Independent Weibull Models Fit to the TCDD Liver Tumor Data versus Administered Dose (AD), Liver Concentration at Terminal Sacrifice (Cl), Area under the Liver Concentration Curve at Terminal Sacrifice (AUCl), or Lifetime Average Body Concentration (Cb), with an assumed RPF of 0.5 for 4-PeCDF 4-PeCDF No. dose observed/ group no. at riska 0 6 20 44 92 200 Liver Tumor Incidence Data and Modeled Dose Responses Figure 1 displays the poly-3-adjusted liver tumor incidence for the TCDD and 4-PeCDF studies along with the independent Weibull model fits to each data set. There is only a relatively modest increase in liver tumor incidence (~16%) for 4-PeCDF even at the highest dose tested, and this is consistent with NTP’s characterization of the 4-PeCDF findings as providing only ‘‘some evidence of carcinogenicity’’ versus the ‘‘clear evidence of carcinogenicity’’ in the TCDD bioassay. It is also readily apparent that 4-PeCDF is less than half as potent as TCDD on the administered dose scale. Tests of the Adequacy of 0.5 as an RPF for 4-PeCDF Tables 2 and 3 provide the results of chi-squared tests of the null hypothesis that an RPF of 0.5 for 4-PeCDF is consistent NTP Bioassay Liver Tumor Responses 0.7 # with Tumor / # at Risk AD Cl AUCl 0. 0.005 0.1 0.8 4.7 23.4 0.0001 16.4 36.7 37.9 36.1 37.2 0.00007 3.1 35.6 37.9 36.1 37.2 Cb 0. 0.8 12.9 36.7 36.1 37.2 129.2 (5) 103.0 (5) 143.6 (5) Chi-squared 20.9 (5) (df)b p value 0.00084 3.1 3 1029 3.5 3 1026 1.3 3 1020 RESULTS 0.6 1/42.0 0/38.1 1/36.7 1/37.9 3/36.1 6/37.2 Number predicted with liver tumors a Number at risk is the poly-3 adjusted number of animals at risk of developing liver tumors. b Because predicted numbers of tumor-bearing animals in the control group were either exactly zero or extremely small, only the five nonzero dose groups were included in the chi-squared computations. with liver or the combined liver and lung tumor incidences observed in the TCDD and 4-PeCDF bioassays. For each of these tumor endpoints, four tests of the null hypothesis were undertaken (one for each dose metric), and in every case the TABLE 3 Observed and Predicted Numbers of Liver or Lung Tumor-Bearing Animals in the 4-PeCDF Bioassay Using Independent Weibull Models Fit to the TCDD Liver or Lung Tumor Data versus Administered Dose (AD), Liver Concentration at Terminal Sacrifice (Cl), Area under the Liver Concentration Curve at Terminal Sacrifice (AUCl), or Lifetime Average Body Concentration (Cb), with an Assumed RPF of 0.5 for 4-PeCDF TCDD 0.5 4-PeCDF No. dose observed/ group no. at riska 0.4 0.3 0.2 0 6 20 44 92 200 4-PeCDF 0.1 0 0 50 100 150 AD 0.4 0.4 0.4 0.9 4.6 26.0 Cl 0.4 17.4 36.7 37.9 36.1 38.1 Chi-squared 15.2 (6) 143.2 (6) (df) p value 0.019 2.1 3 1028 200 Administered Dose (ng/kg/day) FIG. 1. Separate Weibull model fits of the TCDD and 4-PeCDF liver tumor incidence data. 1/42.0 0/38.1 1/36.7 1/37.9 3/36.1 8/38.1 Number predicted with liver or lung tumors a AUCl 0.4 3.0 36.3 37.9 36.1 38.1 128.5 (6) 2.7 3 1025 Cb 0.4 0.9 13.4 37.4 36.1 38.1 103.0 (6) 5.9 3 10–20 Number at risk is the poly-3 adjusted number of animals at risk of developing liver or lung tumors. RELATIVE POTENCY FACTORS FOR 4-PeCDF null hypothesis was rejected; i.e., there was no indication that an RPF of 0.5 was consistent with the tumor data. The test using liver tumor data and administered dose was highly significant (p ¼ 0.00084), with the remaining tests yielding even more highly significant differences and far smaller p values (p < 1020), indicating that once the prominent pharmacokinetic differences between TCDD and 4-PeCDF are accounted for with an appropriate internal dose metric (e.g., AUCl), the tumor incidence rates predicted for 4-PeCDF with an RPF of 0.5 are simply not compatible with the observed rates. Figure 2 displays the differences between observed and predicted numbers of liver tumor-bearing animals as a function of the four different 4-PeCDF dose metrics. The large discrepancies between observed and predicted incidence arise in the dose range where there are significant treatment-related effects for TCDD. In the lower dose range, where there are just a few cases of liver tumors, the differences between observed and predicted incidence are much smaller. For all dose metrics, use of a 0.5 RPF value led to overprediction of tumor incidence rates. 279 for administered dose, and 0.043 (0.015), 0.026 (0.013) and 0.022 (0.020), respectively, for lifetime average body concentration. The dependence of the RPF estimates on the response probabilities at which they are estimated is a characteristic that would not arise if the dose–response relationships for TCDDand 4-PeCDF-induced liver tumor incidence were parallel to one another; i.e., parallel dose responses necessarily give the same RPF value at all response probabilities. This incidence RPF Sampling Distributions at Selected Dose–Response Levels When separate Weibull models were fitted to simulated data sets for TCDD and 4-PeCDF, the estimated mean RPFs and their sampling distributions varied with tumor incidence rate as well as dose metric. Examples of the RPF distributions obtained using administered dose and lifetime average body concentration are shown Figure 3a and 3b, respectively. Three sampling distributions are provided in each frame of the figure, representing the RPFs obtained at predicted liver tumor incidence rates of 10%, 50%, and 90%. The incidence ratedependent distributions have estimated means (standard deviations) of 0.30 (0.12), 0.19 (0.10), and 0.16 (0.15), respectively, Assuming RPF = 0.5 40 # Observed - # Predicted Administered Dose 30 Body Concentration 20 Liver AUC Liver Concentration 10 0 -10 -20 -30 -40 -50 0 3 10 22 46 100 Dose, TEQ (ng/kg/day) FIG. 2. Observed – Predicted numbers of animals with liver tumors using four different dose metrics. FIG. 3. a. The RPFx sampling distributions at 10%, 50%, and 90% liver tumor incidence rates for 4-PeCDF based on administered dose. b. The RPFx sampling distributions at 10%, 50%, and 90% liver tumor incidence rates for 4-PeCDF based on lifetime average body concentration. 280 BUDINSKY ET AL. RPFx and RPFx/ Std Dev for Average Body Concentration 3.0 0.06 RPFx / Std Dev 0.05 Mean RPFx imately 0.036 (0.011) for the lifetime average body concentration dose-metric. 3.5 0.04 RPF Distributions Assuming Parallel Dose–Response Curves 2.5 2.0 RPFx 0.03 1.5 0.02 1.0 0.01 0.5 0.00 RPFx / Std Dev 0.07 0.0 0 0.25 0.5 0.75 1 Estimated Fraction with Liver Tumor FIG. 4. The estimated mean RPFx and mean RPFx divided by its estimated standard deviation (S.D.) plotted versus fraction with liver tumor. The relative standard deviation (S.D./RPFx) is minimized at an incidence rate of approximately 0.16. rate dependence also motivates consideration of whether there is an ‘‘optimal’’ tumor response rate at which the relative error in the estimated RPFs is minimized. In Figure 4, the mean RPF values obtained for the average body concentration dose metric are plotted as a function of response probability at the 5%, 10%, 25%, 50%, 75%, 90%, and 95% incidence points; the figure also displays each mean RPF estimate divided by its estimated standard deviation, a ‘‘figure of merit’’ that is useful in assessing the relative precision of the different responsedependent RPF estimates, with larger RPF/S.D. ratios implying smaller relative uncertainty. The response probability at which the estimated RPF yields the smallest relative error is approximately 16%, irrespective of which dose metric is considered (data not shown). As indicated in Table 4, this response rate corresponds to a mean RPF value (standard deviation) of approximately 0.26 (0.081) for administered dose and approx- Relative potency factor sampling distributions obtained assuming parallel dose responses for TCDD- and 4-PeCDFinduced liver tumors with each of four dose metrics are presented in Figure 5. Table 4 provides the corresponding mean RPF estimates and associated standard deviations, and Table 5 presents selected cumulative percentiles of these RPF sampling distributions for the four dose metrics that were considered. The mean RPF estimates for each dose metric are 0.27 (administered dose), 0.014 (liver concentration), 0.022 (liver AUC), and 0.036 (average body concentration). These values are very close to the corresponding RPF16 values derived without enforcing the constraint of parallel tumor responses (see Table 4). It is noteworthy that the standard deviations of these RPF estimates are about 30% smaller than those obtained without imposition of this constraint. Table 4 also presents the Weibull model parameter estimates obtained with the two estimation approaches. Of particular note is the fact the power parameter (a) for 4-PeCDF is substantially smaller than that for TCDD for all four dose metrics, indicating a considerably less nonlinear dose–response relationship than that observed for TCDD, and thus providing some evidence of non-parallel dose responses. The Weibull model parameters obtained with the joint approach assuming parallelism strongly resemble those obtained for TCDD alone. This may reflect the relative weakness of the liver tumor dose response observed in the 4-PeCDF bioassay. DISCUSSION When the 1997 WHO TEF evaluation was conducted, carcinogenicity data for 4-PeCDF (and most other dioxin-like TABLE 4 Weibull Modela Parameters from the Individual and Joint Fits of the TCDD and 4-PeCDF Liver Tumor TCDDb Data set Dose metric AD Cl AUCl Cb 4-PeCDFb Joint w/parallel dose responsec a b c a b c RPF16d S.D. a b c RPF S.D. 2.52 5.16 3.02 2.59 0.996 0.991 0.996 0.997 0. 0. 0. 0. 1.36 1.53 1.51 1.36 0.170 0.168 0.168 0.170 0.014 0.014 0.014 0.014 0.26 0.013 0.021 0.036 0.081 0.0034 0.0059 0.011 2.38 5.10 2.87 2.44 0.945 0.945 0.947 0.945 0.010 0.016 0.011 0.011 0.27 0.014 0.022 0.037 0.054 0.0018 0.0039 0.0073 Note. Incidence, together with relative potency estimates and estimated standard deviations obtained with both procedures. a P(d) ¼ c þ (1c)(1exp(b * da)). b Doses scaled so that the d ¼ 1 for the highest dose group. c Doses scaled so that the d ¼ 1 for the highest TCDD dose group. d RPF16 is the relative potency of 4-PeCDF evaluated at the liver tumor incidence rate giving the minimum relative error in potency, namely, 0.16. RELATIVE POTENCY FACTORS FOR 4-PeCDF potentially more congruent with probabilistic risk assessment needs; and (3) generation of empirical sampling distributions for these RPFs. We propose that the cancer-based RPFs and RPF distributions derived herein be used in conducting human cancer risk assessments whenever 4-PeCDF is a significant congener in the exposure mixture of concern. However, the WHO TEFs, which were currently being re-evaluated at the time this manuscript was prepared, may still prove useful for other exposure circumstances and matrices. RPF Sampling Distributions Probability Density, Arbitrary Units 50 45 Administered Dose 40 Liver AUC 35 30 25 Body Concentration Liver Concentration 20 15 10 Adequacy of an RPF of 0.5 for 4-PeCDF 5 0 0.001 0.01 0.1 1 RPF FIG. 5. The RPF sampling distributions obtained from simultaneous fits to the TCDD and 4-PeCDF liver tumor data assuming dose additivity (parallel dose–response curves). compounds) were not available. The new NTP carcinogenicity data permit development of RPFs and estimates of potential human risk specifically for the cancer endpoint. Relative potency factors and their distributions derived from these tumor data obviate the need to rely on the TEF of 4-PeCDF, which by definition provides only an order-of-magnitude estimate of potency based on data for a range of biological endpoints, most of which have limited relevance to the carcinogenic potential of 4-PeCDF. Data from the NTP TCDD and 4-PeCDF bioassays (NTP, 2003a, 2003b) permit (1) an objective assessment of whether or not an RPF of 0.5 (the current 4-PeCDF TEF value) provides an adequate description of the 4-PeCDF carcinogenicity data in relation to that obtained for TCDD; (2) derivation of RPFs and their distributions with and without the assumption of parallelism, and suitable for use with administered dose and other internal dose metrics TABLE 5 Cumulative Percentiles of the RPF Sampling Distributions for Different Dose Metrics Obtained with a Common Weibull Model for TCDD- and 4-PeCDF–Induced Liver Tumors, Assuming Parallel Dose Responses Cumulative percentile 1 5 10 25 50 75 90 95 99 281 Administered dose RPF Liver concentration RPF Liver AUC RPF Body concentration RPF 0.147 0.185 0.205 0.235 0.270 0.306 0.335 0.355 0.385 0.009 0.011 0.012 0.013 0.014 0.015 0.015 0.016 0.017 0.013 0.016 0.017 0.019 0.022 0.024 0.026 0.027 0.029 0.021 0.026 0.029 0.033 0.037 0.042 0.046 0.049 0.053 Our analyses show that an RPF of 0.5 does not adequately characterize the carcinogenic potency of 4-PeCDF relative to that of TCDD. Indeed, it significantly overstates this potency. There is also some evidence of non-parallelism of the liver tumor dose responses for TCDD and 4-PeCDF, in that the exponents of dose (the Weibull model parameter a) in the separately estimated models were quite different, with the dose response for TCDD-induced liver tumors being considerably steeper than that for 4-PeCDF. However, the low statistical power of the bioassay designs noted by Walker et al. (2005) precludes a rigorous test of this hypothesis. Despite the large number of dose groups employed in these studies, maximal tumor responses for 4-PeCDF and TCDD do not appear to have been achieved in any of the treated groups. Indeed, the 4-PeCDF study failed to reach a 20% liver tumor response rate, even at the highest dose tested (200 ng/kg per day). Although these data are strong enough to reveal the comparatively low carcinogenic potency of 4-PeCDF relative to that of TCDD, they fall short of providing a definitive characterization of the shape of the dose–response curve for 4PeCDF–induced liver tumors. It is noteworthy that the more sensitive assessments of parallelism by Toyoshiba et al. (2004) using liver enzyme activity levels determined during the course of these bioassays provide compelling evidence of the inappropriateness of this assumption. The practical need for RPFs in human cancer risk assessments thus motivates the consideration of additional approaches to identifying RPFs that do not depend on this questionable assumption. RPF Estimates Without the Assumption of Parallelism In our analyses, which did not assume parallelism, the RPF estimates depend on the response level at which they were estimated, implying that no single RPF value can completely characterize the potency of 4-PeCDF relative to that of TCDD. This dependence on response level, illustrated in Figure 3a and 3b, and Figure 4, appears monotone, decreasing with increasing probability of being tumor-bearing, and is steepest at the lower end of the dose–response curve where there is minimal information about dose response for either TCDD or 4-PeCDF. However, the uncertainty of this estimate, reflected by the spread in the RPF sampling distributions (depicted in Figure 3a and 3b), also increases as the probability of being 282 BUDINSKY ET AL. tumor bearing declines. The result of these two tendencies is to create an optimum response rate at which the relative standard error of the estimated RPF is minimized. As shown in Figure 4 for lifetime average body concentration Cb, this minimum occurred at a response probability of approximately 0.16 (the tumor incidence in the highest 4-PeCDF dose group), where the central RPF estimate is approximately 0.036. The highest tested 4-PeCDF dose thus provides the least uncertain estimate of a ‘‘point of departure’’ suitable for low-dose risk extrapolation. RPF Estimates Assuming Parallelism The central RPF estimates we obtained with the assumption of parallel dose–response curves were nearly identical to those obtained previously with the independent model approach for the response probability of 0.16 (see Table 4). However, it is worth noting that the estimated standard deviations of the RPFs obtained with the parallel dose–response assumption are approximately 33% smaller than those obtained without it at the response probability of 0.16 (shown in Table 4). The greater variance of the non-parallel RPF estimates is a reflection of the additional freedom that non-parallel dose–response curves have to diverge from each another. In fact, the assumption of parallelism leads to estimates of RPF sampling variability that are biased low when dose responses are not parallel. Allowance for non-parallelism, at least for 4-PeCDF, reveals additional uncertainty that should be accounted for when using TEFs (or RPFs) in risk assessment. Recently, Walker et al. (2005) published results from independent and simultaneous fits of the new NTP bioassay data against administered dose for TCDD, 4-PeCDF, PCB126, and the presumably equipotent mixture of the three chemicals with saturable (Hill type) dose–response models. For bile duct tumors (cholangiocarcinomas), liver adenomas, and lung tumors (cystic keratinizing epitheliomas), they obtained endpoint-specific RPFs (standard errors) for 4-PeCDF of 0.03 (0.10), 0.19 (0.25), and 0.36 (not determined), respectively, without assuming parallel dose responses, and RPF50s of 0.16 (0.04), 0.34 (0.08), and 0.26 (0.12), respectively, with the assumption of parallel dose responses. These values are quite close to our results for liver tumors (combined cholangiocarcinomas and hepatocellular adenomas), and the pattern of larger variability of RPF estimates obtained without assuming parallelism is also consistent with our results. RPF Sampling Distributions The development of empirical RPF sampling distributions allows for objective tests of a hypothesis that implicitly underlies the TEF concept, namely, that RPFs for different endpoints are actually just estimates of a common underlying population parameter, namely, the ‘‘true’’ TEF. They also permit the development of TEF estimates using objective weighting— e.g., inverse variance weighting—of endpoint- specific RPF estimates. In our view, this potential for objectivity is a marked improvement over previous approaches to the development of TEF values. Relative potency factor sampling distributions such as those we have developed also facilitate the exploration of probabilistic risk assessment approaches that accommodate uncertainty and variability in toxicity parameters as well as exposure inputs (USEPA, 2004). It is noteworthy that EPA recently concluded that sufficient data exist to conduct Monte Carlo analyses for 4-PeCDF: ‘‘For some chemicals, such as PCB 126, PeCDD and 4-PeCDF, there are sufficient data to apply these methods.’’ (USEPA, 2003a, Part II, Chapter 9). RPFs for Different Dose Metrics It is important to recognize that RPFs derived with administered dose do not account for important pharmacokinetic differences between congeners such as the more pronounced sequestration in the liver of 4-PeCDF relative to that of TCDD. Others have suggested that TEFs should account for congener-specific differences in pharmacokinetics (DeVito and Birnbaum, 1995; Neubert et al., 1992). DeVito et al. (1997) recommended that TEF values be developed on both an intake and a tissue concentration equivalent basis. In addition, the use of internal dose metrics has been endorsed by the USEPA for risk assessment purposes: . . .the scientific principle that the internal dose to the tissue of interest is the ultimate determinant of toxicity. . . .In its 1994 report, the NRC states that: . . .the target-site dose is the ultimate determinant of risk . . .. Thus, the manner or route of systemic exposure is secondary (except as it affects internal doses, e.g., by first-pass metabolism) to the internal dose. Others in government also reflect this fundamental principle. (USEPA, 2004). Certainly, TEFs developed with a body concentration dose metric would be the most compatible with the USEPA’s current use of lifetime average body burden when assessing the potential human risks posed by dioxin-like compounds. The RPF values we obtained assuming parallel dose–response curves range from about 0.27 using administered dose to about one order of magnitude smaller using liver concentration (0.014), area under the liver concentration curve (0.022), or lifetime average body concentration (0.036). The corresponding RPF16 estimates we obtained without the parallel dose– response assumption are very close to these values. If potential human cancer risk is the primary concern, it would seem that an appropriate RPF for 4-PeCDF would be either 0.27 or 0.036, depending on whether administered dose or lifetime average body burden is the preferred dose metric. Lipid concentration is another internal dose metric that merits discussion. At low exposure levels, it should be tightly correlated with both administered dose and liver concentration, but when exposure is sufficiently high to cause CYP1A2 induction and substantial sequestration of TCDD or 4-PeCDF in the liver, these two dose metrics should diverge from one 283 RELATIVE POTENCY FACTORS FOR 4-PeCDF another. This could be important for non-hepatic lesions such as gingival tumors, lung, pancreatic and uterine tumors, where the target organ ligand concentrations are presumably better represented by those in fat. Even in the liver, the most appropriate dose metric is unclear because the CYP1A2-bound fraction may be biologically inactive. It is worth noting in this regard that there is some evidence of hepatic sequestration in humans, particularly for more highly chlorinated congeners (Grassman et al., 2000; Iida et al., 1999; Kitamura et al., 2001). Sequestration is thus relevant for estimating the potential hazards of dioxin-like compounds to humans. Given the fact of hepatic sequestration, the appropriate dose metric for estimating relative potency depends, at least in part, on whether CYP1A2-bound dioxins are inactive or active. Studies conducted in CYP1A2 knockout mice have reported mixed results concerning this issue. Some report enhanced sensitivity to dioxin-induced immunotoxicity in CYP1A2 (/) mice (Smialowicz et al., 2004), others report no difference in dioxin-induced enzyme induction or hepatic oxidative stress between CYP1A2 (–/–) mice and CYP1A2 (þ/þ) mice (Liang et al., 1997; Slezak et al., 1999), and still others report that hepatotoxicity is dependent on CYP1A2 expression (Smith et al., 2001). Interestingly, enzyme activity data collected during the NTP bioassays suggest that the hepatic-sequestered fraction may not be inactive. In 4-PeCDF-treated animals receiving 100 ng TEQ/kg per day, CYP1A1 activity at 53 weeks was approximately double (3674 ± 401 pmol/min per milligram) that in TCDD-treated animals (1871 ± 109 pmol/min per milligram) receiving the same TEQ dose, despite comparably induced levels of CYP1A2 and a nearly 30-fold greater hepatic sequestration of 4-PeCDF than TCDD (Toyoshiba et al., 2004). If the CYP1A2 bound fraction were biologically inactive, one would expect much less, rather than more, CYP1A1 activity in the 4-PeCDF-exposed animals. An apparent limitation of RPFs based on internal dose metrics relative to those derived using administered doses relates to the uncertainty with which internal doses are estimated. The tissue concentration data that we used to estimate internal doses came from satellite groups of approximately 10 animals that may or may not be representative of the main study groups in the NTP bioassays, and we just assumed representativeness. There is also measurement error associated with the resulting internal dose estimates, and we have not taken this explicitly into account. One would expect that the internal dose metric RPF sampling distributions would widen, possibly substantially, if measurement errors were properly accounted for, compared to those we derived based solely on the binomial sampling variability of tumor incidence. However, it is worth noting in this regard that some distribution widening should also occur in the administered dose RPF sampling distributions, provided one properly accounted for the substantial uncertainty associated with the assumption that administered dose is a perfect linear surrogate for the unknown causal dose metric. Interesting questions, therefore, remain about what the most appropriate dose metric is for estimating, evaluating, and using RPFs and TEFs. Although these questions are certainly of scientific interest, they may be academic when it comes to practical risk assessment applications, which typically rely on estimated intake or extrapolated body burdens. We offer several recommendations regarding the practical application of RPFs in risk assessment. First, the RPFs for carcinogenicity of 4-PeCDF should not be combined with RPFs obtained from enzyme induction or other non-cancer endpoints, such as immune function changes. It is noteworthy that draft EPA guidance on the use of TEFs in ecological risk assessments already permits the use of endpoint-specific RPFs in place of generic TEFs. Specifically, the guidance states: Selection of an RPF based on a few data points, or even a single RPF value, is appropriate if the RPF data are of high quality and the overall species, endpoint, and dose specificity is greater than for the comparable TEF. (USEPA, 2003b) Second, cancer risk assessments should consider the full range of 4-PeCDF RPFs that can be generated from the available NTP (2003a, 2003b) bioassay data. Finally, we encourage the use of probabilistic risk assessment approaches that can accommodate both RPF point estimates and their sampling distributions. Such approaches provide a great deal more transparency regarding variability and uncertainty than the current practice of using upper bound point estimate TEFs (Finley et al., 2003). ACKNOWLEDGMENTS D.P. and T.B.S. provide consulting services to The Dow Chemical Company and they received financial support for conducting this research. D.P. is also serving as an expert witness to The Dow Chemical Company on dioxin-related matters. The authors appreciate the assistance of Dr. Edmund Crouch who provided the body burden estimates and the maximum likelihood estimation routines. REFERENCES Bailer, A. J., and Portier, C. J. (1988). Effects of treatment-induced mortality and tumor-induced mortality on tests for carcinogenicity in small samples. Biometrics 44, 417–431. Brewster, D. W., and Birnbaum, L. S. (1987). Disposition and excretion of 2,3,4,7,8-pentachlorodibenzofuran in the rat. Toxicol. Appl. Pharmacol. 90, 243–252. Brown, R. P, Delp, M. D., Lindstedt, S. L., Rhomberg, L. R., and Beliles, R. P. (1997). Physiological parameters values for PBPK models. Toxicol. Ind. Health 13, 407–484. Budinsky, R., Starr, T. B., Paustenbach, D., Landenberger, B., and Fontaine, D. (2004). A preliminary evaluation of the toxic equivalency factor (TEF) for 2,3,4,7,8-PCDF (4-PCDF) using data from the recent NTP dioxin bioassays. Organohalogen Cmpd. 66, 3433–3438. Carrier, G., Brunet R. C., and Brodeur, J. (1995). Modeling of the toxicokinetics of polychlorinated dibenzo-p-dioxins and dibenzofurans in 284 BUDINSKY ET AL. mammalians, including humans. II. Kinetics of absorption and disposition of PCDDs. Toxicol. Appl. Pharmacol. 131, 267–276. DeVito, M. J., and Birnbaum, L. S. (1995). The importance of pharmacokinetics in determining the relative potency of 2,3,7,8-tetrachlorodibenzo-pdioxin and 2,3,7,8-tetrachlorodiebenzofuran. Fundam. Appl. Toxicol. 24, 145–148. DeVito, M. J., Diliberto, J. J., Ross, D. G., Menache, M. G., and Birnbaum, L. S. (1997). Dose-response relationships for polyhalogenated dioxins and dibenzofurans following subchronic treatment in mice. Toxicol. Appl. Pharmacol. 147, 267–280. Finley, B. L., Connor, K. T., and Scott, P. K. (2003). The use of toxic equivalency factor distributions in probabilistic risk assessment for dioxins, furans, and PCBs. J. Toxicol. Environ. Health 66, 533–550. Fontaine, D. D., Paustenbach, D., Landenberger, B. D., Budinsky, R. A., and Starr, T. B. (2004). An evaluation of the toxic equivalency factor (TEF) for 2,3,4,7,8-PCDF (4-PCDF) using data from the recent NTP dioxin bioassays. Paper W8.7. Society for Risk Analysis Annual Meeting, 2004. Grassman, J. A., Needham, L. L., Masten, S. A., Patterson, D., Portier, C. J., Lucier, G. W., and Walker, N. J. (2000). Evidence of hepatic sequestration of dioxins in humans? An examination of tissue levels and CYP1A2 expression. Organohalogen Cmpds. 48, 87–90. Harper, N., Connor, K., and Safe, S. (1993). Immunotoxic potencies of polychlorinated biphenyl (PCB), dibenzofuran (PCDF) and dibenzo-p-dioxin (PCDD) congeners in C57BL/6 and DBA/2 mice. Toxicology 80, 217–227. Haws, L. C., Su, S. H., Harris, M., DeVito, M. J., Walker, N. J., Farland, W. H., Finley, B., and Birnbaum, L. S. (2005). Development of a refined database of mammalian relative potency estimates for dioxin-like compounds. Toxicol. Sci. 89, 4–30. Iida, T., Hirakawa, H., Matsueda, T., Nagayama, J., and Nagata, T. (1999). Polychlorinated dibenzo-p-dioxins and related compounds: Correlations of levels in human tissues and in blood. Chemosphere 38, 2767–2774. Kociba, R. J., Keyes, D. G., Beyer, J. E., Carreon, R. M., Wade, C. E., Dittenber, D. A., Kalnins, R. P., Frauson, L. E., Park, C. N., Barnard, S. D., et al. (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. Kitamura, K., Nagao, M., Yamada, T., Sunaga, M., Hata, J., and Watanabe, S. (2001). Dioxins in bile in relation to those in the human liver and blood. J. Toxicol. Sci. 26, 327–336. NTP (2003c). Toxicology and carcinogenesis studies of 3,3#,4,4#,5-pentachlorobiphenyl (PCB 126) in female Harlan Sprague-Dawley rats. NTP TR 520, NIH Publication No 04–4454. NTP (2003d). Toxicology and carcinogenesis studies of a mixture of 2,3,7,8tetrachlorodibenzo-p-dioxin (TCDD), 2,3,4,7,8-pentachlorodibenzofuran (PeCDF) and 3,3#,4,4#,5-pentachlorobiphenyl (PCB 126) in female Harlan Sprague-Dawley rats. NTP TR 526, NIH Publication No 04–4462. Olson, J. R., Bellin, J. S., and Barnes. D. C. (1989). Reexamination of data used for establishing toxicity equivalence factors (TEFS) for chlorinated dibenzo-p-dioxins and dibenzofurans (CDDS and CDFS) Chemosphere 18, 371–381. Patterson, D. G., Todd, G. D., Turner, W. E., Maggio, V., Alexander, L. R., and Needham, L. L. (1994). Levels of non-ortho-substituted (coplanar), monoand di-ortho-substituted polychlorinated biphenyls, dibenzo-p-dioxins, and dibenzofurans in human serum and adipose tissue. Environ. Health Perspect. 103(Suppl. 1), 195–204. Petreas, M., She, J., Winkler, J., Visita, P., McKinney, M., Reynolds, P., Smith, D., Gilliss, D., Hurley, S., Jeffrey, S., et al. (2000). Body burdens of organohalogens in California populations. Organohalogen Cmpds. 48, 17–20. Pluss, N., Poiger, H., Hohbach, C., Suter, M., and Schlatter, C. (1988). Subchronic toxicity of 2,3,4,7,8-pentachlorodibenzofuran (PeCDF) in rats. Chemosphere 17, 1099–1110. Safe, S. H. (1998). Development validation and problems with the toxic equivalency factor approach for risk assessment of dioxins and related compounds. J. Animal Sci. 76, 134–141. Slezak, B. P., Diliberto, J. J., and Birnbaum, L. S. (1999). 2,3,7,8-Tetrachlorodibenzo-p-dioxin-mediated oxidative stress in CYP1A2 knockout (CYP1A2–/–) mice. Biochem. Biophys. Res. Commun. 264, 376–379. Smialowicz, R. J., Burgin, D. E., Williams, W. C., Diliberto, J. J., Setzer, R. W., and Birnbaum, L. S. (2004). CYP1A2 is not required for 2,3,7,8tetrachloordibenzo-p-dioxin-induced immunosuppression Toxicology 197, 15–22. Smith, A. G., Clothier, B., Carthew, P., Childs, N. I., Sinclar, P. R., Nebert, D. W., and Dalton, T. P. (2001). Protection of the Cyp 1a2 (–/–) null mouse against urophorphyia and hepatic injury following exposure to 2,3,7,8tetrachlorodibenzo-p-dioxin. Toxicol. Appl. Pharmacol. 173, 89–98. Toyoshiba, H., Walker, N. J., Bailer, J. A., and Portier, C. J. (2004). Evaluation of toxic equivalency factors for induction of cytochrome P450 CYP1A1 and CYP1A2 enzyme activity by dioxin-like compounds. Toxicol. Appl. Pharmacol. 194, 156–168. Kreuzer, P. E., Csanady, G. A., Baur, C., Kessler, W., Papke, O., Greim, H., and Filser, J. G. (1997). 2,3,7,8-Tetrachlorodbenzo-p-dioxin (TCDD) and congeners in infants. A toxicokinetic model of human lifetime body burden by TCDD with special emphasis on its uptake by nutrition. Arch. Toxicol. 71, 383–400. USEPA. (1988). A cancer risk-specific dose estimate for 2,3,7,8-TCDD. Appendices A Through F. EPA/600/6–88/007-AB. Liang, H. C. L., McKinnon, R. A., and Nebert, D. (1997). Sensitivity of CYP1A1 mRNA inducibility by dioxin is the same in CYP1a2 (þ/þ) wild-type and Cyp1A2 (/) null mutant mice. Biochem. Pharmacol. 54, 1127–1131. USEPA. (2003a). Exposure and Human Health Reassessment of 2,3,7,8Tetrachlorodibenzo-p-Dioxin (TCDD) and Related Compounds. Washington, DC, National Academy Sciences (NAS) Review Draft, December 2003. Neubert, D., Golor, G., and Neubert, R. (1992). TCDD-toxcity equivalencies for PCDD/PCDF congeners: Prerequisites and limitations. Chemosphere 25, 65–70. USEPA. (2003b). Framework for Application of the Toxicity Equivalence Methodology for Polychlorinated Dioxins, Furans, and Biphenyls in Ecological Risk Assessment. EPA/630/P-03/002A, June 2003. NHANES (2005). National Center for Health Statistics. Third National Report on Human Exposure to Environmental Chemicals. U. S. Department of Health and Human Services, Centers for Disease Control and Prevention, National Center for Environmental Health, NCEH Pub. No. 05–0579. USEPA. (2004). An Examination of EPA Risk Assessment Principles and Practices. EPA/100/B-04/001, March 2004. NTP (2003a). Toxicology and carcinogenesis studies of 2,3,4,7,8-pentachlorodibenzofuran (PeCDF) in female Harlan Sprague-Dawley rats. NTP TR 525, NIH Publication No 04–4461. NTP (2003b). Toxicology and carcinogenesis studies of 2,3,7,8-tetrachlorodibenzo-p-dioxin in female Harlan Sprague-Dawley rats. NTP TR 521, NIH Publication No 04–4455. USEPA. (2001). Benchmark Dose Software. Version 1.3.2 downloaded from http://cfpub.epa.gov/ncea/cfm/recordisplay.cfm?deid¼20167, March 2004. van Birgelen, A. P. J. M., DeVito, M. J., Orzech, D., Walker, N., Birnbaum, L. S., Bucher, J., and Lucier, G. (1997). Design of 2-year bioassays with dioxin-like compounds in female Sprague-Dawley rats. Organohalogen Cmpds. 34, 154–159. Van den Berg, M., Bouwman, C., and Seinen, W. (1989). Hepatic retention of PCDDs and PCDFs in C57BL/6 and DBA/2 mice. Chemosphere 19, 795–782. Van den Berg, M., Birnbaum, L., Bosveld, A. T., Brunstrom, B., Cook, P., Feeley, M., Giesy, J. P., Hanberg, A., Hasegawa, R., Kennedy, S. W., et al. RELATIVE POTENCY FACTORS FOR 4-PeCDF 285 (1998). Toxic equivalency factors (TEFs) for PCBs, PCDDs, PCDFs for humans and wildlife. Environ. Health Perspect. 106, 775–792. Dose-additive carcinogenicity of a defined mixture of dioxin-like compounds. Environ. Health Perspect. 113, 43–48. Waern, F., Flodstrom, S., Busk, L., Kornevi, T., Nordgren, I., and Ahlborg, U. G. (1991). Relative liver tumour promoting activity and toxicity of some polychlorinated dibenzo-p-dioxin- and dibenzofuran-congeners in female Sprague-Dawley rats. Pharmacol. Toxicol. 69, 450–458. Wittsiepe, J., Furst, P., Schrey, P., Lemm, F., Kraft, M., Eberwein, G., Winneke, G., and Wilhelm, M. (2004). PCDD/F and dioxin-like PCB in human blood and milk from German mothers. Organohalogen Cmpds. 66, 2865–2872. Walker, N. J., Crockett, P., Nyska, A., Brix, A., Kokinen, M. P., Sells, D. M., Hailey, J. R., Easterling, M., Haseman, J. K., Yin, M., et al. (2005). Wittsiepe, J., Schrey, P., Ewers, U., Selenka, F., and Wilhelm, M. (2000). Decrease of PCDD/F levels in human blood from Germany over the past ten years (1989–1998). Chemosphere 40, 103–110.
© Copyright 2025 Paperzz