1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 PUBLIC RELEASE DRAFT December 2024 EPA Document# EPA-740-D-24-027 December 2024 United States Office of Chemical Safety and Environmental Protection Agency Pollution Prevention Draft Non-cancer Human Health Hazard Assessment for Diisobutyl Phthalate (DIBP) Technical Support Document for the Draft Risk Evaluation CASRN: 84-69-5 December 2024 ------- PUBLIC RELEASE DRAFT December 2024 28 TABLE OF CONTENTS 29 KEY ABBREVIATIONS AND ACRONYMS 5 30 SUMMARY 6 31 1 INTRODUCTION 9 32 1.1 Human Epidemiologic Data: Approach and Preliminary Conclusions 9 33 1.2 Laboratory Animal Data: Summary of Existing Assessments, Approach, and Methodology .... 11 34 1.2.1 Summary of Existing Assessments 11 35 1.2.2 Approach to Identifying and Integrating Laboratory Animal Data 14 36 1.2.3 New Literature Identified and Hazards of Focus for DIBP 16 37 2 TOXICOKINETICS 17 38 2.1 Oral Route 17 39 2.2 Inhalation Route 17 40 2.3 Dermal Route 17 41 3 NON-CANCER HAZARD IDENTIFICATION 19 42 3.1 Effects on the Developing Male Reproductive System 19 43 3.1.1 Summary of Available Epidemiological Studies 19 44 3.1.1.1 Previous Epidemiology Assessment (Conducted in 2019 or Earlier) 19 45 3.1.1.1.1 Health Canada (2018b) 20 46 3.1.1.1.2 Radkeetal. (2019b; 2018) 21 47 3.1.1.1.3 NASEM (2017) 22 48 3.1.1.1.4 Summary of the Existing Assessments of Male Reproductive Effects 23 49 3.1.1.2 EPA Summary of New Studies (2018 to 2019) 23 50 3.1.2 Summary of Laboratory Animals Studies 26 51 3.1.2.1 Developing Male Reproductive System 32 52 3.1.2.2 Other Developmental Outcomes 33 53 4 DOSE-RESPONSE ASSESSMENT 35 54 4.1 Selection of Studies and Endpoints for Non-cancer Health Effects 36 55 4.2 Non-cancer Oral Points of Departure for Acute, Intermediate, and Chronic Exposures 36 56 4.2.1 Studies Considered for the Non-Cancer POD 36 57 4.2.2 Options Considered by EPA for Deriving the Acute Non-Cancer POD 41 58 4.2.2.1 Option 1. NOAEL/LOAEL Approach 41 59 4.2.2.2 Option 2. Application of a Data-Derived Adjustment Factor 41 60 4.2.2.3 Option 3. BMD Analysis of Individual Fetal Testicular Testosterone Studies 42 61 4.2.3 POD Selected for Acute, Intermediate, and Chronic Durations 42 62 4.3 Weight of The Scientific Evidence Conclusion: POD for Acute, Intermediate, and Chronic 63 Durations 46 64 5 CONSIDERATION OF PESS AND AGGEGRATE EXPOSURE 48 65 5.1 Hazard Considerations for Aggregate Exposure 48 66 5.2 PESS Based on Greater Susceptibility 48 67 6 POINTS OF DEPARTURE USED TO ESTIMATE RISKS FROM DIBP EXPOSURE, 68 CONCLUSOINS, AND NEXT STEPS 56 69 REFERENCES 57 Page 2 of 94 ------- PUBLIC RELEASE DRAFT December 2024 70 APPENDICES 67 71 Appendix A Existing Assessments of DIBP 67 72 Appendix B Fetal Testicular Testosterone as an Acute Effect 71 73 Appendix C Calculating Daily Oral Human Equivalent Doses and Human Equivalent 74 Concentrations 72 75 C. 1 DIBP Non-cancer HED and HEC Calculations for Acute, Intermediate, and Chronic 76 Duration Exposures 73 77 Appendix D Considerations for Benchmark Response (BMR) Selection for Reduced Fetal 78 Testicular Testosterone 75 79 D.l Purpose 75 80 D.2 Methods 75 81 D.3 Results 76 82 D.4 Weight of Scientific Evidence Conclusion 77 83 Appendix E BENCHMARK DOSE MODELING OF FETAL TESTICULAR 84 TESTOSTERONE 80 85 E.l BMD Model Results (Gray et al. 2021) 81 86 E.2 BMD Model Results (Howdeshell et al. 2008) 84 87 E.3 BMD Model Results (Hannas et al. 2011) 88 88 89 LIST OF TABLES 90 Table 1-1. Summary of DIBP Non-cancer PODs Selected for Use by other Regulatory Organizations. 12 91 Table 3-1. Summary of Scope and Methods Used in Previous Assessments to Evaluate the Association 92 Between DIBP Exposure and Male Reproductive Outcomes 19 93 Table 3-2. Summary of Epidemiologic Evidence of Male Reproductive Effects Associated with 94 Exposure to DIBP 21 95 Table 3-3. Summary of Studies of DIBP Evaluating Developmental and Reproductive Outcomes 27 96 Table 4-1. Summary of NASEM (2017) Meta-Analysis and BMD Modeling for Effects of DIBP in Fetal 97 Testosterone ab 38 98 Table 4-2. Overall Analyses of Rat Studies of DIBP and Fetal Testosterone (Updated Analysis 99 Conducted by EPA) 39 100 Table 4-3. Benchmark Dose Estimates for DIBP and Fetal Testosterone in Rats 39 101 Table 4-4. Summary of Dichotomous BMD Analysis of Data from Saillenfait et al. (2008) by Blessinger 102 et al. (2020)° 40 103 Table 4-5. Dose-Response Analysis of Selected Studies Considered for Acute, Intermediate, and 104 Chronic Exposure Scenarios 44 105 Table 5-1. PESS Evidence Crosswalk for Biological Susceptibility Considerations 49 106 107 LIST OF FIGURES 108 Figure 1-1. Overview of DIBP Human Health Hazard Assessment Approach 15 109 Figure 3-1. Hypothesized Phthalate Syndrome Mode of Action Following Gestational Exposure 32 110 111 LIST OF APPENDIX TABLES 112 Table Apx A-l. Summary of Peer-review, Public Comments, and Systematic Review for Existing 113 Assessments of DIBP 67 Page 3 of 94 ------- 114 115 116 117 118 119 120 121 122 123 124 125 126 127 PUBLIC RELEASE DRAFT December 2024 TableApx D-l. Comparison of BMD/BMDL Values Across BMRs of 5%, 10%, and 40% with PODs and LOAELs for Apical Outcomes for DEHP, DBP, DIBP, BBP, DCHP, and DINP .... 79 Table Apx E-l. Summary of BMD Model Results for Decreased Ex Vivo Fetal Testicular Testosterone 81 Table Apx E-2. Ex Vivo Fetal Rat Testicular Testosterone Data (Gray et al. 2021) 81 Table Apx E-3. BMD Model Results Ex Vivo Fetal Testicular Testosterone (Gray et al. 2021) 82 Table Apx E-4. Ex Vivo Fetal Rat Testicular Testosterone Data (Howdeshell et al. 2008) 84 Table Apx E-5. BMD Model Results Ex Vivo Fetal Testicular Testosterone (Howdeshell et al. 2008). 86 Table Apx E-6. Ex Vivo Fetal Rat Testicular Testosterone Data (Hannas et al. 2011) 88 Table Apx E-7. BMD Model Results Ex Vivo Fetal Testicular Testosterone (All Dose Groups) (Hannas etal. 2011) 90 Table Apx E-8. BMD Model Results Ex Vivo Fetal Testicular Testosterone (Highest Dose Group Removed) (Hannas et al. 2011) 91 Page 4 of 94 ------- 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 PUBLIC RELEASE DRAFT December 2024 KEY ABBREVIATIONS AND ACRONYMS ADME Absorption, distribution, metabolism, and excretion BMD Benchmark dose BMDL Benchmark dose lower bound BMR Benchmark response CASRN Chemical abstracts service registry number CPSC Consumer Product Safety Commission (U.S.) BBP Butyl -b enzy 1 -phthal ate DBP Dibutyl phthal ate DEHP Di-ethylhexyl phthalate DIBP Diisobutyl phthalate DINP Di-isononyl phthalate ECHA European Chemicals Agency EPA Environmental Protection Agency (U.S.) GD Gestational day HEC Human equivalent concentration HED Human equivalent dose LOAEL Lowest-observable-adverse-effect level LOEL Lowest-observable-effect level MOA Mode of action MOE Margin of exposure NICNAS National Industrial Chemicals Notification and Assessment Scheme NOAEL No-observed-adverse-effect level OECD Organisation for Economic Co-operation and Development OPPT Office of Pollution Prevention and Toxics PBPK Physiologically based pharmacokinetic PECO Population, exposure, comparator, and outcome PESS Potentially exposed or susceptible subpopulations PND Postnatal day POD Point of departure RPF Relative Potency Factor SACC Science Advisory Committee on Chemicals SD Sprague-Dawley TSCA Toxic Substances Control Act UF Uncertainty factor U.S. United States Page 5 of 94 ------- 164 165 166 167 168 169 170 171 172 173 174 175 176 111 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 PUBLIC RELEASE DRAFT December 2024 ACKNOWLEGEMENTS This report was developed by the United States Environmental Protection Agency (U.S. EPA or the Agency), Office of Chemical Safety and Pollution Prevention (OCSPP), Office of Pollution Prevention and Toxics (OPPT). Acknowledgements The Assessment Team gratefully acknowledges the participation, review, and input from EPA OPPT and OSCPP senior managers and science advisors.The Agency is also grateful for assistance from the following EPA contractors for the preparation of this draft technical support document: ICF (Contract No. 68HERC23D0007); and SRC, Inc. (Contract No. 68HERH19D0022). Special acknowledgement is given for the contributions of technical experts from EPA's Office of Research and Development (ORD) including Justin Conley, Earl Gray, and Tammy Stoker. As part of an intra-agency review, this technical support document was provided to multiple EPA Program Offices for review. Comments were submitted by EPA's Office of General Counsel (OGC) and ORD. Docket Supporting information can be found in the public docket, Docket ID EPA-HQ-QPPT-2018-0434. Disclaimer Reference herein to any specific commercial products, process, or service by trade name, trademark, manufacturer, or otherwise does not constitute or imply its endorsement, recommendation, or favoring by the United States Government. Authors: Collin Beachum (Management Lead), Brandall Ingle-Carlson (Assessment Lead), Myles Hodge (Human Health Hazard Assessment Lead), Anthony Luz (Human Health Hazard Discipline Lead), Christelene Horton (Human Health Hazard Assessor) Contributors: Azah Abdallah Mohamed, Devin Alewel, John Allran, Rony Arauz Melendez, Sarah Au, Lillie Barnett, Jone Corrales, Maggie Clark, Daniel DePasquale, Lauren Gates, Amanda Gerke, Annie Jacob, Ryan Klein, Sydney Nguyen, Ashley Peppriell, Brianne Raccor, Maxwell Sail, Joe Valdez, Leora Vegosen, Susanna Wegner Technical Support: Kelley Stanfield, Hillary Hollinger, S. Xiah Kragie This draft technical support document was reviewed and cleared for release by OPPT and OCSPP leadership. Page 6 of 94 ------- 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 PUBLIC RELEASE DRAFT December 2024 SUMMARY This technical support document is in support of the TSCA Draft Risk Evaluation for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024i). This document describes the use of available information to identify the non-cancer hazards associated with exposure to DIBP and the points of departure (PODs) to be used to estimate risks from DIBP exposures in the draft risk evaluation of DIBP. Environmental Protection Agency (EPA, or the Agency) summarizes the cancer and genotoxicity hazards associated with exposure to DIBP in the Draft Cancer Raman Health Hazard Assessment for Di(2-ethylhexyl) Phthalate (DEHP), Dibutyl Phthalate (DBP), Diisobutyl Phthalate (DIBP), Butyl Benzyl Phthalate (BBP) and Dicyclohexyl Phthalate (DCHP) (U.S. EPA. 2025a). EPA identified effects on the developing male reproductive system as the most sensitive and robust non- cancer hazard associated with oral exposure to DIBP in experimental animal models (Section 3.1). Existing assessments of DIBP also identified effects on the developing male reproductive system as the most sensitive and robust non-cancer effect following oral exposure to DIBP. Existing assessments included those by the U.S. Consumer Product Safety Commission (U.S. CPSC. 2014. 2011). Health Canada (ECCC/HC. 2020; EC/HC. 2015b). European Chemicals Agency (2017a. b), and the Australian National Industrial Chemicals Notification and Assessment Scheme (NICNAS. 2008a). as well as a systematic review by Yost et al., (2019). which drew conclusions consistent with those of the aforementioned regulatory bodies. EPA also considered epidemiologic evidence qualitatively as part of hazard identification and characterization. However, epidemiologic evidence for DIBP was not considered further for dose response analysis due to limitations and uncertainties in exposure characterization (discussed further in Section 1.1). Use of epidemiologic evidence qualitatively is consistent with phthalates assessment by Health Canada, U.S. CPSC, NICNAS, and ECHA. As discussed further in Section 3.1.2, EPA identified 13 oral exposure studies (11 of rats, 2 of mice) that have investigated the developmental and reproductive effects of DIBP following gestational and/or perinatal exposure to DIBP (Gray et al.. 2021; Pan et al.. 2017; Saillenfait et al.. 2017; Wang et al.. 2017; Sedha et al.. 2015; Furr et al.. 2014; Hannas et al.. 2012; Hannas et al.. 2011; Howdeshell et al.. 2008; Saillenfait et al.. 2008; BASF. 2007; Borch et al.. 2006; Saillenfait et al.. 2006). No one- or two- generation reproduction studies of DIBP are available for any route of exposure. Across available studies, the most sensitive developmental effects identified by EPA include effects on the developing male reproductive system consistent with a disruption of androgen action and the development of phthalate syndrome. EPA is proposing a POD of 24 mg/kg-day (human equivalent dose [HED] of 5.7 mg/kg-day) based on phthalate syndrome-related effects on the developing male reproductive system (i.e., decreased fetal testicular testosterone) to estimate non-cancer risks from oral exposure to DIBP for acute, intermediate, and chronic durations of exposure in the draft risk evaluation of DIBP. The proposed POD was derived from benchmark dose (BMD) modeling of ex vivo fetal testicular testosterone data and supports a 95 percent lower confidence limit on the BMD associated with a benchmark response (BMR) of 5 percent (BMDLs) of 24 mg/kg-day (Gray et al.. 2021). The Agency has performed 3/4 body weight scaling to yield the HED and is applying the animal to human extrapolation factor (i.e., interspecies extrapolation; UFa) of 3x and a within human variability extrapolation factor (i.e., intraspecies extrapolation; UFh) of 10x. Thus, a total uncertainty factor (UF) of 30x is applied for use as the benchmark margin of exposure (MOE). Based on the strengths, limitations, and uncertainties discussed Section 4.3, EPA reviewed the weight of the scientific evidence and has robust overall confidence in the proposed POD based on decreased fetal testicular testosterone for use in characterizing risk from exposure to DIBP for acute, intermediate, and chronic exposure scenarios. The applicability and relevance of this POD for all exposure durations (acute, intermediate, and chronic) is described in the introduction to Section 4 and additionally in Page 7 of 94 ------- 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 PUBLIC RELEASE DRAFT December 2024 Section 4.2 and Appendix B. For purposes of assessing non-cancer risks, the selected POD is considered most applicable to women of reproductive age, pregnant women, and male infants. Use of this POD to assess risk for other age groups (e.g., older children, adult males, and the elderly) is considered to be conservative and appropriate for a screening level assessment for these other age groups. No data are available for the dermal or inhalation routes that are suitable for deriving route-specific PODs. Therefore, EPA is using the acute/intermediate/chronic oral PODs to evaluate risks from dermal and inhalation exposure to DIBP. For the dermal route, differences in absorption are being accounted for in dermal exposure estimates in the draft risk evaluation for DIBP. For the inhalation route, EPA is extrapolating the oral HED to an inhalation human equivalent concentration (HEC) per EPA's Methods for derivation of inhalation reference concentrations and application of inhalation dosimetry (U.S. EPA. 1994) using the updated human body weight and breathing rate relevant to continuous exposure of an individual at rest provided in EPA's Exposure factors handbook: 2011 edition (U.S. EPA. 201 lb). Table ES-1 and Section 6 summarize EPA's selection of the oral HED and inhalation HEC values used to estimate non-cancer risk from acute/intermediate/chronic exposure to DIBP in the draft risk evaluation of DIBP. EPA is soliciting comments from the Science Advisory Committee on Chemicals (SACC) and the public on the non-cancer hazard identification, dose-response and weight of evidence analyses, and the selected POD for use in risk characterization of DIBP. Table ES-1. Non-cancer HED and HEC Used to Estimate Ris ks Exposure Scenario Target Organ System Species Duration POD (mg/kg- day) Effect HED (mg/ kg-day) HEC (mg/m3) [ppm] Benchmark MOE Reference Acute, intermediate, chronic Developmental toxicity Rat 4 days during gestation (GDs 14- 18) BMDLS= 24 I ex vivo fetal testicular testosterone production 5.7 30.9 [2.71] UFa= 311 ufh=io Total UF=30 Grav et al.. 2021 HEC = human equivalent concentration; HED = human equivalent dose; MOE = margin of exposure; NOAEL = no-observed-adverse- effect level; POD = point of departure; UF = uncertainty factor "EPA used allometric bodv weight scaling to the three-quarters power to derive the HED. Consistent with EPA Guidance (U.S. EPA, 201 let. the UFa was reduced from 10 to 3. Page 8 of 94 ------- 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 PUBLIC RELEASE DRAFT December 2024 1 INTRODUCTION In December 2019, EPA designated diisobutyl phthalate (DIBP) (CASRN 85-69-5) as a high-priority substance for risk evaluation following the prioritization process as required by Section 6(b) of the Toxic Substances Control Act (TSCA) and implementing regulations (40 CFR 702). The Agency published the draft and final scope documents for DIBP in 2020 (U.S. EPA. 2020a. b). Following publication of the final scope document, one of the next steps in the TSCA risk evaluation process is to identify and characterize the human health hazards of DIBP and conduct a dose-response assessment to determine the toxicity values to be used to estimate risks from DIBP exposures. This technical support document summarizes the non-cancer human health hazards associated with exposure to DIBP and proposes non-cancer toxicity values to be used to estimate risks from DIBP exposures. Cancer human health hazards associated with exposure to DIBP are summarized in EPA's Draft Cancer Raman Health Hazard Assessment for Di (2-e thy Ihexyl) Phthalate (DEHP), Dibutyl Phthalate (DBP), Diisobutyl Phthalate (DIBP), Butyl Benzyl Phthalate (BBP) and Dicyclohexyl Phthalate (DCHP) (U.S. EPA. 2025a). Over the past several decades, the human health effects of DIBP have been reviewed by several regulatory and authoritative agencies, including: the U.S. Consumer Product Safety Commission (U.S. CPSC); Health Canada; the European Chemicals Agency (ECHA); the Australian National Industrial Chemicals Notification and Assessment Scheme (NICNAS); and The National Academies of Sciences, Engineering, and Medicine (NASEM). EPA relied on information published in these assessments as a starting point for its human health hazard assessment of DIBP. Additionally, EPA considered new literature published since the most recent existing assessments of DIBP to determine if newer information might support the identification of new human health hazards or lower PODs for use in estimating human risk. EPA's process for considering and incorporating new DIBP literature is described in the Draft Systematic Review Protocol for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024k). EPA's approach and methodology for identifying and using human epidemiologic data and experimental laboratory animal data is described in Sections 1.1 and 1.2 1.1 Human Epidemiologic Data: Approach and Preliminary Conclusions To identify and integrate human epidemiologic data into the draft DIBP Risk Evaluation, EPA first reviewed existing assessments of DIBP conducted by regulatory and authoritative agencies, as well as several systematic reviews of epidemiologic studies of DIBP published by Radke et al., in the open literature. Although the authors (i.e., Radke et al.) are affiliated with the U.S. EPA's Center for Public Health and Environmental Assessment, the reviews do not reflect EPA policy. Existing epidemiologic assessments reviewed by EPA are listed below. As described further in Appendix A, most of these assessments have been subjected to peer-review and/or public comment periods and have employed formal systematic review protocols of varying structure and scope. The assessments and open literature used as a baseline in this risk evaluation are listed below. Supporting documentation: Evaluation of epidemiologic studies on phthalate compounds and their metabolites for hormonal effects, growth and development and reproductive parameters (Health Canada. 2018b); Supporting documentation: Evaluation of epidemiologic studies on phthalate compounds and their metabolites for effects on behaviour and neurodevelopment, allergies, cardiovascular function, oxidative stress, breast cancer, obesity, and metabolic disorders (Health Canada. 2018a): Application of systematic review methods in an overall strategy for evaluating low-dose toxicity fi'om endocrine active chemicals ^NASEM. 2017); Page 9 of 94 ------- 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 PUBLIC RELEASE DRAFT December 2024 Phthalate exposure and male reproductive outcomes: A systematic review of the human epidemiological evidence (Radke et al.. 2018); Phthalate exposure andfemale reproductive and developmental outcomes: A systematic review of the human epidemiological evidence (Radke et al.. 2019b); Phthalate exposure and metabolic effects: A systematic review of the human epidemiological evidence (Radke et al.. 2019a); and Phthalate exposure and neurodevelopment: A systematic review and meta-analysis of human epidemiological evidence (Radke et al.. 2020a). Next, EPA sought to identify new population, exposure, comparator, and outcome (PECO)-relevant literature published since the most recent existing assessment(s) of DIBP by applying a literature inclusion cutoff date. For DIBP, the applied cutoff date was based on existing assessments of epidemiologic studies of phthalates by Health Canada (2018a. b), which included literature up to January 2018. The Health Canada (2018a. b) epidemiologic evaluations were considered the most appropriate existing assessments for setting a literature inclusion cutoff date because those assessments provided a robust and the most recent evaluation of human epidemiologic data for DIBP. Health Canada evaluated epidemiologic study quality using the Downs and Black method (Downs and Black. 1998) and reviewed the database of epidemiologic studies for consistency, temporality, exposure-response, strength of association, and database quality to determine the level of evidence for association between urinary DIBP metabolites and health outcomes. New PECO-relevant literature published between 2018 to 2019 was identified through the literature search conducted by EPA in 2019, as well as references published between 2018 to 2023 that were submitted with public comments to the DIBP Docket (https://www.regulations.gov/docket/EPA-HQ-QPPT-2018-0434). and these studies were evaluated for data quality and extracted consistent with EPA's Draft Systematic Review Protocol Supporting TSCA Risk Evaluations for Chemical Substances (U.S. EPA. 2021). Data quality evaluations for new studies reviewed by EPA are provided in the Data Quality Evaluation Information for Raman Health Hazard Epidemiology for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024c). As described further in the Draft Systematic Review Protocol for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024k). EPA considers phthalate metabolite concentrations in urine to be an appropriate proxy of exposure from all sourcesincluding exposure through ingestion, dermal absorption, and inhalation. As described in the Application of US EPA IRIS systematic review methods to the health effects of phthalates: Lessons learned and path forward (Radke et al.. 2020b). the "problem with measuring phthalate metabolites in blood and other tissues is the potential for contamination from outside sources" (Calafat et al.. 2015). Phthalate diesters present from exogenous contamination can be metabolized to the monoester metabolites by enzymes present in blood and other tissues, but not urine." Therefore, EPA has focused its epidemiologic evaluation on urinary biomonitoring data; new epidemiologic studies that examined DIBP metabolites in matrices other than urine were considered supplemental and not evaluated for data quality. EPA used epidemiologic studies of DIBP qualitatively. This is consistent with Health Canada, U.S. CPSC, and ECHA. EPA did not use epidemiology studies quantitatively for dose-response assessment, primarily due to uncertainty associated with exposure characterization. Primary sources of uncertainty include the source(s) of exposure; timing of exposure assessment that may not be reflective of exposure during outcome measurements; and use of spot-urine samples, which due to rapid elimination kinetics may not be representative of average urinary concentrations that are collected over a longer term or calculated using pooled samples. The majority of epidemiological studies introduced additional uncertainty by considering DIBP in isolation and failing to account for confounding effects from co- exposure to mixtures of multiple phthalates (Shin et al.. 2019; Aylward et al.. 2016). Conclusions from Page 10 of 94 ------- 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 PUBLIC RELEASE DRAFT December 2024 Health Canada (2018a. b), NASEM (2017) and systematic review articles by Radke et al. (2020a; 2019b; 2019a; 2018) regarding the level of evidence for association between urinary DIBP metabolites and each health outcome were reviewed by EPA and used as a starting point for its human health hazard assessment. The Agency also evaluated and summarized new epidemiologic studies identified by EPA's systematic review process (as described in the Draft Systematic Review Protocol for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024k)) to use qualitatively during evidence integration to inform hazard identification and the weight of scientific evidence (Shin et al.. 2019; Aylward et al.. 2016). 1.2 Laboratory Animal Data: Summary of Existing Assessments, Approach, and Methodology 1.2.1 Summary of Existing Assessments The human health hazards of DIBP have been evaluated in existing assessments by the U.S. CPSC (2014. 2011). Health Canada (ECCC/HC. 2020; EC/HC. 2015b). ECHA (2017a. b), and Australia NICNAS (2016. 2008a. b). These assessments have consistently identified toxicity to the developing male reproductive system as the most sensitive and robust outcome for use in estimating human risk from exposure to DIBP. The PODs from these assessments are shown in Table 1-1. Additionally, a recent systematic review of animal toxicology studies of DIBP was published by Yost et al. (2019) in the open literature. Although the authors (i.e., Yost et al.) are affiliated with the U.S. EPA's Center for Public Health and Environmental Assessment, the review does not reflect EPA policy. Consistent with existing assessments of DIBP by regulatory bodies, Yost et al. (2019) concluded that there was: robust evidence that DIBP causes male reproductive toxicity and robust evidence that DIBP causes developmental toxicity. Additionally, Yost et al. (2019) concluded that there was slight evidence for female reproductive toxicity and effects on the liver, and indetermincmt evidence for effects on kidney. However, for these hazards, evidence was "limited by the small number of studies, experimental designs that were suboptimal for evaluating outcomes, and study evaluation concerns such as incomplete reporting of methods and results." Page 11 of 94 ------- PUBLIC RELEASE DRAFT December 2024 394 Table 1-1. Summary of DIBP Non-cancer POPs Selected for Use by other Regulatory Organizations Brief Study Description NOAEL/ LOAEL (mg/kg- day) Critical Effect o w u in a. U t/5 P ECCC/HC (2020) NICNAS (2008a) 3 03 (N O w < X u H 3 O w < X u UJ Pregnant Sprague-Dawley rats (20-22 pregnant rats/dose) gavaged with 0, 250, 500, 750, 1000 mg/kg-day DIBP on GD 6-20 (non-guideline study) (Saillenfait et al 2006) 250/500 I fetal body weight (both sexes); t incidence of cryptorchidism Pregnant Sprague-Dawley rats (11-14 dams/dose) gavaged with 0, 125, 250, 500, 625 mg/kg-day of DIBP on GD 12-21 (non-guideline study) (Saillenfait et al 2008) 125/250 I AGD, NR, testicular pathology (degeneration of seminiferous tubules) and oligo-/azoospermia in epididymis) None/ 125 Testicular pathology (degeneration of seminiferous tubules) and oligo-/azoospermia in epididymis) Pregnant rats (6-8/group) exposed to 0, 20, 200, 2000, or 10,000 ppm DBP via diet from GDI5 - PND21 (equivalent to 0, 1.5, 14, 148, 712 mg/kg- day [males]; 0, 3, 29, 291, 1372 mg/kg-day [females]). F1 evaluated at PND14, PND21, & PNW 8-11 (non-suideline study) (Lee et al 2004)6 None/ 2.5b Reduced spermatocyte development (PND 21) and mammary gland changes (vacuolar degeneration, alveolar hypertrophy) in adult male offspring6 Page 12 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Brief Study Description NOAEL/ LOAEL (mg/kg- day) Critical Effect o (N- u in a. U t/5 P o (N O (N u u u S3 00 o o w in < Z u aj (N O w < B U H X>l 03 O w < B U H 11ECHA (2012a. b) considered the study by Saillenfait et al. to support a LOAEL of 125 mg/kg-day based on increased incidence of testicular pathology. h ECHA (2017a. b) concluded "Few reproductive toxicity studies have been published on [DIBP] compared to DEHP and DBP. No two-generation studies are available and the substance has not been tested at doses <100 mg/kg bw/d. Current data suggest that DIBP could have similar effects to DBP, if studied at lower dose levels. If the potency difference between DIBP and DBP, as a very rough estimate of the observed effects in Saillenfait et al. (2008) (type of effects seen at 500 and 625 mg/kg bw-day, corresponding to a difference of 25%). is extrapolated from the high dose area to the lower dose area, an estimated LOAEL for DIBP would be 25% higher than the current LOAEL for DBP (2 mg/kg bw-day). Available information is shown in Table B7. A LOAEL for DIBP of 2.5 mg/kg bw-day is selected for use in the current combined risk assessment Page 13 of 94 ------- 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 PUBLIC RELEASE DRAFT December 2024 1.2.2 Approach to Identifying and Integrating Laboratory Animal Data Figure 1-1 provides an overview of EPA's approach to identifying and integrating laboratory animal data into the draft DIBP Risk Evaluation. EPA first reviewed existing assessments of DIBP conducted by various regulatory and authoritative agencies. Existing assessments reviewed by EPA are listed below. The purpose of this review was to identify sensitive and human relevant hazard outcomes associated with exposure to DIBP, and while these authoritative sources identified a broader pool of studies to inform hazard identification, EPA only selected those studies used quantitatively for dose- response analysis in prior assessments for further consideration in estimating human risk. As described further in Appendix A, most of these assessments have been subjected to external peer-review and/or public comment periods but have not employed formal systematic review protocols. Toxicity review of diisobutylphthalate (DiBP, CASRN84-69-5) (U.S. CPSC. 2011); Chronic Hazard Advisory Panel on Phthalates and Phthalate Alternatives (with appendices) (U.S. CPSC. 2014); State of the science report: Phthalate substance grouping: Medium-chain phthalate esters: Chemical Abstracts Service Registry Numbers: 84-61-7; 84-64-0; 84-69-5; 523-31-9; 5334-09- 8; 16883-83-3; 27215-22-1; 27987-25-3; 68515-40-2; 71888-89-6 (EC/HC. 2015b): Screening assessment - Phthalate substance grouping (ECCC/HC. 2020); Existing chemical hazard assessment report: Diisobutyl phthalate (NICNAS. 2008a); Phthalates hazard compendium: A summary of physicochemical and human health hazard data for 24 ortho-phthalate chemicals (NICNAS. 2008b); C4-6 side chain transitional phthalates: Human health tier II assessment (NICNAS. 2016); Committee for Risk Assessment (RAC) Opinion on an Annex XV dossier proposing restrictions on four phthalates (ECHA. 2012b); Committee for Risk Assessment (RAC) Committee for Socio-economic Analysis (SEAC): Background document to the Opinion on the A nnex XV dossier proposing restrictions on four phthalates (ECHA. 2012a); Opinion on an Annex XV dossier proposing restrictions on four phthalates (DEHP, BBP, DBP, DIBP) (ECHA. 2017b); Annex to the Background document to the Opinion on the Annex XV dossier proposing restrictions on four phthalates (DEHP, BBP, DBP, DIBP) (ECHA. 2017a); Application of systematic review methods in an overall strategy for evaluating low-dose toxicity fi'om endocrine active chemicals (NASEM. 2017); and Hazards of diisobutyl phthalate (DIBP) exposure: A systematic review of animal toxicology studies (Yost et al.. 2019). Next, EPA sought to identify new PECO-relevant literature published since the most recent existing assessment(s) of DIBP by applying a literature inclusion cutoff date. Along with existing assessments, EPA used the systematic review in the open literature by Yost et al. (2019) as the starting point for this draft document (publicly available at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8596331/). The systematic review by Yost et al. employed a systematic review protocol and included scientific literature up to July 2017. Further, Yost et al. (2019) considered a range of human health hazards (e.g., developmental toxicity, male and female reproductive toxicity, liver and kidney toxicity, and cancer) across all durations (i.e., acute, intermediate, subchronic, chronic) and routes of exposure (i.e., oral, dermal, inhalation). Likewise, Yost et. al reached similar conclusions related to the human health hazards of DIBP, as other assessments by U.S. CPSC, Health Canada, NICNAS, ECHA, and NASEM. Page 14 of 94 ------- 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 PUBLIC RELEASE DRAFT December 2024 EPA considered literature published between 2017 to 2019 further as shown in Figure 1-1. EPA first screened titles and abstracts and then full texts for relevancy using PECO screening criteria described in the Draft Systematic Review Protocol for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024k). EPA then identified PECO-relevant literature published since the most recent and comprehensive existing assessment of DIBP by applying a literature inclusion cutoff date from this assessment. Figure 1-1. Overview of DIBP Human Health Hazard Assessment Approach 11 Any study that was considered for dose-response assessment, not necessarily limited to the study used for POD selection. h Extracted information includes PECO relevance, species, exposure route and type, study duration, number of dose groups, target organ/systems evaluated, study-wide LOEL, and PESS categories. Next, EPA reviewed and extracted key study information from those new studies including: PECO relevance; species tested; exposure route, method, and duration of exposure; number of dose groups; target organ/systems evaluated; information related to potentially exposed or susceptible subpopulations (PESS); and the study-wide lowest-observable-effect level (LOEL) (Figure 1-1). New information for DIBP was limited to one new oral exposure study, and the study LOEL was converted to an HED by allometric scaling across species using the 3A power of body weight (BW3 4) for oral data, which is the approach recommended by U.S. EPA when physiologically based pharmacokinetic models (PBPK) or other information to support a chemical-specific quantitative extrapolation is absent (U.S. EPA. 2011c). EPA's use of allometric body weight scaling is described further in Appendix C. Effects on the developing male reproductive system are a focus of EPA's DIBP hazard assessment. Therefore, EPA also considered literature identified outside of the 2019 TSCA literature search that was identified through development of EP A's Draft Proposed Approach for Cumulative Risk Assessment of High-Priority Phthalates and a Manufacturer-Requested Phthalate under the Toxic Substances Control Act (U.S. EPA. 2023a). which focused on the developing male reproductive system. Data quality evaluations for DIBP animal toxicity studies are provided in the Data Quality Evaluation Information for Human Health Hazard Animal Toxicology for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024b). Notably, Yost et al. (2019) included data quality evaluations, which are documented and publicly available in the Health Assessment Workspace Collaborative (HAWC) (https://hawc.epa.gov/assessment/497/). As described further in the Draft Systematic Review Protocol for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024k). EPA relied on the data quality evaluations completed by Yost et al. (2019). which were imported from HAWC to Distiller and are included in the Data Quality Evaluation Information for Human Health Hazard Animal Toxicology for Diisobutyl Page 15 of 94 ------- 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 PUBLIC RELEASE DRAFT December 2024 Phthalate (DIBP) (U.S. EPA. 2024b). Further, as described in the Draft Systematic Review Protocol for DiisobutylPhthalate (DIBP) (U.S. EPA. 2024k). OPPT harmonized its draft TSCA systematic review protocol for human health animal toxicology and epidemiologic study data quality evaluations with the process described in the IRIS Systematic Review Handbook (U.S. EPA. 2022). Therefore, the data quality evaluations completed by Yost et al. (2019) are reflective of the harmonized TSCA data quality evaluation process. 1.2.3 New Literature Identified and Hazards of Focus for DIBP In its review of literature published between 2017 to 2019 for new information on sensitive human health hazards not previously identified in existing assessments, including information that may indicate a more sensitive POD, EPA identified two new PECO-relevant studies that provided information pertaining to one primary hazard outcome (i.e., reproductive/developmental toxicity) (Gray et al.. 2021; Pan et al.. 2017). These new studies of DIBP are discussed further in Section 3.1.2. Based on information provided in existing assessments of DIBP for developmental and reproductive effects in combination with new information identified by EPA, the Agency focused its non-cancer human health hazard assessment on developmental and reproductive toxicity (Section 3.1). Further, EPA reviewed and supports the conclusions of the systematic review and hazard identification for DIBP published by Yost et al. (2019). EPA did not identify any new literature that would change the conclusions of Yost et al. (2019) pertaining to slight evidence for female reproductive effects and liver effects and indeterminant evidence for kidney effects. Therefore, EPA did not further characterize these non-cancer hazards in this assessment or carry them forward to dose-response assessment in Section 4. Genotoxicity and carcinogenicity data for DIBP are summarized in EPA's Draft Cancer Raman Health Hazard Assessment for Di (2-e thy Ihexyl) Phthalate (DEHP), Dibutyl Phthalate (DBP), Diisobutyl Phthalate (DIBP), Butyl Benzyl Phthalate (BBP) and Dicyclohexyl Phthalate (DCHP) (U.S. EPA. 2025a). Page 16 of 94 ------- 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 PUBLIC RELEASE DRAFT December 2024 2 TOXICOKINETICS 2.1 Oral Route No in vivo studies of experimental animal models are available that have evaluated the absorption, distribution, metabolism, and excretion (ADME) properties of DIBP for the oral exposure route. One intentional human dosing study is available that investigates urinary elimination of DIBP (Koch et al.. 2012). In this study, an individual volunteer (36-year-old male, 87 kg) was administered a single oral dose of 60 |ig/kg deuterium-labelled DIBP (5.001 mg total), and urine samples were collected up to 48 hours following dosing (Koch et al.. 2012). Three urinary metabolites of DIBP were detected: Monoisobutyl phthalate (MIBP); 20H-MIBP; and 30H-MIBP. MTBP was the primary urinary metabolite of DIBP (70 to 71 percent of excreted DIBP over 24 to 48 hours), while 20H-MIBP (approximately 19 percent of excreted DIBP over 24 to 48 hours) and 30H-MIBP (0.7 percent of excreted DIBP over 24 to 48 hours) were minor urinary metabolites. After 24 hours, 90.27 percent of the administered dose was recovered in urine, indicating DIBP is absorbed across the gastrointestinal tract and urine is the primary elimination route. After 48 hours, 90.84 percent was recovered. Peak urinary metabolite concentrations occurred 2.83 hours post-dosing. Urinary elimination half-lives were similar for MIBP (3.9 hours), 20H-MIBP (4.2 hours) and 30H-MIBP (4.1 hours), indicating rapid absorption and urinary elimination. Fecal and biliary excretion were not investigated in this study. MIBP has been measured in human milk in the United States (Hartle et al.. 2018). Korea (Kim et al.. 2020; Kim et al.. 2018; Kim et al.. 2015). Italy (Del Bubba et al.. 2018; Latini et al.. 2009). Germany (Fromme et al.. 2011). Taiwan (Lin et al.. 2011). Switzerland (Schlumpf et al.. 2010). and Sweden (Hogberg et al.. 2008). indicating that absorbed DIBP can partition into human milk. Furthermore, because human biomonitoring data reflects recent aggregate exposure, it cannot quantitatively be attributed to a specific route although it is assumed to predominately come from oral exposure; however, exposure from the dermal and inhalation routes may also contribute. For the draft DIBP risk evaluation, EPA will assume 100 percent oral absorption of DIBP. Notably, other regulatory agencies have also assumed 100 percent oral absorption of DIBP (ECCC/HC. 2020; ECHA. 2017a. b; EC/HC. 2015b; U.S. CPSC. 2014. 2011). 2.2 Inhalation Route No controlled human exposure studies or in vivo animal studies are available that evaluate the ADME properties of DIBP for the inhalation route. EPA will assume 100 percent absorption via inhalation for the draft DIBP risk evaluation. Notably, ECHA (2017a. b) has also assumed 100 percent absorption via the inhalation route for DIBP. 2.3 Dermal Route No in vitro or controlled human exposure studies are available that evaluate the ADME of DIBP for the dermal route. One in vivo ADME study is available that indicates dermally absorbed DIBP is widely distributed to tissues in rats (Elsisi et al.. 1989). Skin on the backs of male Fischer 344 (F344) rats was shaved one hour before DIBP administration (rats with visual signs of abrasions were eliminated from the study). Neat carbon-14 labelled DIBP (14C-DIBP) in an ethanol vehicle (30 to 40 mg/kg) was applied to a circular area of the skin 1.3 centimeters in diameter, which represents a dose of 5 to 8 mg/cm2. Ethanol was allowed to evaporate and then the application site was covered with a perforated circular plastic Page 17 of 94 ------- 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 PUBLIC RELEASE DRAFT December 2024 cup. Rats were then housed in metabolic cages for 7 days during which time urine and feces were collected every 24 hours. Following 7 days of dermal exposure to 14C-DIBP, Elsisi et al. measured low levels of radioactivity associated with 14C-DIBP in adipose tissue (0.11 percent of applied dose), muscle (0.22 percent of applied dose), skin (0.2 percent of applied dose) and other tissues (less than 0.5 percent of applied dose found in the brain, lung, liver, spleen, small intestine, kidney, testis, spinal cord, and blood). Thirty-five percent of the applied dose was recovered from the skin at the application site, while six percent was recovered from the plastic cap. Total recovery of the applied dose was 93 percent. After 24 hours of exposure, approximately 6 percent of the applied dose was recovered in urine, while approximately 1 percent was recovered in feces. After seven days, approximately a total of 51 percent of the applied dose was excreted in urine and feces. As described in Section 2.3 of the Draft Consumer and Indoor Exposure Assessment for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2025b). the rate of transport of DIBP across the dermal barrier is considered flux-limited, rather than delivery limited. The physicochemical properties of DIBP (high molecular weight, large size, and low solubility in water) impede its ability to cross the dermal barrier, limiting the rate of flux independent of the concentration on the skin. Therefore, to estimate dermal exposures to DIBP for workers, consumers and the general population, EPA used a flux-limited dermal absorption approach for liquids and solid articles. Dermal absorption data from the study by Elsisi et al. (1989) was used to estimate a steady-state flux of neat DIBP of 2.43 x 10"2 mg/cm2/hr, which is considered representative for dermal contact with liquids or formulations containing DIBP. No empirical data exists to estimate flux-limited dermal absorption of DIBP from solid articles. Therefore, EPA used a framework based on physical chemical properties of DIBP to estimate flux for solid articles. Briefly, EPA assumes that DIBP first migrates from the solid matrix to a thin layer of moisture on the skin surface. Therefore, absorption of DIBP from solid matrices is considered limited by aqueous solubility and is estimated using an aqueous absorption model, which is described further in Section 2.3.1 of the Draft Consumer and Indoor Exposure Assessment for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2025b). Overall, EPA estimated an 8-hour time weighted average flux value of 1.7 x 10"4 mg/cm2/hr, which is considered a representative value for worker dermal exposures to solids or articles containing DIBP. For consumers, dermal flux values range from 1.51 x 10"4 to 9.62 x 10"4 mg/cm2/hr depending on model input parameters used in the dermal models for different consumer exposure scenarios for solids. For further information pertaining to EPA's dermal approach, see Section 2.3 of the Draft Consumer and Indoor Exposure Assessment for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2025b) and Appendix C.2.1 of the Draft Environmental Release and Occupational Exposure Assessment for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2025c). Page 18 of 94 ------- 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 PUBLIC RELEASE DRAFT December 2024 3 NON-CANCER HAZARD IDENTIFICATION 3.1 Effects on the Developing Male Reproductive System As discussed in Section 1.2, the effects on the developing male reproductive system has consistently been identified as the most sensitive effects associated with oral exposure to DIBP in experimental animal models in existing assessments of DIBP (ECCC/HC. 2020; ECHA. 2017a. b; NASEM. 2017; EC/HC. 2015b; U.S. CPSC. 2014; ECHA. 2012a. b; U.S. CPSC. 2011; NICNAS. 2008a) as well as prior systematic reviews (Yost et al.. 2019). EPA identified no new information through systematic review that would change this conclusion. Therefore, EPA focused its non-cancer hazard characterization on the developing male reproductive system. Evidence from epidemiological and laboratory animal studies for developmental and reproductive outcomes is summarized in Sections 3.1.1 and 3.1.2, respectively. 3.1.1 Summary of Available Epidemiological Studies 3.1.1.1 Previous Epidemiology Assessment (Conducted in 2019 or Earlier) EPA reviewed and summarized conclusions from previous assessments conducted by Health Canada (2018b) and NASEM (2017). as well as systematic review articles by Radke et al. (2019b; 2018). that investigated the association between exposure to DIBP and its metabolites and male and female developmental and reproductive outcomes. As can be seen from Table 3-1, epidemiologic assessments by Health Canada (2018b). NASEM (2017). and systematic review articles by Radke et al. (2019b; 2018) varied in scope and considered different developmental and reproductive outcomes. Further, these assessments used different approaches to evaluate epidemiologic studies for data quality and risk of bias in determining the level of confidence in the association between phthalate exposure and evaluated health outcomes (Table 3-1). Section 3.1.1.1.1, Section 3.1.1.1.2, and Section 3.1.1.1.3 provide further details on previous assessments of DIBP by Health Canada (2018b). Radke et al., (2019b; 2018) and NASEM (2017). respectively, including conclusions related to exposure to DIBP and health outcomes. Additionally, EPA also evaluated epidemiologic studies published after the Health Canada (2018b) assessment as part of its literature search (i.e., published between 2018 and 2019) to determine if newer epidemiologic studies would change the conclusions of existing epidemiologic assessments or provide useful information for evaluating exposure-response relationship (Section 3.1.1.2). Table 3-1. Summary of Scope and Methods Used in Previous Assessments to Evaluate the Association Between DIBP Exposure and Male Reproductive Outcomes Previous Assessment Outcomes Evaluated Method Used for Study Quality Evaluation Health Canada (2018b) Hormonal effects: Sex hormone levels (e.g., testosterone) Growth & Development: AGD Birth measures Male infant genitalia (e.g., hypospadias/cryptorchidism) Placental development and gene expression Preterm birth and gestational age Downs and Black (Downs and Black, 1998) Page 19 of 94 ------- 617 618 619 620 621 622 623 624 625 626 627 628 629 630 PUBLIC RELEASE DRAFT December 2024 Previous Assessment Outcomes Evaluated Method Used for Study Quality Evaluation Postnatal growth DNA methylation Reproductive: Altered male puberty Gynecomastia Changes in semen parameters Sexual dysfunction (males) Sex ratio Radke et al. (2018) AGD Hypospadias/cryptorchidism Pubertal development Semen parameters Time to pregnancy Testosterone Timing of pubertal development Approach included study sensitivity as well as risk of bias assessment consistent with the study evaluation methods described in (U.S. EPA. 2022) Radke et al. (2019b) Pubertal development Time to pregnancy (Fecundity) Preterm birth Spontaneous abortion ROBINS-I (Sterne et al., 2016) NASEM (2017) AGD Hypospadias (incidence, prevalence, and severity/grade) Testosterone concentrations (measured at gestation or delivery) OHAT (based on GRADE) (TSTTP. 2015) Abbreviations: AGD = anogenital distance; ROBINS-I= Risk of Bias in Non-randomized Studies of Interventions; OHAT = National Toxicology Program's Office of Health Assessment and Translation; GRADE = Grading of Recommendations, Assessment, Development and Evaluation. 3.1.1.1.1 Health Canada (2018b) Health Canada evaluated studies that looked at individual phthalates (or their metabolites) and health outcomes and did not consider studies that only looked at summed exposure to multiple phthalates due to the challenging nature of interpreting results for the sum of several phthalates. The outcomes that were evaluated are listed in Table 3-1. To evaluate the quality of individual studies and risk of bias, Health Canada (2018b) used the Downs and Black evaluation criteria (Downs and Black. 1998). which is based on the quality of the epidemiology studies and the strength and consistency of the relationship between a phthalate and each health outcome. The level of evidence for association of a phthalate and each health outcome was established based on the quality of the epidemiology studies and the strength and consistency of the association. Health Canada (2018b) evaluated several studies that investigated the association between urinary metabolites of DIBP and several developmental and reproductive outcomes. Health Canada concluded Page 20 of 94 ------- 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 PUBLIC RELEASE DRAFT December 2024 that there was some limited evidence of association1 for DIBP and several outcomes, including changes in serum levels of sex hormones (e.g., follicle stimulating hormone, luteinizing hormone, testosterone), increased sperm DNA damage and apoptosis, and changes in infant sex ratio at birth. For other health outcomes, Health Canada concluded there was inadequate evidence of association1 (i.e., for changes in thyroid and other miscellaneous hormones, changes in semen parameters, pregnancy complication and loss, sexual dysfunction in males and females, and age at menopause). In addition, there was no evidence of association1 based on lack of changes in AGD, birth weight, birth length, head circumference, femur length, preterm birth, gestational age, altered male puberty, gynecomastia, time to pregnancy, uterine leiomyoma, and polycystic ovary syndrome, or that the level of evidence of association could not be established due to limitations in the available studies (i.e., for changes in placental development, postnatal growth, altered female puberty, altered fertility). 3.1.1.1.2 Radke et al. (2019b: 2018) Radke et al. conducted systematic reviews of male (Radke et al.. 2018) and female (Radke et al.. 2019b) developmental and reproductive outcomes. These systematic review articles are considered herein. Radke et al. (2018) evaluated the associations between DIBP or its metabolite (MIBP) and male reproductive outcomes, including AGD and hypospadias/cryptorchidism following in utero exposures; pubertal development following in utero or childhood exposures, and semen parameters, time to pregnancy (following male exposure), and testosterone following adult exposures. Male reproductive outcomes and level of confidence in the associations is listed in Table 3-2. Data quality evaluation criteria and methodology used by Radke et al. (2018) were qualitatively similar to those used by NASEM (2017) (i.e., OHAT methods) and Health Canada (2018b). Similar to NASEM (2017) and Health Canada (2018b). most studies reviewed by Radke et al. (2018) relied on phthalate metabolite biomarkers for exposure evaluation. Therefore, different criteria were developed for short- chain (DIBP, DEP, DBP, BBP) and long-chain (DEHP, DINP) phthalates due to better reliability of single measures for short-chain phthalates. Radke et al. (2018) used data quality evaluations to inform overall study confidence classifications, and ultimately evidence conclusions of "Robust," "Moderate," "Slight," "Indeterminate," or "Compelling evidence of no effect." "Robust" and "Moderate" evidence of an association is distinguished by the amount and caliber of data that can be used to rule out other possible causes for the findings. "Slight" and "Indeterminate" describe evidence for which uncertainties prevent drawing a causal conclusion in either direction. Table 3-2. Summary of Epidemiologic Evidence of Male Reproductive Effects Associated with Exposure to DIBP Timing of Exposure Outcome Level of Confidence in Association In utero Anogenital distance Slight Hypospadias/cryptorchidism Slight In utero or childhood Pubertal development Indeterminate 1 Health Canada defines limited evidence as "evidence is suggestive of an association between exposure to a phthalate or its metabolite and a health outcome; however, chance, bias or confounding could not be ruled out with reasonable confidence." Health Canada defines inadequate evidence as "the available studies are of insufficient quality, consistency or statistical power to permit a conclusion regarding the presence or absence of an association." Health Canada defines no evidence of association as "the available studies are mutually consistent in not showing an association between the phthalate of interest and the health outcome measured." Page 21 of 94 ------- 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 PUBLIC RELEASE DRAFT December 2024 Timing of Exposure Outcome Level of Confidence in Association Adult Semen parameters Slight Time to pregnancy Slight Testosterone Moderate Male Reproductive Outcomes Overall Moderate "Table from Figure 3 in Radke et al. (2018). Similar to the conclusions of Health Canada, Radke et al. (2019b; 2018) found moderate evidence of an association2 between exposure to DIBP and decreased testosterone levels in males, while evidence of an association between exposure to DIBP and other male and female reproductive outcomes was found to the slight {i.e., for decreased AGD, hypospadias and/or cryptorchidism, changes in semen parameters, time to pregnancy [based on male exposure to DIBP]) or indeterminant {i.e., for male and female pubertal development, spontaneous abortion, time to pregnancy [based on female exposure to DIBP]). 3.1.1.1.3 NASEM (2017) NASEM (2017) included a systematic review of the epidemiological evidence of the associations between exposure to various phthalates or their monoester or oxidative metabolites including DIBP, and the following male reproductive outcomes 1) AGD measurements, 2) incidence, prevalence, and severity/grade of hypospadias, and 3) testosterone concentrations measured at gestation or delivery. In contrast to Health Canada (2018b). and Radke et al. (2018). NASEM (2017) relied on methodological guidance from the National Toxicology Program's Office of Health Assessment and Translation (OHAT) to assign confidence ratings and determine the certainty of the evidence to ultimately draw hazard conclusions (NTP. 2015). NASEM (2017) concluded that there was inadequate evidence to establish an association between prenatal exposure to DIBP and hypospadias due to the limited number of studies and dissimilar matrices utilized to evaluate them (urine and amniotic fluid). NASEM also concluded that there is inadequate evidence to determine whether fetal exposure to DIBP is associated with a decrease in fetal testosterone in males, given the various matrices used to measure testosterone (amniotic fluid, maternal serum, or cord blood), the differences in timing of exposure (during pregnancy or at delivery), and the limited number of studies. This conclusion is slightly different from those of Health Canada (2018b) and Radke et al. (2019b; 2018). because they are looking at different life stages, each of which found limited and moderate evidence, respectively, of an association between exposure to DIBP and decreased testosterone levels in males. Radke et al. (2018) and Health Canada (2018b) considered the association between exposure to DIBP and testosterone in children and adults while NASEM looked at fetal life stages. NASEM also concluded that there was an inadequate level of evidence to determine an association between DIBP (MIBP) and AGD, although there was moderate confidence in the evidence of association based on three prospective cohort studies. However, NASEM also conducted a meta-analysis 2 Radke et al. (2019b: 2018) define Robust and Moderate evidence descriptors as "evidence that supports a hazard, differentiated by the quantity and quality of information available to rule out alternative explanations for the results." Slight and indeterminant evidence descriptors are defined as "evidence that could support a hazard or could support the absence of a hazard. These categories are generally limited in terms of quantity or confidence level of studies and serve to encourage additional research across the exposure range experienced by humans." Page 22 of 94 ------- 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 PUBLIC RELEASE DRAFT December 2024 of three studies (Jensen et al.. 2016; Swan et al.. 2015; Swan. 2008) and found that the available studies do not support the association between DIBP exposure and decreased AGD (% change [95% CI] = -2.23 [-5.15, 0.70] [p = 0.13]). The AGD effect estimates in the NASEM (2017) meta-analyses are slope estimates based on the assumption that exposure and effect have a monotonic dose-response relationship. This conclusion is similar to the conclusions of Radke et al. (2018). who found slight evidence of an association between DIBP exposure and decreased AGD. 3.1.1.1.4 Summary of the Existing Assessments of Male Reproductive Effects Each of the three assessments discussed above provided qualitative support as part of the weight of scientific evidence for the association between DIBP exposure and male reproductive outcomes. The existing assessments and review article came to similar conclusion on the effect of exposure to DIBP and male reproductive outcomes. Radke et al. (2018) concluded that there was a slight level of confidence in the association between exposure to DIBP and AGD, while Health Canada (2018b) and NASEM (2017) found inadequate evidence of an association. Further, Radke et al. (2018) found that there was moderate evidence for the association between testosterone and exposure to DIBP, while Health Canada (2018b) found that total testosterone (TT) and free testosterone (fT) had negative associations (i.e., increase exposure to DIBP with decrease testosterone) in peri pubertal or adolescent boys (6-12, 8-14 or 12-20 years) per IQR increase with exposure to DIBP and its metabolite MIBP, and negative associations for total testosterone in adult males 17 to 52 years. Radke et al. (2018). also found a slight level of confidence in the association between exposure to DIBP and cryptorchidism/hypospadias, but this association was not consistent with the findings of Health Canada (2018b) or NASEM (2017). The scope and purpose of the assessments by Health Canada (2018b). systematic review articles by Radke et al. (2018). and the report by NASEM (2017) differ and may be related to differences in quality evaluation and confidence conclusions drawn. Health Canada (2018b) was the most comprehensive review, and considered prenatal and perinatal exposures, as well as peripubertal exposures and multiple different outcomes. NASEM (2017) evaluated fewer epidemiological outcomes than Health Canada (2018b) and systematic review articles by Radke et al. (2018). but also conducted a second systematic review of the animal literature, which will be discussed further in Section 4.The results of the animal and epidemiological systematic reviews were considered together by NASEM (2017) to draw hazard conclusions. Each of the existing assessments covered above considered a different number of epidemiological outcomes and used different data quality evaluation methods for risk of bias. Despite these differences, and regardless of the limitations of the epidemiological data, each assessment provides qualitative support as part of the weight of scientific evidence. 3.1.1.2 EPA Summary of New Studies (2018 to 2019) EPA also evaluated epidemiologic studies published after the Health Canada (2018b) assessment as part of its literature search (i.e., published between 2018 and 2019). EPA identified 40 new epidemiologic studies (24 developmental and 16 reproductive) that evaluated the association between urinary DIBP and its metabolite (MIBP) and reproductive and developmental outcomes. Studies reporting a significant association are discussed further below. Further information (i.e., data quality evaluations and data extractions) on the new studies identified by EPA can be found in: Draft Data Quality Evaluation Information for Raman Health Hazard Epidemiology for DiisobutylPhthalate (DIBP) (U.S. EPA. 2024c). and Draft Data Extraction Information for Environmental Hazard and Human Health Hazard Animal Toxicology and Epidemiology for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024a). Page 23 of 94 ------- 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 PUBLIC RELEASE DRAFT December 2024 In text below, EPA discussed the evaluation of the new studies by outcome that contribute to the weight of scientific evidence. Developmental Outcomes for Males Twenty-four studies were evaluated for the association between DIBP and developmental outcomes including birth measures, size trajectory, fetal loss, pubertal development, and gestational duration. Of those studies, 1 was high confidence, 17 were of medium confidence and 6 were of low confidence. There were only four studies with significant results, one high confidence study (Harlev et al.. 2019). one medium confidence study (Burns et al.. 2022) and two low confidence study (Durmaz et al.. 2018; Yang et al.. 2018). The remaining 20 studies evaluating developmental outcomes in males did not show any significant results and are not discussed further in this document. However, further information for these 20 studies can be found in the Draft Data Quality Evaluation Information for Raman Health Hazard Epidemiology for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024c) and Draft Data Extraction Information for Environmental Hazard and Human Health Hazard Animal Toxicology and Epidemiology for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024a). In the evaluation of pubertal development and DIBP exposure, one high confidence (Harlev et al.. 2019) and one medium confidence (Burns et al.. 2022) study examined the relationship between exposure to MIBP and pubertal onset and both reported increasing developmental delay in association with MIBP exposure. The high confidence study (Harlev et al.. 2019) examined the relationship between prenatal MIBP exposure and pubertal timing (thelarche, pubarche, menarche, gonadarche) among 159 boys and 179 girls enrolled in the CHAMACOS Study and found significant positive association between prenatal MIBP exposure (measured via maternal urinary MIBP) and age at thelarche among girls in exposure quartile 2 vs. quartile 1 [6.5 month mean shift in age at thelarche, 95% CI (1.0, 12.3)]. However, no significant associations were found for Q2 or Q4 vs. Ql, and no significant associations were found for other pubertal timing outcomes among girls or boys. The medium confidence study (Burns et al.. 2022) examined the association between prepubertal MIBP exposure (assessed via urinary MIBP concentrations) in relation to age at pubertal onset among 304 boys enrolled in the Russia Children's Study. Puberal onset outcomes were defined as testicular volume greater than 3 mL, Tanner Genitalia Stage greater than or equal to 2, and Tanner Pubarche Stage greater than or equal to 2. Significant positive associations were found for all three outcomes. Significant mean delays in testicular growth were found across all quartiles, as compared to Ql [Q2 vs Ql: 8.5 months, 95% CI (3.7, 13.5); Q3 vs Ql: 6.4, 95% CI (1.1, 11.7); Q4 vsQl: 5.7(0.2, 11.1). Significant mean delays reaching a Tanner Genital Stage > 2 were found for Q2 and Q3 vs Ql [Q2 vs Ql: 6.4 months, 95% CI (0.2, 12.6); Q3 vs Ql: 7.2 (0.5, 13.8)] but not for Q4 vs Ql. Significant mean delays in reaching Pubarche Stage > 2c were found for Q3 and Q4 vs Ql [Q3 vs Ql: 10.2 months, 95% CI (2.9, 17.5); Q4 vs Ql: 12.8, 95% CI (5.3, 20.3)], but not for Q2 vs Ql. Trend tests were only significant for increasing quartiles of MIBP exposure for Pubarche Stage greater than or equal to 2c. Other Developmental Outcomes Other developmental outcomes such as body mass index (BMI) trajectories were also assessed. One medium confidence study (Heggeseth et al.. 2019) and two low confidence study (Durmaz et al.. 2018; Yang et al.. 2018) examined BMI trajectories in relation to MIBP exposure. Heggeseth et al. (2019) (medium confidence) used growth mixture models and functional principal components analysis to assess whether prenatal phthalate exposure helped explain variation in size trajectory among 162 boys and 173 girls enrolled in Center for the Health Assessment of Mothers and Children of Salinas (CHAMACOS) Study. The study, although no effect estimates were provided, found that urinary concentrations of MIBP at greater than or equal to 1.7 ng/mL explains variation in BMI in boys. One low confidence study (Yang et al.. 2018) examining BMI trajectories in relation to MIBP exposure Page 24 of 94 ------- 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 PUBLIC RELEASE DRAFT December 2024 among 239 children from Mexico City enrolled in the Early Life in Mexico to Environmental Toxicants (ELEMENT) found, without reporting effect estimates, that exposure to the first tertile of MIBP predicted the lowest BMI trajectory in infancy and early childhood but crossed over to predict the highest BMI by age 14. The other low confidence study (Durmaz et al.. 2018) examined the relationship between MIBP exposure and BMI and weight in 29 girls between the ages of 4 years and 8 years with premature thelarche, from Antalya City, Turkey and found significant positive associations for both weight (Spearman correlation coefficient: 0.742, p< 0.01) and BMI (Spearman correlation coefficient: 0.574, p = 0.002). Reproductive Outcomes for Males Five medium confidence studies evaluated the association between DIBP exposure and male reproductive outcomes; however, only one (Wenzel et al.. 2018) found significant results. Epidemiologic literature that identified male reproductive effects associated with DIBP exposure found one medium confidence study (Wenzel et al.. 2018) of infants in Charleston, South Carolina that reported a significant positive association between maternal urinary concentrations of MIBP and anoscrotal distance in white infants only [Beta (95% CI) per unit increase in MIBP for anoscrotal distance = 1.68 (0.09, 3.27)]. No other significant results were reported for other anthropometric measurements or when results were not stratified by race/ethnicity. Studies on other male reproductive effects such as anthropometric measures of male reproductive organs, sperm parameters, prostate and male reproductive hormones found no significant associations. However, further information for these 5 studies can be found in the Draft Data Quality Evaluation Information for Raman Health Hazard Epidemiology for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024c) and Draft Data Extraction Information for Environmental Hazard and Human Health Hazard Animal Toxicology and Epidemiology for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024a). Reproductive Outcomes for Females Eleven studies (1 high confidence, 9 medium confidence, 1 uninformative) evaluated the association between DIBP exposure and female reproductive outcomes. Of those studies, two medium confidence studies (Chin et al.. 2019: Machtinger et al.. 2018) and one low confidence studv(Durmaz et al.. 2018) had significant results. The remaining eight studies evaluating reproductive outcomes in females did not show any significant results and are not discussed further in this document. However, further information for these 9 studies can be found in the Draft Data Quality Evaluation Information for Human Health Hazard Epidemiology for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024c) and Draft Data Extraction Information for Environmental Hazard and Human Health Hazard Animal Toxicology and Epidemiology for Diisobutyl Phthalate (DIBP) (U.S. EPA. 2024a). Female reproductive effects associated with DIBP exposure were identified in two medium confidence studies (Chin et al.. 2019: Machtinger et al.. 2018) and one low confidence study (Durmaz et al.. 2018). Chin et al. (2019) (medium confidence study) investigated North Carolina women without known fertility issues and reported significantly increased odds of a shorter time between ovulation and implantation [OR (95% CI) for early implantation = 2.09 (95% CI=1.18, 3.69)]. The other medium confidence study (Machtinger et al.. 2018) examined women undergoing in vitro fertilization (IVF) in Israel and reported a significantly reduced mean number of total oocytes in tertile 2 compared to tertile 1 of urinary MIBP in women undergoing a fresh IVF cycle [Mean difference (95%) CI for tertile 2 = 8.7 (7.9, 9.6)]. This study further reported a significantly reduced mean number of mature oocytes in both tertiles 2 and 3 compared to tertile 1 of MIBP exposure [Mean difference (95% CI) for tertile 2 = 6.7 (6.0, 7.5); Mean (95% CI) for tertile 3 = 8.0 (7.2, 8.8)]. The mean number of fertilized oocytes was also significantly reduced in tertile 2 compared to tertile 1 of MIBP exposure [Mean difference (95% CI) for tertile 2 = 4.6 (4.0, 5.3)]. Women with higher MIBP exposure also had a significantly reduced mean Page 25 of 94 ------- 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 PUBLIC RELEASE DRAFT December 2024 number of top-quality embryos. This study also reported significantly reduced mean number of top- quality embryos [Mean difference (95% CI) for tertile 2 = 2.0 (1.7, 2.5); Mean (95% CI) for tertile 3 = 2.2 (1.8, 2.7)]. The low confidence study (Durmaz et al.. 2018) conducted in Turkey reported a significant unadjusted positive correlation between urinary MIBP concentrations and basal follicle stimulating hormone (FSH) in girls with premature thelarche [Spearman correlation coefficient between MIBP and basal FSH = 0.323, p-value = 0.045], Other studies that examined female reproductive measures, such as anthropometric measures of female reproductive organs or fibroids, and association with DIBP exposure found no significant association. Conclusion In conclusion, Health Canada (2018b) and NASEM (2017) found inadequate evidence of association between DIBP and AGD while systematic review articles published by Radke et al. (2018) found slight evidence of association with AGD. Moreover, new studies identified by EPA from 2018 to 2019 do not alter the previous conclusions from Health Canada (2018b) and NASEM (2017). and systematic review articles published by Radke et al. (2018). Although there is slight evidence of an association between DIBP and AGD. the results for testosterone were measured at different life stages (i.e., fetal/infants to adults) and causality could not be established, thus the overall evidence does not support an association between DIBP and AGD or testosterone. Furthermore, EPA preliminarily concludes that the existing epidemiological studies do not support quantitative exposure-response assessment due to uncertainty associated with exposure characterization of individual phthalates, including source or exposure and timing of exposure as well as co-exposure confounding with other phthalates, discussed in Section 1.1. The epidemiological studies provide qualitative support as part of the weight of scientific evidence. 3.1.2 Summary of Laboratory Animals Studies EPA identified 13 oral exposure studies (11 of rats, 2 of mice) that have investigated the effects of DIBP on the developing male reproductive system (Gray et al.. 2021; Pan et al.. 2017; Saillenfait et al.. 2017; Wang et al.. 2017; Sedha et al.. 2015; Furr et al.. 2014; Hannas et al.. 2012; Hannas et al.. 2011; Howdeshell et al.. 2008; Saillenfait et al.. 2008; BASF. 2007; Borch et al.. 2006; Saillenfait et al.. 2006). No studies evaluating the developmental and/or reproductive toxicity of DIBP are available for the inhalation or dermal exposure routes. Available oral exposure studies of DIBP evaluating developmental and reproductive outcomes are summarized in Table 3-3. Most of the available studies evaluate effects on the developing male reproductive system consistent with a disruption of androgen action following gestational, perinatal, or pre-pubertal oral exposures to DIBP. However, several studies are available that evaluate other developmental outcomes (e.g., post-implantation loss, resorptions, fetal body weight, skeletal variations, etc.). Effects on the developing male reproductive system and other developmental and reproductive outcomes are discussed in Sections 3.1.2.1 and 3.1.2.2, respectively. Page 26 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Table 3-3. Summary of Studies of DII P Evaluating Developmental and Reproductive Outcomes Reference Brief Study Description NOAEL/ LOAEL (mg/kg-day) Effect at LOAEL Remarks (Howdeshell et al.. 2008) Pregnant SD rats (5-8 dams/dose) gavaged with 0, 100, 300, 600, 900 mg/kg- day DIBP onGDs8-18 100/300 I ex vivo testicular testosterone production (40%) on GD 18 Maternal Effects - i Maternal body weight on GD 18 (>600 mg/kg-day) and weight gain (900) Developmental Effects -1 fetal morality (900 mg/kg-day) - i # of live fetuses (900 mg/kg-day) -1 total resorptions (900 mg/kg-day) (Hannas et al.. 2011) Pregnant SD rats (3 dams/dose) gavaged with 0, 100, 300, 600, 900 mg/kg- day DIBP on GDs 14-18 100/300 I ex vivo fetal testicular testosterone production (56%) and j expression of steroidogenic genes in fetal testes on GD 18 Maternal Effects - None Developmental Effects -1 ex vivo fetal testicular testosterone production on GD 18 (>300 mg/kg-day) -1 Fetal testis mRNA levels of StAR (>300 mg/kg-day) and Cvplla on GD 18 (>100 mg/kg-day) Unaffected Outcomes - Maternal mortality, clinical signs of toxicity, maternal body weight, litter size (Saillenfait et al.. 2008) Pregnant SD rats (11-14 dams/dose) gavaged with 0, 125, 250, 500, 625 mg/kg- day DIBP on GDs 12-21 None/125 t Testicular pathology (degeneration of seminiferous tubules) and oligo-/azoospennia in epididymis) Maternal Effects - None Developmental Effects -1 Male AGD (absolute) on PND 1 (>250 mg/kg-day) -1 Male NR on PNDs 12-14 and at necropsy on PNW 11-12 or PNW 16-17 (>250 mg/kg-day) - i Male pup weight on PND 1 and PND 21 (625 mg/kg-day) -1 hypospadias (>500 mg/kg-day), cleft prepuce (625 mg/kg-day), exposed os penis (>500 mg/kg-day), non-scrotal testes at necropsy (PNW 11-12 or 16-17) (>500 mg/kg-day) - Delayed PPS (>500 mg/kg-day) -1 male offspring body weight on PNW 11-12 and PNW 16-17 (>500 mg/kg- day) - i absolute prostate weight on PNW 11-12 (>250 mg/kg-day) and 16-17 (>500 mg/kg-day); j absolute testis, epididymis, and SV weight on PND 11-12 and 16-17 (>500 mg/kg-day) Unaffected Outcomes Page 27 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Reference Brief Study Description NOAEL/ LOAEL (mg/kg-day) Effect at LOAEL Remarks - Maternal weight gain (GD 0-12, GD 12-21, PND 1-21); post-implantation loss; % live pups; pup survival (PND 1-4, PND 4-21); female offspring body weight on PND 4, 7, 14, 21 (Saillenfait et al.. 2006) Pregnant SD rats (20-22 pregnant rats/dose) gavaged with 0, 250, 500, 750, 1000 mg/kg-day DIBP on GDs 6- 20 250/500 i fetal body weight (7%) (both sexes); t incidence of undescended testes (unilateral or bilateral) and degree of trans- abdominal testicular migration. J, Maternal weight gain Maternal Effects -1 Maternal weight gain on GD 6-9 and GD 15-18 (>500 mg/kg-day) Developmental Effects -1 % Resorptions per litter (>750 mg/kg-day) -1 % Post-implantation losses per litter (>750 mg/kg-day) - i Number of live fetuses per litter (>750 mg/kg-day) - i Fetal body weight (|7%) (both sexes) (>500 mg/kg-day) -1 Total number of fetuses with external, visceral, and skeletal malformations (>750 mg/kg-day) -1 Total number of litters with visceral and skeletal malformations (>750 mg/kg-day) -1 incidence of visceral and skeletal variations, including ectopic testis (>750 mg/kg-day), increased degree of trans-abdominal testicular migration (>500) Unaffected Outcomes - Maternal mortality; maternal food consumption; overall maternal weight gain corrected for gravid uterine weight; % dead fetus per litter; sex ratio (BASF. 2007) Pregnant Wistar rats (22- 23/dose) administered diets containing 0, 1000, 4000, 11,000 ppm DIBP on GDs 6- 20 (equivalent to 88, 363, 942 mg/kg-day) Adhered to OECD TG 414; GLP-compliant 363/942 i maternal food consumption, J, maternal body weight gain, J, fetal body weight (5%); skeletal variations Maternal Effects - i Maternal food consumption (approximately 5% below control across GD 6- 20) (942 mg/kg-day) - i Maternal body weight gain (approximately 11% below control across GD 6- 20) (942 mg/kg-day) Developmental Effects - i Fetal (both sexes) body weight (approximately 5% below control) (942 mg/kg-day) -1 skeletal variations, including incomplete ossification of sternebra and unilateral ossification of sternebra (942 mg/kg-day) Unaffected Outcomes - Maternal mortality; no clinical signs; post-implantation loss; resorptions; # of viable fetuses; sex ratio; external or visceral malformations or variations Page 28 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Reference Brief Study Description NOAEL/ LOAEL (mg/kg-day) Effect at LOAEL Remarks (Saillenfait et al.. 2017) Pregnant SD rats (15-20 /dose) gavaged with 0 or 250 mg/kg-day DIBP on GDs 13- 19 None/250 I AGD, I testicular testosterone (45%) and, androstenedione (27%) production; altered mRNA expression of steroidogenesis genes in the testes Maternal Effects - None Developmental Effects - i AGD (normalized to cubic root of body weight) - i (27-45%) ex vivo testis testosterone and androstenedione production i gene expression in cholesterol and steroid synthesis in fetal testes (Hmg- CoAR, Hmg-CoAS, SR-B1, StAR, P450cl7, 170-HSD) Unaffected Outcomes - Dam body weight gain; gravid uterine weight; post-implantation loss; # live fetuses per litter; sex ratio; fetal body weight (Wans et al.. 2017) Pregnant ICR Mice (15-18 offspring/dose) fed diets containing 0 or 2.8 g DIBP/kg diet (dry weight) (equivalent to 450 mg/kg- day) from GDs 0-21 (designated TC) or from GDs 0 to PND 21 (designated TT) None/450 i absolute testes weight on PND 21; J, serum and testes testosterone; j expression of steroidogenic genes in testes; |sperm concentration and motility on PND 80 Maternal Effects - None Developmental Effects - i absolute testes weight on PND 21 (TT group only) - i serum and testes testosterone in PND 21 males (TC and TT groups) - i serum and testes testosterone in PND 80 males (TT group only) - i mRNA and protein expression of steroidogenic genes in testes of PND 21 and PND 80 males (e.g., Cypl 7a 1) (TC and TT groups) - i sperm concentration and motility for PND 80 males (TT group only) Unaffected Outcomes - Maternal weight gain; litter size; fetal viability; PND 21 male offspring body weight; offspring liver weight; AGD (Hannas et al.. 2012) Pregnant SD rats (3/dose) gavaged with 0 or 500 mg/kg-day DIBP on GDs 14- 18 None/500 I ex vivo fetal testicular testosterone production (-25%) on GD 18 Unaffected Outcomes - Maternal body weight gain maternal liver weight, # of live fetuses. Pregnant SD rats gavaged with 0, 100, 300, 600, 900 mg/kg-day DIBP on GDs 14- 18 100/300 (LOEL) i fetal testicular mRNA levels of steroidogenic genes -1 mRNA expression levels of StAR, Cvpllal, Hsd3b, Cvpl7al, Scarbl, InsB, Cypl lb 1 (>300) (Borch et al.. 2006) Pregnant Wistar Rats (6/dose) gavaged with 0 or None/600 I ex vivo testes testosterone production Maternal Effects - None Page 29 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Reference Brief Study Description NOAEL/ LOAEL (mg/kg-day) Effect at LOAEL Remarks 600 mg/kg-day DIBP on GDs 7-19 or GDs 7-20/21 (96%), | AGD, t testicular histopathology Developmental Effects - i Testes testosterone content on GD20/21 (effect on GD 19 not statistically significant) -1 ex vivo testis testosterone production on GD 20/21, but not GD 19 -1 absolute AGD on GD 19 and GD 20/21; J, AGD (normalized to cubic root of body weight) on GD 20/21 - i fetal body weight on GD 19 -1 testicular pathology (Leydig cell clusters on GD 19 and GD 20/21), Sertoli cell vacuolization MNGs, central localization of gonocytes on GD 20/21) -1 immunohistochemistry staining for StAR and P450scc in Leydig cells Unaffected Outcomes - Maternal weight gain during pregnancy; litter size; fetal viability; number of resorptions (Furr et al.. 2014)° Pregnant SD rats (3-5/group) gavaged 0 or 750 mg/kg-day DIBP on GDs 14-18 (Block 2) None/750 I ex vivo fetal testicular testosterone production (81%) on GD 18 Unaffected Outcomes - Fetal viability on GDI8 - Dam body weight gain Pregnant SD rats (3-4/group) gavaged with 0 or 500 mg/kg-day DIBP on GDs 14- 18 (Block 14) None/500 I ex vivo fetal testicular testosterone production (70%) on GD 18 Unaffected Outcomes - Fetal viability on GDI8 - Dam body weight gain Pregnant SD rats (2-4/group) gavaged with 0 or 200 mg/kg-day DIBP on GDs 14- 18 (Block 30) None/200 I ex vivo fetal testicular testosterone production (47%) on GD 18 Unaffected Outcomes - Fetal viability on GDI8 - Dam body weight gain (Sedha et al.. 2015) UtcrotroDhic Assav Young female Wistar rats (20 days old) (>6 mice/dose) gavaged 0, 250, or 1250 mg/kg-day DIBP for 3 days None/250 i Body weight gain - i Body weight gain (>250 mg/kg-day) - Lack of effect on uterus and ovary wet weight indicate DIBP lacks estrogenic potential Unaffected Outcomes - No clinical signs; uterus and ovary wet weight Pubertal Assav Young female Wistar rats (20 days old) (>6 mice/dose) None/250 i Body weight gain - i Body weight gain (>250 mg/kg-day) Page 30 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Reference Brief Study Description NOAEL/ LOAEL (mg/kg-day) Effect at LOAEL Remarks gavaged 0, 250, or 1250 DIBP for 20 days (PND 21- 41) - Lack of effect on reproductive organ weight and vaginal opening indicate DIBP lacks estrogenic potential Unaffected Outcomes - Absolute and relative uterus, ovary, and vagina weight; vaginal opening New Studies of DIBP Since Yost et al. (2019) (Pan et al.. 2017) Young (6-8 week old) male ICR mice (20/dose) fed diets containing 0 or 2.8 g DIBP/kg chow (equivalent received dose of 450 mg/kg- day) for 28 days None/450 Sperm effects, [ serum & testes testosterone, [ mRNA & protein levels of steroidogenesis genes - i Epididymal sperm concentration sperm motility, and progressiveness (450 mg/kg-day) -1 Sperm malformation (450 mg/kg-day) -j Serum and testis testosterone, j serum follicle stimulating hormone levels (450 mg/kg-day) - i mRNA and protein levels of steroidogenic genes in testes (e.g., P450cc, StAR, 3fi-hsd) Unaffected Outcomes - Body weight gain; food intake; absolute and relative testes and epididymis weight; serum levels of estradiol and luteinizing hormone (Gray et al.. 2021)° Pregnant SD rats (3-4 dams/dose) were gavaged with 0, 100, 300, 600, or 900 mg/kg-day DIBP on GDs 14- 18 100/300 I ex vivo testicular testosterone production (34%) on GD 18 -1 ex vivo testicular testosterone production on GD18; Block 67 (>300 mg/kg- day) - i mRNA expression of Phase I metabolism genes (e.g., Cypl lb 1. Cypl lal, Cypl7al, ALDH2) (900 mg/kg-day) - i mRNA expression of lipid signaling and cholesterol metabolism gene (>900 mg/kg-day) Unaffected Outcomes - Maternal liver weight (Block 19) Abbreviations: [ = statistically significant decrease; t = statistically significant increase; NOAEL = No observed adverse effect level; LOAEL = Lowest observed adverse effect level; GD = Gestational Day; PND = Postnatal Day AGD = Anogenital distance; GLP = Good Laboratory Practice; MNG = multinucleated gonocytes; NR = Nipple Retention; PPS = preputial separation; SD = Sprague Dawley; SV = Seminal Vesicles; TT = pups exposed both prenatally and postnatally; TC = pups exposed prenatally only "These studies were conducted by EPA's Office of Research and Development (ORD). 882 Page 31 of 94 ------- 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 PUBLIC RELEASE DRAFT December 2024 3.1.2.1 Developing Male Reproductive System EPA previously developed a weight of scientific evidence analysis and concluded that oral exposure to DIBP can induce effects on the developing male reproductive system consistent with a disruption of androgen action (see EP A's Draft Proposed Approach for Cumulative Risk Assessment of High-Priority and a Manufacturer-Requested Phthalate under the Toxic Substances Control Act (U.S. EPA. 2023 a)). Notably, EPA's conclusion was supported by the Science Advisory Committee on Chemicals (SACC) (U.S. EPA. 2023b). A brief summary of the MOA for phthalate syndrome and data available for DIBP supporting this MOA is provided below in Figure 3-1. Readers are directed to see EPA's Draft Proposed Approach for Cumulative Risk Assessment of High-Priority and a Manufacturer-Requested Phthalate under the Toxic Substances Control Act (U.S. EPA. 2023 a) for a more thorough discussion of DIBP's effects on the developing male reproductive system and EPA's MOA analysis. Effects on the developing male reproductive system are considered further for dose-response assessment in Section 4. As shown in Figure 3-1, a MOA for phthalate syndrome has been proposed to explain the link between gestational or perinatal exposure to DIBP and effects on the male reproductive system in rats. The MOA has been described in greater detail in EPA's Draft Proposed Approach for Cumulative Risk Assessment of High-Priority Phthalates and a Manufacturer-Requested Phthalate under the Toxic Substances Control Act (U.S. EPA. 2023a) and is described briefly below. Chemical Structure and Properties >=> Molecular Initiating Event Cellular Responses o Organ Responses Adverse Organism Outcomes Phthalate exposure during critical window of development Fetal Male Tissue J, AR dependent mRNA/protein synthesis ¦=> Metabolism to monoester & transport to fetal testes l=> Unknown MIE (not believed to be AR or PPARa mediated) 4- Testosterone synthesis IT Key genes involved in the AOP \ for phthalate syndrome Scarbl Cher? StAF Ebp Cypllcl Fdps Cypllbl Hmgcr Cypllb2 Hmgcsl Cypl7ol Hsd3b CypSl Fldll Mvd Ela3b Nsdhl Insl3 RGD1564999 Lhcgr Tm7sf2 inha Cyp46al NrObl Ldlr RhoxlO Insigl Wnt7a 4/ Gene expression (INSL3, lipid > metabolism, cholesterol and androgen synthesis and transport) 17 4- INSL3 synthesis Fetal Leydig cell ^> Abnormal cell apoptosis/ proliferation (Nipple/areolae retention, >1/ AGD, Disrupted testis tubules, Leydig cell clusters, MNGs, agenesis of reproductive tissues) Suppressed gubernacular cord development {inguinoscrotal phase) Suppressed gubernacular cord development (transabdominal phase) 4- Androgen- dependent tissue weights, testicular pathology [e.g., seminiferous tubule atrophy), malformations (e.g., hypospadias), 4* sperm production <0 Impaired s. fertility Undescended testes Figure 3-1. Hypothesized Phthalate Syndrome Mode of Action Following Gestational Exposure Figure taken directly from ( J.S. EPA. 2023a) and adapted from (Conlev et al.. 2021; Gray et al.. 2021; Schwartz et al.. 2021; Howdeshell et al.. 2017). AR = androgen receptor; INSL3 = insulin-like growth factor 3; MNG = multinucleated gonocytes; PPARa = peroxisome proliferator-activated receptor alpha. Phthalate syndrome is characterized by both androgen-dependent (e.g., reduced AGD, increased male NR.) and -independent effects (e.g., germ cell effects) on the male reproductive system ( J.S. EPA. 2023a). The MOA underlying phthalate syndrome has not been fully established; however, key cellular-, Page 32 of 94 ------- 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 PUBLIC RELEASE DRAFT December 2024 organ-, and organism4evel effects are generally understood (Figure 3-1). The molecular events preceding cellular changes remain unknown. Although androgen receptor antagonism and peroxisome proliferator-activated receptor alpha activation have been hypothesized to play a role, studies have generally ruled out the involvement of these receptors (Foster. 2005; Foster et al.. 2001; Parks et al.. 2000). Exposure to DIBP during the masculinization programming window (i.e., GDs 15.5 to 18.5 for rats; GDs 14 to 16 for mice; gestational weeks 8 to 14 for humans), in which androgen action drives development of the male reproductive system, can lead to antiandrogenic effects on the male reproductive system (MacLeod et al.. 2010; Welsh et al.. 2008; Carruthers and Foster. 2005). Consistent with the MOA outlined in Figure 3-1, seven studies (5 of rats, 2 of mice) of DIBP have demonstrated that oral exposure to DIBP during the masculinization programming window can reduce mRNA and/or protein expression of insulin-like growth factor 3 (INSL3), as well as genes involved in steroidogenesis in the testes of rats (Gray et al.. 2021; Saillenfait et al.. 2017; Hannas et al.. 2012; Hannas et al.. 2011; Borch et al.. 2006) and mice (Pan et al.. 2017; Wang et al.. 2017). Consistently, nine studies (7 of rats, 2 of mice) have also demonstrated that oral exposure to DIBP during the masculinization programming window can reduce testicular testosterone content and/or testosterone production in rats (Gray et al.. 2021; Saillenfait et al.. 2017; Furr et al.. 2014; Hannas et al.. 2012; Hannas et al.. 2011; Howdeshell et al.. 2008; Borch et al.. 2006) and mice (Pan et al.. 2017; Wang et al.. 2017). Oral exposure of rats to DIBP during the masculinization programming window has also been shown to reduce male pup anogenital distance (AGD) in three studies (Saillenfait et al.. 2017; Saillenfait et al.. 2008; Borch et al.. 2006) and cause male pup nipple retention (NR) in one study (Saillenfait et al.. 2008). which are two hallmarks of anti androgenic substances (see Sections 3.1.3.3 and 3.1.3.4 of (U.S. EPA. 2023 a) for additional discussion). Additional effects consistent with phthalate syndrome observed in mice and rats following oral exposure to DIBP during the critical window of development include: reproductive tract malformations (i.e., hypospadias, undescended testes, exposed os penis, cleft prepuce) in two studies of rats (Saillenfait et al.. 2008; Saillenfait et al.. 2006); delayed preputial separation (PPS) in one study of rats (Saillenfait et al.. 2008); testicular pathology in two studies of rats (e.g., degeneration of seminiferous tubules, oligospermia, azoospermia, Leydig cell aggregation, Sertoli cell vacuolation, multinucleated gonocytes) (Saillenfait et al.. 2008; Borch et al.. 2006); and decreased sperm concentration and motility in two studies of mice (Pan et al.. 2017; Wang et al.. 2017). Collectively, available studies consistently demonstrate that oral exposure to DIBP during the masculinization programming window in rats and mice can disrupt androgen action, leading to a spectrum of effects on the developing male reproductive system consistent with development of phthalate syndrome. As noted above, this conclusion was supported by the Science Advisory Committee on Chemicals (SACC) (U.S. EPA. 2023b) and readers are directed to EPA's Draft Proposed Approach for Cumulative Risk Assessment of High-Priority and a Manufacturer-Requested Phthalate under the Toxic Substances Control Act (U.S. EPA. 2023 a) for additional discussion of DIBP's effects on the developing male reproductive system and EPA's MOA analysis. 3.1.2.2 Other Developmental Outcomes In addition to effects on the developing male reproductive system, other developmental effects (e.g., decreased fetal weight, decreased offspring body weight, resorptions, post-implantation loss, skeletal variations) have been observed in experimental animal models following oral exposure to DIBP. However, these effects occur at higher doses than those that result in effects on the developing male reproductive system and frequently coincide with maternal toxicity (Table 3-3). Data supporting other developmental effects of DIBP are discussed below. Page 33 of 94 ------- 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 PUBLIC RELEASE DRAFT December 2024 In a study that adhered to OECD test guideline 414, pregnant Wistar rats (22 to 23 per dose) were administered diets containing 0, 1000, 4000, or 11,000 ppm DIBP (equivalent to 88, 363, or 942 mg/kg- day) from GD 6 through 20 and then sacrificed on GD 20 (BASF. 2007). Maternal and developmental effects were limited to the high-dose group and included a 5 percent decrease in maternal food consumption as well as an 11 percent decrease in maternal bodyweight gain from GD 6 through 20, a 5 percent decrease in fetal body weight, and increased incidences of skeletal variations (e.g., incomplete ossification of sternebra, unilateral ossification of sternebra). No significant increases in malformations were observed. No developmental or maternal toxicity was observed in the low- or mid-dose groups. In a second study, pregnant SD rats (20 to 22 per dose) were exposed to 0, 250, 500, 750, or 1000 mg/kg-day DIBP from GD 6 through 20 via gavage and then sacrificed on GD 21 (Saillenfait et al.. 2006). Maternal effects were limited to a decrease in weight gain on GD 6 through 9 and GD 15 through 18 in dams treated with 500 mg/kg-day DIBP and above; however, dam body weight gain on GD 6 through 21 corrected for gravid uterine weight was unaffected. Developmental toxicity was observed at 500 mg/kg-day and above. Observed developmental effects included: increased resorptions and post- implantation loss per litter and decreased live fetuses per litter at 750 mg/kg-day and above; increased incidence of total number of fetuses and/or litters with external, visceral, and skeletal malformations at 750 mg/kg-day and above; and increased incidence of undescended testes and decreased fetal body weight (both sexes) at 500 mg/kg-day and above. Howdeshell et al. (2008) reported increased fetal mortality and total resorptions, and decreased numbers of live fetuses in pregnant SD rats gavaged with 900 mg/kg-day DIBP from GDs 8 to GDI 8 and sacrificed on GD 18. Additionally, Borch et al. (2006) reported reduced fetal body weight on GD 19 in pregnant Wistar rats gavaged with 600 mg/kg-day DIBP on GD 7 through 19. In addition to decreased fetal weight, decreased offspring body weight was observed following gestational exposures. Saillenfait et al. (2008) reported reduced male offspring body weight on PND1, PND21 as well as PNW11 to 12 and PNW16 to 17 following gestational exposure to 500 to 625 mg/kg-day DIBP on GD 12 through 21. Collectively, available studies provide consistent evidence that gestational exposure to DIBP can result in a spectrum of developmental effects in addition to those of the developing male reproductive system. However, effects on the developing male reproductive system (Section 3.1.2.1) occur at much lower doses than the aforementioned other developmental effects. Therefore, effects on the developing male reproductive system are the most sensitive to DIBP exposure and are consistent with a disruption of androgen action and phthalate syndrome. Furthermore, the lowest LOAELs for effects on the developing male reproductive system range from 125 to 300 mg/kg-day, while the lowest LOAELs for other developmental outcomes range from 500 to 600 mg/kg-day (Table 3-3). Page 34 of 94 ------- 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 PUBLIC RELEASE DRAFT December 2024 4 DOSE-RESPONSE ASSESSMENT EPA considered reproductive/developmental toxicity as the sole non-cancer hazard endpoint for dose- response analysis. This hazard endpoint was selected for dose-response analysis because EPA has the highest confidence in this hazard endpoint for estimating risk to human health; effects were consistently observed across species and durations of exposure and occurred in a dose-related manner. Other non- cancer hazard endpoints considered by EPA (i.e., liver and kidney toxicity) were not utilized for dose- response analysis due to limitations and uncertainties that reduce EPA's confidence in using these endpoints for estimating risk to human health. For toxicologically similar phthalates (i.e., DEHP, DBP, BBP, DCHP), which include larger databases of animal toxicology studies including numerous well- conducted subchronic and chronic toxicity studies, effects on the developing male reproductive system consistent with a disruption of androgen action have consistently been identified by EPA as the most sensitive and well-characterized hazard in experimental animal models. This is demonstrated by the fact that the preliminary acute/intermediate/chronic PODs selected by EPA for use in risk characterization for DEHP (U.S. EPA. 2024h). DBP (U.S. EPA. 2024f). BBP (U.S. EPA. 2024e). DCHP (U.S. EPA. 2024g) are all based on effects related to phthalate syndrome. According to previous assessments, liver is a target organ following DIBP exposure (U.S. CPSC. 2011; NICNAS. 2008a); however, Health Canada (2015b) concluded that DIBP has low systemic toxicity based on a limited number of repeated oral dose toxicity studies. Additionally, a systematic review by Yost et al. (2019) stated that several studies indicate dose dependent increases in liver weight following intermediate and chronic DIBP exposure in rats and male mice (Wang et al.. 2017; Foster et al.. 1982; Oishi andHiraga. 1980; University of Rochester. 1953). However, there are no available data on other hepatic endpoints, such as clinical chemistry (e.g., ALT, ALT, bilirubin) and histology effects, following oral DIBP exposure. The lack of such data reduces EPAs confidence in using effects on the liver as an endpoint from which to derive a POD, because there is uncertainty about adversity without corroborating clinical chemistry or histology (Hall et al.. 2012; U.S. EPA. 2002a). Likewise, effects on the kidney following exposure to DIBP were evaluated by a limited number of studies, wherein inconsistencies across species were observed, as summarized in previous assessments and publications (Yost et al.. 2019; ECHA. 2017b; NICNAS. 2016; U.S. CPSC. 2011; NICNAS. 2008a). No new studies were identified that provided data on hepatic or renal effects following exposure to DIBP were identified through the TSCA systematic review process; therefore, EPA is in agreement with the conclusions of these previous assessments as well as those of the systematic review by Yost et al. (2019) [as described previously in Section 1.2.3], EPA used a BMD modeling approach for individual data sets of fetal testicular testosterone changes for the dose-response analysis. EPA did consider NOAEL and LOAEL values from oral toxicity studies in experimental animal models. The use of a NOAEL/LOAEL approach is supported by consistency across several studies that have evaluated effects on the developing male reproductive system consistent with phthalate syndrome that are similar and cluster around a single human equivalent dose (HED) NOAEL value, which supports identification of a consensus NOAEL. For reduced fetal testicular testosterone in rats, EPA conducted meta-analysis and benchmark dose modeling using the approach previously published by NASEM (2017). which is further described in EPA's Draft Meta-Analysis and Benchmark Dose Modeling of Fetal Testicular Testosterone for Di (2-ethylhexyl) Phthalate (DEHP), Dibutyl Phthalate (DBP), Butyl Benzyl Phthalate (BBP), Diisobittyl Phthalate (DIBP), Dicyclohexyl Phthalate (DCHP), andDiisononylPhthalate (U.S. EPA. 2024d). Acute, intermediate, and chronic non-cancer NOAEL, LOAEL, and BMDLs values identified by EPA are discussed further in Section 4.2. As discussed further in Section 4.2, EPA considers effects on the developing male reproductive system consistent with a disruption of androgen action relevant for setting a POD for acute exposure durations. However, because these acute effects are the most sensitive effects following exposure to DIBP, they are also considered protective of intermediate and chronic duration exposures. As described in Appendix C, Page 35 of 94 ------- 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 PUBLIC RELEASE DRAFT December 2024 EPA converted oral PODs derived from animal studies to human equivalent doses (HEDs) using allometric body weight scaling to the three-quarters power (U.S. EPA. 2011c). Species differences in dermal and oral absorption are corrected for as part of the dermal exposure assessment (U.S. EPA. 2025c). In the absence of inhalation studies, EPA performed route-to-route extrapolation to convert oral HEDs to inhalation human equivalent concentrations (HECs) (Appendix C). 4.1 Selection of Studies and Endpoints for Non-cancer Health Effects EPA considered the suite of oral animal toxicity studies primarily indicating effects on the developing male reproductive system consistent with phthalate syndrome when considering non-cancer PODs for estimating risks for acute, intermediate, and chronic exposure scenarios as described in Section 4.2. EPA considered the following factors during study and endpoint selection for POD determination from relevant non-cancer health effects: Exposure duration; Dose range; Relevance (e.g., considerations of species, whether the study directly assesses the effect, whether the endpoint is the best marker for the toxicological outcome, etc); Uncertainties not captured by the overall quality determination; Endpoint/POD sensitivity; and Total uncertainty factors (UFs). EPA considers the overall uncertainty with a preference for selecting studies that provide a lower uncertainty (e.g., lower benchmark MOE) because provides higher confidence (e.g., use of a NOAEL or BMDLs vs. a LOAEL with additional UFl applied). The following sections provide comparisons of the above attributes for studies and hazard outcomes relevant to each of these exposure durations and details related to the studies considered for each exposure duration scenario. 4.2 Non-cancer Oral Points of Departure for Acute, Intermediate, and Chronic Exposures 4.2.1 Studies Considered for the Non-Cancer POD EPA considered 11 developmental toxicity studies (10 of rats, 1 of mice) with endpoints relevant to acute, intermediate, and chronic exposure duration (U.S. EPA. 1996. 1991). summarized in Table 4-5. Of the considered studies, all 11 evaluated gestational or perinatal exposures to DIBP. No one or two- generation studies on the effects of DIBP on reproduction have been identified by EPA. Further, of the 11 studies considered, 5 only evaluated one exposure level of DIBP (i.e., did not evaluate dose-response across multiple exposure levels) ranging from 200 to 750 mg/kg-day (Table 4-5) (Saillenfait et al.. 2017; Wang et al.. 2017; Furr et al.. 2014; Hannas et al.. 2012; Borch et al.. 2006). Of the six remaining studies considered, four tested doses as low as 100 to 125 mg/kg-day (Table 4-5) (Gray et al.. 2021; Hannas et al.. 2011; Howdeshell et al.. 2008; Saillenfait et al.. 2008). however, no studies evaluating effects on the developing male reproductive system consistent with a disruption of androgen action have been conducted with DIBP that have evaluated doses below 100 mg/kg-day. Available studies considered for dose-response are discussed further below. As discussed in Sections 3.1.2.1 and 3.1.2.2, oral exposure to DIBP can cause effects on the developing male reproductive system consistent with a disruption of androgen action and other developmental effects (i.e., decreased fetal weight, resorptions, post-implantation loss, skeletal variations). Effects on Page 36 of 94 ------- 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 PUBLIC RELEASE DRAFT December 2024 the developing male reproductive system are more sensitive than other observed developmental effects. This is demonstrated by the fact that the lowest LOAELs for effects on the developing male reproductive system range from 125 to 300 mg/kg-day, while the lowest LOAELs for other developmental outcomes range from 500 to 600 mg/kg-day (Table 3-3, Table 4-5). Therefore, EPA's dose-response assessment in this section focuses on effects on the developing male reproductive system consistent with a disruption of androgen action. Although single dose studies evaluating the effects of DIBP on the developing male reproductive system are not available, studies of the toxicologically similar phthalate dibutyl phthalate (DBP) have demonstrated that a single exposure during the critical window of development can disrupt expression of steroidogenic genes and decrease fetal testes testosterone. Therefore, EPA considers effects on the developing male reproductive system consistent with a disruption of androgen action to be relevant for setting a POD for acute, intermediate, and chronic duration exposures (see Appendix B for further discussion). Notably, SACC agreed with EPA's decision to consider effects on the developing male reproductive system consistent with a disruption of androgen action to be relevant for setting a POD for acute durations during the July 2024 peer-review meeting of the DINP human health hazard assessment (U.S. EPA. 2024m). Studies considered for dose-response assessment are summarized in Table 4-5. Of the 11 developmental toxicity studies considered for dose-response, two studies (BASF. 2007; Saillenfait et al.. 2006) were not considered further for dose-response analysis because of limitations and other factors that increase uncertainty. In Saillenfait et al. (2006). rats were exposed to doses of DIBP ranging from 250 to 1000 mg/kg-day on GD 6 through 20 via gavage. Decreased fetal body weight and increased incidence of cryptorchidism were observed at 500 mg/kg-day. Based on these effects, EPA identified a NOAEL of 250 mg/kg-day. Similarly, BASF (2007) conducted a dietary study of pregnant Wistar rats in which animals were exposed to 88 to 942 mg/kg-day of DIBP from GDs 6 through 20. A NOAEL of 363 mg/kg-day was identified based on decreases in fetal body weight, maternal food consumption, and maternal body weight gain at 942 mg/kg-day. However, the doses at which developmental effects were observed in these studies were higher than doses at which more sensitive effects of androgen insufficiency (e.g., decreased fetal testicular testosterone) were observed in other studies. Therefore, EPA did not select these studies and endpoints because they do not provide the most sensitive robust endpoint for an acute/intermediate/chronic POD. Seven studies reported across five publications (Saillenfait et al.. 2017; Wang et al.. 2017; Furr et al.. 2014; Hannas et al.. 2012; Borch et al.. 2006) that exposed pregnant mice or rats to DIBP via gavage have observed effects on the developing male reproductive system. However, experiments in each of these studies only tested one dose level in addition to vehicle controls, and support LOAELs ranging from 200 to 750 mg/kg-day DIBP. These studies do not allow for the identification of a NOAEL, which increases the uncertainty in the data set. Ultimately, these studies were not further considered because other developmental studies of DIBP are available that test more than one dose level, including doses less than 200 mg/kg-day and support identification of more sensitive NOAELs. In contrast, three studies of pregnant SD rats provide consistent evidence of dose-related reductions in ex vivo fetal testicular testosterone production and support NOAEL and LOAEL values of 100 and 300 mg/kg-day, respectively (Table 4-5) (Gray et al.. 2021; Hannas et al.. 2011; Howdeshell et al.. 2008). Notably, the magnitude of effect on ex vivo fetal testicular testosterone production was consistent across tested doses in all three studies when measured on GDI8. For example, the response compared to the control ranged from 95 to 110 percent at 100 mg/kg-day and 44 to 66 percent at 300 mg/kg-day. Across the three studies, there is consistent evidence of no effect on ex vivo fetal testicular testosterone production in rats dosed with 100 mg/kg-day DIBP. Page 37 of 94 ------- 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 PUBLIC RELEASE DRAFT December 2024 In 2017, NASEM (2017) assessed experimental animal evidence for effects on fetal testicular testosterone following in utero exposure to DIBP using the systematic review methodology developed by the National Toxicology Program's (NTP) Office of Health Assessment and Translation (OHAT). Based on results from two studies of rats (Hannas et al.. 2011; Howdeshell et al.. 2008). NASEM found high confidence in the body of evidence and a high level of evidence that fetal exposure to DIBP is associated with a reduction in fetal testosterone in rats. NASEM further conducted a meta-regression analysis and benchmark dose (BMD) modeling analysis on decreased fetal testicular testosterone production data from the same two prenatal exposure studies of rats (Hannas et al.. 2011; Howdeshell et al.. 2008). NASEM found a statistically significant overall effect and linear trends in logio(dose) and dose, with an overall large magnitude of effect (greater than 50 percent) in its meta-analysis for DIBP. BMD analysis determined BMDLs and BMDL40 values of 23 and 225 mg/kg-day, respectively, the 95 percent lower confidence limits of the BMDs associated with a benchmark response (BMR) of 5 and 40 percent (Table 4-1). Table 4-1. Summary of NASEM (2017) Meta-Analysis and BMD Modeling for Effects of DIBP in Fetal Testosterone ab Database Supporting Outcome Confidence in Evidence Evidence of Outcome Heterogeneity in Overall Effect Model with Lowest AIC BMDs mg/kg- day (95% CI) BMD40 mg/kg- day (95% CI) 2 rat studies High High I2 > 60% Linear 27 (23, 34)' 270 (225, 340) 0 R code supporting NASEM's meta-regression and BMD analysis of DIBP is publicly available through GitHub (https ://github. com/wachiuphd/NASEM-2017 -Endocrine-Low-Dose). b NASEM (2017) calculated BMD40s for this endpoint because "previous studies have shown that reproductive-tract malformations were seen in male rats when fetal testosterone production was reduced by about 40%." c EPA noted an apparent discrepancy in the NASEM (2017) report. In Table 3-26, NASEM (2017) notes that no BMD/BMDL estimates could be generated at the 5% response level for DIBP because "the 5% change was well below the range of the data, but it will be 10 times lower because a linear model was usedHowever, in Table C6-12 of the NASEM (2017) report, BMD/BMDL estimates at the 5% response level are provided for DIBP for the best-fit linear model. In EPA's replicate analysis, which is provided in EPA's Draft Meta-Analysis and BMD of Fetal Testicular Testosterone for DEHP, DBP, BBP, DIBP, and DCHP (U.S. EPA. 2024d). identical BMD/BMDL estimates for the 5% response level were obtained. Therefore, BMD/BMDL estimates at the 5% response level for DIBP are reported in this table. Since EPA identified new fetal testicular testosterone data (Gray et al.. 2021) for DIBP, an updated meta-analysis was conducted. Using the publicly available R code provided by NASEM (https://github.com/wachiuphd/NASEM-2017-Endocrine-Low-Dose). EPA applied the same meta- analysis and BMD modeling approach used by NASEM, with the exception that the most recent Metafor package available at the time of EPA's updated analysis was used (i.e., EPA used Metafor package Version 4.6.0, whereas NASEM used Version 2.0.0), and an additional BMR of 10 percent was modelled. Appendix D provides justification for the evaluated BMRs of 5, 10, and 40 percent. Fetal rat testosterone data from three studies was included in the analysis (Gray et al.. 2021; Hannas et al.. 2011; Howdeshell et al.. 2008). Overall, the meta-analysis found a statistically significant overall effect and linear trends in logio(dose) and dose, with an overall effect that is large in magnitude (>50% change) (Table 4-2). There was substantial, statistically significant heterogeneity in all cases (I2>60%). The statistical significance of these effects was robust to leaving out individual studies. The linear-quadratic model provided the best fit (based on lowest AIC) (Table 4-2). BMD estimates from the linear-quadratic model were 270 mg/kg-day [95% confidence interval: 136, 517] for a 40 percent change (BMR = 40%>) and 55 mg/kg-day [NA, 266] for a 10 percent change (BMR = 10%>), although a BMDL10 could not be estimated (Table 4-3). No BMD could be estimated for a 5 percent change (BMR = 5%). Further Page 38 of 94 ------- 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 PUBLIC RELEASE DRAFT December 2024 methodological details and results (e.g., forest plots, figures of BMD model fits) for the updated meta- analysis and BMD modeling of fetal testicular testosterone data are provided in the Draft Meta-Analysis and Benchmark Dose Modeling of Fetal Testicular Testosterone for Di(2-ethylhexyl) Phthalate (DEHP), Dibutyl Phthalate (DBP), Butyl Benzyl Phthalate (BBP), Diisobiityl Phthalate (DIBP), Dicyclohexyl Phthalate (DCHP), andDiisononylPhthalate (U.S. EPA. 2024d). Table 4-2. Overall Analyses of Rat Studies of DIBP and Fetal Testosterone (Updated Analysis Conducted by EPA) Analysis Estimate Beta CI, Lower Bound CI, Upper Bound P value Tau I2 P value for Heterogeneity AICs Primary Analysis Overall intrcpt -82.21 -122.85 -41.56 0.000 68.02 96.52 0.000 130.45 Trend in loglO(dose) loglO(dose) -165.55 -205.47 -125.64 0.000 19.89 65.48 0.004 106.31 Linear in doselOO doselOO -18.48 -25.14 -11.81 0.000 60.86 96.92 0.000 120.04 LinearQuadratic in doselOO doselOO -19.18 -41.21 2.85 0.088 48.79 94.49 0.000 111.51* LinearQuadratic in doselOO I(dosel00A2) 0.09 -2.70 2.88 0.950 48.79 94.49 0.000 111.51 Sensitivity Analysis Overall minus Gray et al. 2021 intrcpt -82.31 -135.11 -29.52 0.002 71.76 96.96 0.000 87.28 Overall minus Hannas et al. 2011b intrcpt -69.98 -110.63 -29.34 0.001 55.43 95.94 0.000 83.66 Overall minus Howdeshell et al. 2008 intrcpt -94.90 -151.74 -38.06 0.001 78.38 94.86 0.000 88.36 * Indicates lowest AIC. Table 4-3. Benchmark Dose Estimates for DIBP and Fetal Testosterone in Rats Analysis Benchmark Response (BMR) Benchmark Dose (BMD) Confidence Interval, Lower Bound Confidence Interval, Upper Bound Linear in doselOO 5% 28 20 43 Linear in doselOO 10% 57 42 89 Linear in doselOO 40% 276 203 432 LinearQuadratic in doselOO* 5% NA NA 207 LinearQuadratic in doselOO* 10% 55 NA 266 LinearQuadratic in doselOO* 40% 270 136 517 * Indicates model with lowest AIC. 'NA' indicates a BMD or BMDL estimate could not be derived. Since no BMDLs could be derived through the updated meta-analysis and BMD modeling analysis, EPA modelled individual fetal testicular testosterone data from the three studies included in the updated meta- analysis using EPA's BMD Software (BMDS version 3.3.2) (Gray et al.. 2021; Hannas et al.. 2011; Howdeshell et al.. 2008). This analysis included the full suite of standard continuous models Page 39 of 94 ------- 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 PUBLIC RELEASE DRAFT December 2024 (Exponential, Hill, Polynomial, Power, Linear), compared to the meta-analysis that only included the linear and linear-quadratic models. Further methodological details and results from this BMD analysis are provided in Appendix E. As can be seen from Table Apx E-l, no models adequately fit the fetal testicular testosterone data from Hannas et al. (2011). In contrast, BMDs and BMDLs values of 63 and 24 mg/kg-day were derived from the fetal testicular testosterone data reported in Gray et al. (2021) based on the best fitting exponential 3 model (constant variance), while BMDs and BMDLs values of 103 and 52 mg/kg-day were derived from the fetal testicular testosterone data reported in Howdeshell et al. (2008) based on the best fitting hill model (constant variance). Lastly, Saillenfait et al. (2008) reported the results of oral exposure to 0, 125, 250, 500, or 625 mg/kg- day y DIBP on GD 12 through 21 on F1 male offspring. Treatment-related effects at 250 mg/kg-day DIBP and above include decreased F1 male AGD on PND 1, increased male nipple retention on PND 12 to 14 and PNW 11 to 12 or PNW 16 to 17, while more severe reproductive tract malformations (e.g., hypospadias, exposed os penis, nonscrotal testes) were observed at 500 mg/kg-day DIBP and above. In the low dose group (125 mg/kg-day), low incidence of testicular pathology was observed in F1 males from PNW 11 to 12, including oligospermia (low sperm) (incidence: 0/24, 1/20, 3/28, 2/22, 1/20), azoospermia (no sperm present) (0/24, 1/20, 3/28, 10/22, 18/20), and tubular degeneration, which showed evidence of increasing severity with dose. However, the study is limited due to a lack of statistical analysis on the testicular pathology data and due to the small sample size (only two F1 males were examined per litter). Although the incidence of testicular pathology at 125 mg/kg-day is low, EPA considers the study to support a LOAEL of 125 mg/kg-day (no NOAEL identified) due to the severity of the observed effects (i.e., reduced and/or absence of sperm in 2/20 adult F1 males). EPA considered BMD modeling of data from Saillenfait et al. (2008). However, BMD modeling of data from Saillenfait et al. (2008) has previously been published by EPA's Office of Research and Development (Blessmger et al.. 2020). As can be seen from Table 4-4, the BMDs and BMDLs values for the more sensitive outcomes evaluated by Saillenfait et al. (i.e., combined azoospermia and oligospermia) fall outside of the range of measured tested doses. Table 4-4. Summary of Dichotomous BMD Analysis of Data from Saillenfait et al. (2008) by Blessinger et al. (2020)" Endpoint BMR BMD (mg/kg-day) BMDL (mg/kg-day) Hypospadias 1% extra risk 401 242 Undescended testes 1% extra risk 342 194 Exposed os penis 1% extra risk 361 112 Areola or nipple retention 5% extra risk 317 205 Azoospermia or grade 2-5 oligospermia 5% extra risk 117 60 Tubular degeneration 5% extra risk 480 266 Sloughed cells 5% extra risk 112 56 11 Adapted from Table 6 in Blessinger et al. (2020). See Blessinger et al. for a description of the BMD modeling approach. BMD modeling outputs from Blessinger et al. are available at: httos://doi.ors/10.23719/1503702. Page 40 of 94 ------- 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 PUBLIC RELEASE DRAFT December 2024 4.2.2 Options Considered by EPA for Deriving the Acute Non-Cancer POD In order to derive a non-cancer POD for DIBP, EPA considered three options, including: Option 1. NOAEL/LOAEL approach to identify the highest NOAEL below the lowest LOAEL (Section 4.2.2.1). Option 2. Application of a data-derived adjustment factor based on differences in relative potency to reduced fetal testicular testosterone (Section4.2.2.2). Option 3. BMD modeling of fetal testicular testosterone (Section 4.2.2.3). The strengths and limitations of each of the approaches considered by EPA to derive a non-cancer POD for DIBP are discussed further below, while the POD EPA selected for the draft risk evaluation of DIBP is discussed in Section 4.2.3. 4.2.2.1 Option 1. NOAEL/LOAEL Approach Overall, EPA considers Saillenfait et al. (2008) to support a LOAEL of 125 mg/kg-day based on low incidence of testicular histopathological findings. Three additional studies of fetal testicular testosterone all support a NOAEL of 100 mg/kg-day (Gray et al.. 2021; Hannas et al.. 2011; Howdeshell et al.. 2008). Each of these three studies gavaged pregnant SD rats with the same DIBP doses (0, 100, 300, 600, 900 mg/kg-day) on GDs 14-18 (Gray et al.. 2021; Hannas et al.. 2011) or GDs 8-18 (Howdeshell et al.. 2008). For each of the three studies, ex vivo fetal testicular testosterone production was then measured on GD 18, approximately 2 hours after the final dose of DIBP was administered. Results from these studies did not observe any significant changes in ex vivo fetal testicular testosterone production at 100 mg/kg-day when measured on GDI8; however, at the 300 mg/kg-day DIBP dose, the response compared to the control ranged from 44 to 66 percent, supporting a NOAEL of 100 mg/kg-day in these studies (Gray et al.. 2021; Hannas et al.. 2011; Howdeshell et al.. 2008). Therefore, EPA considers these 4 studies to support a NOAEL for fetal testicular testosterone of 100 mg/kg-day and a LOAEL for testicular histopathology of 125 mg/kg-day. However, there are several lines of evidence that suggest a NOAEL of 100 mg/kg-day may be under- protective, including: The database of studies for DIBP is limited to 11 gestational or perinatal oral exposures studies, 5 of which tested a single high dose level of 200 to 750 mg/kg-day, while no studies have evaluated doses below 100 mg/kg-day. BMD modeling of testicular pathology data from Saillenfait et al. (2008) supports BMDLs values of 56 to 60 mg/kg-day based on incidence of sloughed cells or combined azoospermia/oligospermia (Table 4-4). EPA's updated meta-analysis and BMD modeling analysis of fetal testicular testosterone supported a BMDio of 55 mg/kg-day. No BMDLs could not be derived from the best-fitting linear quadratic model as part of the updated analysis (Table 4-3). 4.2.2.2 Option 2. Application of a Data-Derived Adjustment Factor EPA also considered differences in relative potency between toxicologically similar phthalates to derive a data-derived adjustment factor. As discussed in EPA's Draft Technical Support Document for the Cumulative Risk Analysis of Di(2-ethylhexyl) Phthalate (DEHP), Dibutyl Phthalate (DBP), Butyl Benzyl Phthalate (BBP), Diisobutyl Phthalate (DIBP), Dicyclohexyl Phthalate (DCHP), andDiisononyl Phthalate (DINP) Under the Toxic Substances Control Act (TSCA) (U.S. EPA. 20241). EPA has derived a draft relative potency factor (RPF) of 0.53 for DIBP, based on its relative potency compared to the index chemical, dibutyl phthalate (DBP), at reducing fetal testicular testosterone. The draft POD for the Page 41 of 94 ------- 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 PUBLIC RELEASE DRAFT December 2024 index chemical, DBP, is a BMDLs of 9 mg/kg-day derived from EPA's updated meta-analysis and BMD modeling analysis of fetal testicular testosterone (U.S. EPA. 2024f 1). The draft POD of 9 mg/mg-day for the index chemical (DBP) is approximately 11.1 times lower than the NOAEL of 100 mg/kg-day identified for DIBP identified above in Section 4.2.2.1. In contrast, the draft RPF of 0.53 indicates that the POD for DIBP should be approximately twice that of DBP, since DIBP is approximately half as potent as DBP as reducing fetal testicular testosterone. Therefore, EPA considered adjusting the DIBP NOAEL of 100 by a factor of 5.89 (i.e., (DIBP NOAEL DBP BMDLs) * RPFdibp), which would result in an adjusted NOAEL of 17 mg/kg-day. Notably, ECHA (2017a. b) employed a similar relative potency adjustment for DIBP. When deriving a POD for DIBP for use in risk characterization, ECHA (2017a. b) stated: "Few reproductive toxicity studies have been published on [DIBP] compared to DEHP and DBP. No two-generation studies are available and the substance has not been tested at doses <100 mgkg bw/d. Current data suggest that DIBP coirfd have similar effects to DBP, if studied at lower dose levels. If the potency difference between DIBP and DBP, as a very rough estimate of the observed effects in Saillenfait et al. (2008) (type of effects seen at 500 and 625 mg kg bw-day, corresponding to a difference of 25%), is extrapolatedfrom the high dose area to the lower dose area, an estimatedLOAEL for DIBP would be 25% higher than the current LOAEL for DBP (2 mg kg bw-day). Available information is shown in Table B7. A LOAEL for DIBP of 2.5 mg kg bw-day is selectedfor use in the current combined risk assessment " 4.2.2.3 Option 3. BMD Analysis of Individual Fetal Testicular Testosterone Studies Because no BMDLs could be derived via the updated meta-analysis and BMD analysis of fetal testicular testosterone data, EPA modelled individual ex vivo fetal testicular testosterone production data sets using EPA's BMD Software (Gray et al.. 2021; Hannas et al.. 2011; Howdeshell et al.. 2008). No models adequately fit the Hannas et al. (2011) data set (TableApx E-l). In contrast, BMDs and BMDLs values of 63 and 24 mg/kg-day were derived from the Gray et al. (2021) data set based on the best fitting exponential 3 model, while BMDs and BMDLs values of 103 and 52 mg/kg-day were derived from the Howdeshell et al. (2008) data set based on the best fitting Hill model (Table Apx E-l).The BMDLs of 52 mg/kg-day from Howdeshell et al. (2008) is similar to the derived BMDio of 55 mg/kg-day from EPA's updated meta-analysis (Table 4-3) suggesting the BMDLs of 52 mg/kg-day is not appropriate for use in human health risk characterization. Additionally, although the linear model in EPA's updated meta-analysis did not provide the best-fit (i.e., the linear-quadratic model had a lower AIC), the linear model did appear to adequately fit the data set and supports BMDs and BMDLs values of 28 and 20 mg/kg-day (Table 4-3). The BMDLs of 24 mg/kg-day from Gray et al. (2021) is similar to the BMDLs of 20 mg/kg-day derived using the linear model in the updated meta-analysis. Although there is some uncertainty because derived BMDLs estimates are below the lowest dose with empirical data (i.e., 100 mg/kg-day), EPA considers this BMD analysis to support a BMDLs of 24 mg/kg-day based on reduced fetal testicular testosterone in the study by Gray et al. (2021). 4.2.3 POD Selected for Acute, Intermediate, and Chronic Durations Considering the three options described above in Section 4.2.2,, EPA selected the BMDLs of 24 mg/kg- day (option 3) based on reduced fetal testicular testosterone from the study by Gray et al. (2021). EPA considered the POD derived from the BMD analysis of data in this study to have the least uncertainty and highest confidence upon examination of the weight of evidence provided by the three options. This POD is more sensitive than the NOAEL of 100 mg/kg-day (option 1), which is likely under-protective due to the limited number of studies and lack of testing at doses lower than 100 mg/kg-day. EPA Page 42 of 94 ------- 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 PUBLIC RELEASE DRAFT December 2024 considers the BMDLs of 24 mg/kg-day (option 3) to be more appropriate than using the data-derived adjustment factor based on relative potency to the index chemical, DBP, (Option 2) because the BMDLs of 24 mg/kg-day relies on adequately modeled fetal testosterone production data specific to DIBP. While the application of relative potency is appropriate and necessary for the cumulative risk assessment across phthalates, the use of data exclusive to DIBP has less uncertainty for the individual risk from DIBP. Using allometric body weight scaling to the three-quarters power, (U.S. EPA. 2011c). EPA extrapolated an HED of 5.7 mg/kg-day from the BMDLs of 24 mg/kg-day. A total uncertainty factor of 30 was selected for use as the benchmark margin of exposure (based on an interspecies uncertainty factor (UFa) of 3 and an intraspecies uncertainty factor (UFh) of 10). Consistent with EPA guidance (2022. 2002b. 1993). EPA reduced the UFa from a value of 10 to 3 because allometric body weight scaling to the three-quarter power was used to adjust the POD to obtain a HED (Appendix C). EPA considered reducing the UFa further to a value of 1 based on apparent differences in toxicodynamics between rats and humans. As discussed in Section 3.1.4 of EPA's Draft Proposed Approach for Cumulative Risk Assessment of High-Priority Phthalates and a Manufacturer-Requested Phthalate under the Toxic Substances Control Act (U.S. EPA. 2023 a). several explant (Lamb rot et al.. 2009; Hallmark et al.. 2007) and xenograft studies (van Den Driesche et al.. 2015; Spade et al.. 2014; Heger et al.. 2012; Mitchell et al.. 2012) using human donor fetal testis tissue have been conducted to investigate the antiandrogenicity of mono-2-ethylhexyl phthalate (MEHP; a monoester metabolite of DEHP), DBP, and monobutyl phthalate (MBP; a monoester metabolite of DBP) in a human model. Generally, results from human explant and xenograft studies suggest that human fetal testes are less sensitive than rat testes to the antiandrogenic effects of phthalates, however, effects on Sertoli cells and increased incidence of MNGs have been observed in two human xenograft studies of DBP (van Den Driesche et al.. 2015; Spade et al.. 2014; Heger et al.. 2012; Mitchell et al.. 2012). As discussed in EPA's draft approach document (U.S. EPA. 2023a). the available human explant and xenograft studies have limitations and uncertainties, which preclude definitive conclusions related to species differences in sensitivity. For example, key limitations and uncertainties of the human explant and xenograft studies include: small sample size; human testis tissue was collected from donors of variable age and by variable non-standardized methods; and most of the testis tissue was taken from fetuses older than 14 weeks, which is outside of the critical window of development (i.e., gestational weeks 8 to 14 in humans). Therefore, EPA did not reduce the UFa. Page 43 of 94 ------- PUBLIC RELEASE DRAFT December 2024 1336 Table 4-5. Dose-Response Analysis of Selected Studies Considered for Acute, Intermediate, and Chronic Exposure Scenarios Study Details (Species, Duration, Exposure Route/ Method, Doses [mg/kg- day]) Study POD/ Type (mg/kg-day) Effect HED (mg/kg) Uncertainty Factors"b c Reference(s) Sprague-Dawley rats; GD 14-18; oral/gavage; 0, 100, 300, 600, 900 BMDLs = 24 I ex vivo testicular testosterone production (34%) 5.7 UFa = 3 UFh = 10 Total UF = 30 (Grav et al.. 2021) Sprague-Dawley rats; GD 8-18; 0, 100, 300, 600, 900 BMDLs = 52 I ex vivo testicular testosterone production (40%) 12.3 UFa = 3 UFh = 10 Total UF = 30 (Howdeshell et al.. 2008) Sprague-Dawley rats; GD 14-18; oral/gavage; 0, 100, 300, 600, 900 NOAEL = 100 I ex vivo fetal testicular testosterone production (56%); I expression of steroidogenic genes in fetal testes 23.6 UFa = 3 UFh = 10 Total UF = 30 (Hannas et al.. 2011) Sprague-Dawley rats; GD 12-21; oral/gavage; 0, 125, 250, 500, 625 LOAEL = 125 Testicular pathology (degeneration of seminiferous tubules and oligo-/azoospennia in epididymis) 29.6 UFa = 3 UFh = 10 UFl = 10 Total UF = 300 (Saillenfait et al.. 2008) Sprague-Dawley rats; GD 14-18; oral/gavage; 0, 200 (Block 30) LOAEL = 200 I ex vivo fetal testicular testosterone production 47.3 UFa = 3 UFh = 10 UFl = 10 Total UF = 300 (Furr et al.. 2014) Sprague-Dawley rats; GD 6-20; oral/gavage; 0, 250, 500, 750, 1000 NOAEL = 250 I fetal body weight (both sexes); t incidence of cryptorchidism 59.1 UFa = 3 UFh = 10 Total UF = 30 (Saillenfait et al.. 2006) Sprague-Dawley rats; GD 13-19; oral/gavage; 0, 250 LOAEL = 250 J.AGD, I testicular testosterone & androstenedione production, altered mRNA expression of steroidogenesis genes in the testes 59.1 UFa = 3 UFh = 10 UFl = 10 Total UF = 300 (Saillenfait et al.. 2017) ICR Mice; GD 0-21; oral/gavage; 0, 450 LOAEL = 450 I absolute testes weight on PND 21; J. serum and testes testosterone; J. expression of steroidogenic genes in testes; |sperm concentration and motility on PND 80 59.8 UFa = 3 UFh = 10 UFl = 10 Total UF = 300 (Wane etal..2017) Wistar Rat; oral/diet; 0, 88, 363, 942 NOAEL = 363 I maternal food consumption, [ maternal body weight gain, [ fetal body weight 85.8 UFa = 3 UFh = 10 (BASF. 2007) Page 44 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Study Details (Species, Duration, Exposure Route/ Method, Doses [mg/kg- day]) Study POD/ Type (mg/kg-day) Effect HED (mg/kg) Uncertainty Factors"b c Reference(s) Total UF = 30 Sprague-Dawley rats; GD 14-18; oral/gavage; 0, 500 (Block 14) LOAEL = 500 I ex vivo fetal testicular testosterone production 118 UFa = 3 UFh = 10 UFl = 10 Total UF = 300 (Furr et al.. 2014) Sprague-Dawley rats; GD 14-18; oral/gavage; 0, 500 LOAEL = 500 I ex vivo fetal testicular testosterone production 118 UFa = 3 UFh = 10 UFl = 10 Total UF = 300 (Hannas et al.. 2012) Wistar Rat; GD 7-19 or 7-20/21; oral/gavage; 0, 600 LOAEL = 600 I testes testosterone, [ AGD, | testicular histopathology 142 UFa = 3 UFh = 10 UFl = 10 Total UF = 300 (Borch et al.. 2006) Sprague-Dawley rats; GD 14-18; oral/gavage; 0, 750 (Block 2) LOAEL = 750 I ex vivo fetal testicular testosterone production 177 UFa = 3 UFh = 10 UFl = 10 Total UF = 300 (Furr et al.. 2014) Abbreviations: j = statistically significant decrease; t = statistically significant increase; POD = Point of Departure; HED = Human equivalent Dose; UF = uncertainty factor; NOAEL = No observed adverse effect level; LOAEL = Lowest observed adverse effect level; GD = Gestational Day; PND = Postnatal Day AGD = Anogenital distance; BMD = benchmark dose. "EPA used allometric bodv weieht scalins to the three-auarters rower to derive the HED. Consistent with EPA Guidance (U.S. EPA. 201 lc). the interspecies uncertainty factor (UFA), was reduced from 10 to 3 to account remaining uncertainty associated with interspecies differences in toxicodynamics. b EPA used a default intraspecies (UFH) of 10 to account for variation in sensitivity within human populations due to limited information regarding the degree to which human variability may impact the disposition of or response to DIBP. c EPA used a LOAEL-to-NOAEL uncertainty factor (UFl) of 10 to account for the uncertainty inherent in extrapolating from the LOAEL to the NOAEL. J Two studies with similar desisns were included in the meta-analvsis bv NASEM (2017). each of which exrosed Suraeue-Dawlev rats (< 3 dams/dose) to 0. 100, 300, 600, 900 mg/kg-day DIBP during the masculinization programming window during gestational development. 1337 Page 45 of 94 ------- 1338 1339 1340 1341 1342 1343 1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381 1382 PUBLIC RELEASE DRAFT December 2024 4.3 Weight of The Scientific Evidence Conclusion: POD for Acute, Intermediate, and Chronic Durations EPA considered BMD modelling from the study by Gray et al. to support a BMDLs of 24 mg/kg-day (Gray et al.. 2021). EPA has preliminarily concluded that the HED of 5.7 mg/kg-day (BMDLs of 24 mg/kg-day) based on decreased fetal testicular testosterone production from the gestational exposure study of rats by Gray et al. is appropriate for calculation of risk from acute, intermediate, and chronic durations. A total uncertainty factor of 30 was selected for use as the benchmark margin of exposure (based on an interspecies uncertainty factor (UFa) of 3 and an intraspecies uncertainty factor (UFh) of 10). Consistent with EPA guidance (2022. 2002b. 1993). EPA reduced the UFa from a value of 10 to 3 because allometric body weight scaling to the three-quarter power was used to adjust the POD to obtain a HED (Appendix C). Given the limited database of studies for DIBP that have evaluated outcomes other that developmental toxicity and effects on the developing male reproductive system. For toxicologically similar phthalates (i.e., DEHP, DBP, BBP, DCHP), which include larger databases of animal toxicology studies including numerous well-conducted subchronic and chronic toxicity studies, effects on the developing male reproductive system consistent with a disruption of androgen action have consistently been identified by EPA as the most sensitive and well-characterized hazard in experimental animal models. This is demonstrated by the fact that the preliminary acute/intermediate/chronic PODs selected by EPA for use in risk characterization for DEHP (U.S. EPA. 2024h). DBP (U.S. EPA. 2024ft. BBP (U.S. EPA. 2024e). DCHP (U.S. EPA. 2024g) are all based on effects related to phthalate syndrome. EPA has robust overall confidence in the selected POD based on the following weight of the scientific evidence: EPA has previously considered the weight of scientific evidence and concluded that oral exposure to DIBP can induce effects on the developing male reproductive system consistent with a disruption of androgen action (see EPA's Draft Proposed Approach for Cumulative Risk Assessment of High-Priority and a Manufacturer-Requested Phthalate under the Toxic Substances Control Act (U.S. EPA. 2023 a)). Notably, EPA's conclusion was supported by the SACC (U.S. EPA. 2023b). DIBP exposure resulted in treatment-related effects on the developing male reproductive system consistent with a disruption of androgen action during the critical window of development in 13 studies of rats (Section 3.1.2.1). Observed effects included: reduced fetal testicular testosterone content and/or testosterone production; reduced male pup anogenital distance; male pup nipple retention; reproductive tract malformations (i.e., hypospadias, undescended testes, exposed os penis, cleft prepuce); delayed preputial separation; testicular pathology (e.g., degeneration of seminiferous tubules, oligospermia, azoospermia, Leydig cell aggregation, Sertoli cell vacuolation, multinucleated gonocytes); decreased sperm concentration and motility. The selected POD is a BMDLs based on reduced ex vivo fetal testicular testosterone production in one gestational exposure studies of rats (Gray et al.. 2021). Consistently, other regulatory and authoritative bodies have also concluded that DIBP induces effects on the developing male reproductive system consistent with a disruption of androgen action and phthalate syndrome and that these effects are relevant for estimating human risk (ECCC/HC. 2020: ECHA. 2017a. b; U.S. CPSC. 2014: ECHA. 2012a. b; NICNAS. 2008a). EPA considers effects on the developing male reproductive system consistent with a disruption of androgen action to be relevant for setting a POD for acute, intermediate, and chronic duration exposures, based on studies of the toxicologically similar phthalate dibutyl phthalate (DBP) Page 46 of 94 ------- 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 PUBLIC RELEASE DRAFT December 2024 which have demonstrated that a single exposure during the critical window of development in rats can disrupt expression of steroidogenic genes and decrease fetal testes testosterone production. EPA did not identify any studies conducted via the dermal route relevant for extrapolating human health risk. Therefore, EPA is using the oral HED of 24 mg/kg to extrapolate to the dermal route. EPA's approach to dermal absorption for workers, consumers, and the general population is described in EPA's Draft Environmental Release and Occupational Exposure Assessment for Diisobiityl phthalate (U.S. EPA. 2025c). EPA did not identify any inhalation studies of DIBP. Therefore, EPA is also using the oral HED of 24 mg/kg to extrapolate to the inhalation route. EPA assumes similar absorption for the oral and inhalation routes, and no adjustment was made when extrapolating to the inhalation route. For the inhalation route, EPA extrapolated the daily oral HEDs to inhalation HECs using a human body weight and breathing rate relevant to a continuous exposure of an individual at rest. Appendix C provides further information on extrapolation of inhalation HECs from oral HEDs. Page 47 of 94 ------- 1398 1399 1400 1401 1402 1403 1404 1405 1406 1407 1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434 1435 1436 1437 1438 1439 1440 1441 1442 PUBLIC RELEASE DRAFT December 2024 5 CONSIDERATION OF PESS AND AGGEGRATE EXPOSURE 5.1 Hazard Considerations for Aggregate Exposure For use in the risk evaluation and assessing risks from other exposure routes, EPA conducted route-to- route extrapolation of the toxicity values from the oral studies for use in the dermal and inhalation exposure routes and scenarios. Health outcomes that serve as the basis for acute, intermediate, and chronic hazard values are systemic and assumed to be consistent across routes of exposure. EPA therefore concludes that for consideration of aggregate exposures, it is reasonable to assume that exposures and risks across oral, dermal, and inhalation routes may be additive for the selected PODs in Section 6. 5.2 PESS Based on Greater Susceptibility In this section, EPA addresses subpopulations likely to be more susceptible to DIBP exposure than other populations. Table 5-1 presents the data sources that were used in the potentially exposed or susceptible subpopulations (PESS) analysis evaluating susceptible subpopulations and identifies whether and how the subpopulation was addressed quantitatively in the draft risk evaluation of DIBP. Although ample human epidemiologic data are available on health effects of DIBP (see Section 3.1.1), EPA was unable to identify direct evidence of differences in susceptibility among human populations. Animal studies demonstrating effects on male reproductive development and other developmental outcomes provide direct evidence that gestation is a particularly sensitive lifestage. Evidence from animal studies also suggests that the liver may also be a target organ; however, there is not enough evidence to reliably inform specific health outcomes or to be used in risk quantification. Therefore, EPA is quantifying risks including those for PESS based on reproductive and developmental toxicity in the draft DIBP risk evaluation. As summarized in Table 5-1, EPA identified a range of factors that may have the potential to increase biological susceptibility to DIBP, including lifestage, pre-existing diseases, physical activity, nutritional status, stress, and co-exposures to other environmental stressors that contribute to related health outcomes. The effect of these factors on susceptibility to health effects of DIBP is not known; therefore, EPA is uncertain about the directions and magnitude of any possible increased risk from effects associated with DIBP exposure for relevant subpopulations. For non-cancer endpoints, EPA used a default value of 10 for human variability (UFh) to account for increased susceptibility when quantifying risks from exposure to DIBP. The Risk Assessment Forum, in A Review of the Reference Dose and Reference Concentration Processes (U.S. EPA. 2002b). discusses some of the evidence for choosing the default factor of 10 when data are lacking and describe the types of populations that may be more susceptible, including different lifestages (e.g., of children and elderly). Although U.S. EPA (2002b) did not discuss all the factors presented in Table 5-1, EPA considers the POD selected for use in characterizing risk from exposure to DIBP to be protective of effects on the developing male reproductive system consistent with phthalate syndrome in humans. As discussed in U.S. EPA (2023a). exposure to DIBP and other toxicologically similar phthalates (i.e., DEHP, DBP, BBP, DCHP, DINP) that disrupt androgen action during the development of the male reproductive system cause dose additive effects. Cumulative effects from exposure to DIBP and other toxicologically similar phthalates will be evaluated as part of U.S. EPA's cumulative risk assessment of phthalates. Page 48 of 94 ------- 1443 PUBLIC RELEASE DRAFT December 2024 Table 5-1. PESS Evidence Crosswalk for Biological Susceptibility Considerations Susceptibility Category Examples of Specific Factors Direct Evidence this Factor Modifies Susceptibility to DIBP Description of Interaction Key Citations Indirect Evidence of Interaction with Target Organs or Biological Pathways Relevant to DIBP Description of Interaction Key Citation(s) Susceptibility Addressed in Risk Evaluation? Embryos/ fetuses/infants Lifestage Direct quantitative animal evidence for developmental toxicity (e.g., increased skeletal variations, decreased fetal body weight, increased resorptions, and post-implantation loss). There is direct quantitative animal evidence for effects on the developing male reproductive system consistent with a disruption of androgen action. (U.S. EPA. 2023a) (U.S. EPA. 2023b) (Howdeshell et al 2008) (Hannas et al 2011) (Wang et al.. 2017) (Saillenfait et al.. 2008) (BASF. 2007) (Borch et al.. 2006) (Saillenfait et al.. 2006) Pregnancy/ lactating status Rodent dams not particularly susceptible during pregnancy and lactation, except for effects related to reduced maternal weight gain and food consumption evident only at high concentrations. (Howdeshell et al.. 2008) (Saillenfait et al.. 2006) (BASF. 2007) POD selected for assessing risks from acute, intermediate, and chronic exposures to DIBP is based on developmental toxicity (i.e., reduced fetal testicular testosterone production) and is protective of effects on the fetus and offspring. POD selected for assessing risks from acute, intermediate, and chronic exposures to DIBP based on developmental toxicity (i.e., reduced fetal testicular testosterone production) is protective of effects on dams Page 49 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Susceptibility Category Examples of Specific Direct Evidence this Factor Modifies Susceptibility to DIBP Indirect Evidence of Interaction with Target Organs or Biological Pathways Relevant to DIBP Susceptibility Addressed in Risk Evaluation? Factors Description of Interaction Key Citations Description of Interaction Key Citation(s) Males of reproductive age Consistent evidence of effects on endpoints related to male reproductive development in rats and mice, including steroidogenesis in the testes and effects on sperm (i.e., decreased concentration and motility, increased malformation). (Pan etal.. 2017) POD selected for assessing risks from acute, intermediate, and chronic exposures to DIBP is based on effects on male reproductive development (i.e., reduced fetal testicular testosterone production) is expected to be protective of adult male reproductive effects. Children Reduced rodent offspring body weight gain between PNDs 1 to 21 was observed in three gestational exposure studies. (Saillenfait et al.. 2008) (Wans et al.. 2017) (BASF. 2007) POD selected for assessing risks from acute, intermediate, and chronic exposures to DIBP based on developmental toxicity (i.e., reduced fetal testicular testosterone production) is expected to be protective of effects of offspring bodyweight gain. Use of default lOx UFH Elderly No direct evidence identified Use of default lOx UFH Page 50 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Susceptibility Category Examples of Specific Direct Evidence this Factor Modifies Susceptibility to DIBP Indirect Evidence of Interaction with Target Organs or Biological Pathways Relevant to DIBP Susceptibility Addressed in Risk Evaluation? Factors Description of Interaction Key Citations Description of Interaction Key Citation(s) Health outcome/ target organs No direct evidence identified Several preexisting conditions may contribute to adverse developmental outcomes (e.g., diabetes, high blood pressure, certain viruses). CDC (2023e) CDC (2023 g) Use of default lOx UFH Pre-existing disease or disorder Individuals with chronic liver disease may be more susceptible to effects on these target organs. Viruses such as viral hepatitis can cause liver damage. Toxicokinetics No direct evidence identified Chronic liver disease is associated with impaired metabolism and clearance (altered expression of phase 1 and phase 2 enzymes, impaired clearance), which may enhance exposure duration and concentration of DIBP. Use of default lOx UFH Lifestyle activities Smoking No direct evidence identified Smoking during pregnancy may increase susceptibility for developmental outcomes (e.g., early delivery and stillbirths). CDC (2023f) Page 51 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Susceptibility Category Examples of Specific Direct Evidence this Factor Modifies Susceptibility to DIBP Indirect Evidence of Interaction with Target Organs or Biological Pathways Relevant to DIBP Susceptibility Addressed in Risk Evaluation? Factors Description of Interaction Key Citations Description of Interaction Key Citation(s) Alcohol consumption No direct evidence identified Alcohol use during pregnancy can cause adverse developmental outcomes (e.g., fetal alcohol spectrum disorders). Heavy alcohol use may affect susceptibility to liver disease. CDC (2023d) CDC (2023a) Physical activity No direct evidence identified Insufficient activity may increase susceptibility to multiple health outcomes. Overly strenuous activity may also increase susceptibility. CDC (2022) Sociodemo- graphic status Race/ethnicity No direct evidence identified (e.g., no information on polymorphisms in DIBP metabolic pathways or diseases associated race/ethnicity that would lead to increased susceptibility to effects of DIBP by any individual group). Socioeconomic status No direct evidence identified Individuals with lower incomes may have worse health outcomes due to social needs that are not met, enviromnental concerns, and barriers to health care access. ODPHP (2023b) Page 52 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Susceptibility Category Examples of Specific Factors Direct Evidence this Factor Modifies Susceptibility to DIBP Indirect Evidence of Interaction with Target Organs or Biological Pathways Relevant to DIBP Susceptibility Addressed in Risk Evaluation? Description of Interaction Key Citations Description of Interaction Key Citation(s) Sex/gender Male reproductive development is a sex-specific endpoint and consistent evidence indicates it is the most sensitive effect following gestational or early life DIBP exposure. See discussion in Section 3.1.2.1. POD selected for assessing risks from acute, intermediate, and chronic exposures to DIBP is based on effects on male reproductive development (i.e., reduced fetal testicular testosterone production) Nutrition Diet No direct evidence identified Poor diets can lead to chronic illnesses such as heart disease, type 2 diabetes, and obesity, which may contribute to adverse developmental outcomes. Additionally, diet can be a risk factor for fatty liver, which could be a pre-existing condition to enhance susceptibility to DIBP-induced liver toxicity. CDC (2023e) CDC (2023b) Page 53 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Susceptibility Category Examples of Specific Direct Evidence this Factor Modifies Susceptibility to DIBP Indirect Evidence of Interaction with Target Organs or Biological Pathways Relevant to DIBP Susceptibility Addressed in Risk Evaluation? Factors Description of Interaction Key Citations Description of Interaction Key Citation(s) Malnutrition No direct evidence identified Micronutrient malnutrition can lead to multiple conditions that include birth defects, maternal and infant deaths, preterm birth low birth weight, poor fetal growth, childhood blindness, undeveloped cognitive ability. Thus, malnutrition may increase susceptibility to some developmental outcomes associated with DIBP. CDC (2021) CDC (2023b) Target organs No direct evidence identified Polymorphisms in genes may increase susceptibility to liver or developmental toxicity. Use of default lOx UFH Genetics/ epigenetics Toxicokinetics No direct evidence identified Polymorphisms in genes encoding enzymes (e.g., esterases) involved in metabolism of DIBP may influence metabolism and excretion of DIBP. Use of default lOx UFH Other chemical and nonchemical stressors Built enviromnent No direct evidence identified Poor-quality housing is associated with a variety of negative health outcomes. ODPHP (2023a) Page 54 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Susceptibility Category Examples of Specific Direct Evidence this Factor Modifies Susceptibility to DIBP Indirect Evidence of Interaction with Target Organs or Biological Pathways Relevant to DIBP Susceptibility Addressed in Risk Evaluation? Factors Description of Interaction Key Citations Description of Interaction Key Citation(s) Social environment No direct evidence identified Social isolation and other social determinants (e.g., decreased social capital, stress) can lead to negative health outcomes. CDC (2023c) ODPHP (2023c) Chemical co- exposures Studies have demonstrated that co-exposure to DIBP and other toxicologically similar phthalates (e.g., DEHP, DBP, BBP, DCHP, DINP) and other classes of antiandrogenic chemicals (e.g., certain pesticides and pharmaceuticals - discussed more in (U.S. EPA. 2023a)) can induce effects on the developing male reproductive system in a dose- additive manner. See (TJ.S. EPA. 2023a) and (U.S. EPA 2023b) Co-exposures will be quantitatively addressed as part of the phthalate cumulative risk assessment and are not addressed in the individual DIBP assessment. 1444 Page 55 of 94 ------- 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 PUBLIC RELEASE DRAFT December 2024 6 POINTS OF DEPARTURE USED TO ESTIMATE RISKS FROM DIBP EXPOSURE, CONCLUSOINS, AND NEXT STEPS After considering hazard identification and evidence integration, dose-response evaluation, and weight of the scientific evidence of POD candidates, EPA chose one non-cancer endpoint for use in determining the risk from acute, intermediate, and chronic exposure scenarios (see Table ES-1). The critical effect is disruption to androgen action during the critical window of male reproductive development (i.e., during gestation), leading to a spectrum of effects on the developing male reproductive system consistent with phthalate syndrome. Decreased fetal testicular testosterone was selected as the basis for the POD of 24 mg/kg-day (HED = 5.7 mg/kg-day) for acute, intermediate, and chronic durations. EPA has robust overall confidence in the selected POD for acute, intermediate, and chronic durations. There are no studies conducted via the dermal and inhalation route relevant for extrapolating human health risk. In the absence of inhalation studies, EPA performed route-to-route extrapolation to convert the oral HED to an inhalation human equivalent concentration (HEC) of 30.9 mg/m3 (2.71 ppm). EPA is also using the oral HED to extrapolate to the dermal route. HECs are based on daily continuous (24-hour) exposure, and HEDs are daily values. The POD of 24 mg/kg-day (HED = 5.7 mg/kg-day) will be used in the Draft Risk Evaluation for DIBP (U.S. EPA. 2024i) to estimate acute, intermediate, and chronic non-cancer risk. EPA summarizes the cancer hazards of DIBP in a separate technical support document, Draft Cancer Raman Health Hazard Assessment for Di(2-ethylhexyl) Phthalate (DEHP), Dibutyl Phthalate (DBP), Butyl Benzyl Phthalate (BBP) andDicyclohexylPhthalate (DCHP) (U.S. EPA. 2025a). EPA is soliciting comments from the Science Advisory Committee on Chemicals (SACC) and the public on the non-cancer hazard identification, dose-response and weight of evidence analyses, and the selected POD for use in risk characterization of DIBP. Page 56 of 94 ------- 1470 1471 1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482 1483 1484 1485 1486 1487 1488 1489 1490 1491 1492 1493 1494 1495 1496 1497 1498 1499 1500 1501 1502 1503 1504 1505 1506 1507 1508 1509 1510 1511 1512 1513 1514 1515 1516 1517 PUBLIC RELEASE DRAFT December 2024 REFERENCES Allen. BC; Kavlock. RJ; Kimmel. CA; Faustman. EM. (1994a). Dose-response assessment for developmental toxicity II: Comparison of generic benchmark dose estimates with no observed adverse effect levels. Fundam Appl Toxicol 23: 487-495. http://dx.doi.org/10.1006/faat.1994.1133 Allen. BC: Kavlock. RJ: Kimmel CA: Faustman. EM. (1994b). Dose-response assessment for developmental toxicity III: statistical models. Fundam Appl Toxicol 23: 496-509. http://dx.doi.org/10.1006/faat.1994.1134 Aylward. LL; Hays. SM; Zidek. A. (2016). Variation in urinary spot sample, 24 h samples, and longer- term average urinary concentrations of short-lived environmental chemicals: implications for exposure assessment and reverse dosimetry. J Expo Sci Environ Epidemiol 27: 582-590. http://dx.doi.org/10.1038/ies.2016.54 BASF. (2007). Diisobutylphthalate - prenatal developmental toxicity study in Wistar rats administration in the diet (2007 Update) [TSCA Submission] (pp. 68-155). (Document Control Number: 86070000046). Submitted to the U.S. Environmental Protection Agency under TSCA Section 8d. http://vosemite. epa.gov/oppts/epatscat8. nsf/bv+Service/82FC6103C0E2F95585257B5100479E7 9/$File/86070000046.pdf Blessinger. TP: Euling. SY; Wang. L; Hogan. KA; Cai. C: Klinefelter. G: Saillenfait. AM. (2020). Ordinal dose-response modeling approach for the phthalate syndrome. Environ Int 134: 105287. http://dx.doi.Org/10.1016/i.envint.2019.105287 Borch. J: Axelstad. M; Vinggaard. AM: Dalgaard. M. (2006). Diisobutyl phthalate has comparable anti- androgenic effects to di-n-butyl phthalate in fetal rat testis. Toxicol Lett 163: 183-190. http://dx.doi.Org/10.1016/i.toxlet.2005.10.020 Burns. JS: Sergevev. O: Lee. MM: Williams. PL: Minguez-Alarcon. L; Plaku-Alakbarova. B; Sokolov. S: Kovalev. S: Koch. HM; Lebedev. AT: Hauser. R; Korrick. SA; Russian Children's. S. (2022). Associations of prepubertal urinary phthalate metabolite concentrations with pubertal onset among a longitudinal cohort of boys. Environ Res 212: 113218. http://dx.doi. org/10.1016/i. envres.2022.113218 Calafat. AM: Longnecker. MP: Koch. HM: Swan. SH: Hauser. R: Goldman. LR: Lanphear. BP: Rudel. RA: Engel. SM: Teitelbaum. SL: Whyatt. RM: Wolff. MS. (2015). Optimal exposure biomarkers for nonpersistent chemicals in environmental epidemiology. Environ Health Perspect 123: A166- A168. http://dx.doi.org/10.1289/ehp.1510041 Carruthers. CM: Foster. PMD. (2005). Critical window of male reproductive tract development in rats following gestational exposure to di-n-butyl phthalate. Birth Defects Res B Dev Reprod Toxicol 74: 277-285. http://dx.doi.org/10.1002/bdrb.2005Q CDC. (2021). CDC Health Topics A-Z: Micronutrients [Website], http s: //www, cdc. gov/nutriti on/mi cronutri ent- malnutrition/index.html?CDC AA refVal=https%3A%2F%2Fwww.cdc.gov%2Fimmpact%2Fin dex.html CDC. (2022). CDC Health Topics A-Z: Physical activity [Website], http s: //www, cdc. gov/phy si cal activity/index. html CDC. (2023a). Alcohol and Public Health: Alcohol use and your health [Website], https://www.cdc.gov/alcohol/fact-sheets/alcohol-use.htm CDC. (2023b). CDC Health Topics A-Z: Nutrition [Website], https://www.cdc.gov/nutrition/index.html CDC. (2023c). CDC Health Topics A-Z: Stress at work [Website], https://www.cdc.gov/niosh/topics/stress/ CDC. (2023d). Fetal Alcohol Spectrum Disorders (FASDs): Alcohol use during pregnancy [Website], https://www.cdc.gov/ncbddd/fasd/alcohol-use.html Page 57 of 94 ------- 1518 1519 1520 1521 1522 1523 1524 1525 1526 1527 1528 1529 1530 1531 1532 1533 1534 1535 1536 1537 1538 1539 1540 1541 1542 1543 1544 1545 1546 1547 1548 1549 1550 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560 1561 1562 1563 1564 1565 PUBLIC RELEASE DRAFT December 2024 CDC. (2023e). Pregnancy: During pregnancy [Website], https://www.cdc.gov/pregnancv/during.html CDC. (2023f). Smoking & Tobacco Use: Smoking during pregnancy - Health effects of smoking and secondhand smoke on pregnancies [Website], https://www.cdc.gov/tobacco/basic information/health effects/pregnancv/index.htm CDC. (2023 g). Viral Hepatitis: What is viral hepatitis? [Website], https://www.cdc.gov/hepatitis/abc/index.htm Chin. HB; Jukic. AM: Wilcox. AJ; Weinberg. CR; Ferguson. KK; Calafat AM: McConnaughev. PR: Baird. DP. (2019). Association of urinary concentrations of phthalate metabolites and bisphenol A with early pregnancy endpoints. Environ Res 168: 254-260. http://dx.doi.Org/10.1016/i.envres.2018.09.037 Conlev. JM; Lambright. CS: Evans. N: Cardon. M; Medlock-Kakalev. E; Wilson. VS: Gray. LE. (2021). A mixture of 15 phthalates and pesticides below individual chemical no observed adverse effect levels (NOAELs) produces reproductive tract malformations in the male rat. Environ Int 156: 106615. http://dx.doi.org/10.1016/i.envint.2021.106615 Pel Bubba. M; Ancillotti. C: Checchini. L; Fibbi. P; Rossini. P; Ciofi. L; Rivoira. L; Profeti. C: Orlandini. S: Furlanetto. S. (2018). Petermination of phthalate diesters and monoesters in human milk and infant formula by fat extraction, size-exclusion chromatography clean-up and gas chromatography-mass spectrometry detection. J Pharm Biomed Anal 148: 6-16. http://dx.doi.Org/10.1016/i.ipba.2017.09.017 Powns. SH; Black. N. (1998). The feasibility of creating a checklist for the assessment of the methodological quality both of randomised and non-randomised studies of health care interventions. J Epidemiol Community Health 52: 377-384. http://dx.doi.Org/10.1136/iech.52.6.377 Purmaz. E; Erkekoglu. P; Asci. A: Akcurin. S: Bircan. I; Kocer-Gumusel. B. (2018). Urinary phthalate metabolite concentrations in girls with premature thelarche. Environ Toxicol Pharmacol 59: 172- 181. http://dx.doi.Org/10.1016/i.etap.2018.03.010 EC/HC. (2015a). State of the science report: Phthalate substance grouping 1,2-Benzenedicarboxylic acid, diisononyl ester; 1,2-Benzenedicarboxylic acid, di-C8-10-branched alkyl esters, C9-rich (Piisononyl Phthalate; PINP). Chemical Abstracts Service Registry Numbers: 28553-12-0 and 68515-48-0. Gatineau, Quebec, https://www.ec.gc.ca/ese- ees/default.asp?lang=En&n=47F58AA5-l EC/HC. (2015b). State of the science report: Phthalate substance grouping: Medium-chain phthalate esters: Chemical Abstracts Service Registry Numbers: 84-61-7; 84-64-0; 84-69-5; 523-31-9; 5334-09-8; 16883-83-3; 27215-22-1; 27987-25-3; 68515-40-2; 71888-89-6. Gatineau, Quebec: Environment Canada, Health Canada. https://www.ec.gc.ca/ese-ees/4P845198-761P-428B- A519-7548 !B25B3E5/SoS Phthalates%20%28Medium-chain%29 EN.pdf ECCC/HC. (2020). Screening assessment - Phthalate substance grouping. (Enl4-393/2019E-PPF). Environment and Climate Change Canada, Health Canada. https://www.canada.ca/en/environment-climate-change/services/evaluating-existing- substances/screening-assessment-phthalate-substance-grouping.html ECHA. (2012a). Committee for Risk Assessment (RAC) Committee for Socio-economic Analysis (SEAC): Background document to the Opinion on the Annex XV dossier proposing restrictions on four phthalates. Helsinki, Finland, http://echa.europa.eu/documents/10162/3bc5088a-a231- 498e-86e6-8451884c6a4f ECHA. (2012b). Committee for Risk Assessment (RAC) Opinion on an Annex XV dossier proposing restrictions on four phthalates. (ECHA/RAC/RES-0-0000001412-86-07/F). Helsinki, Finland: European Chemicals Agency :: ECHA. https://echa.europa.eu/documents/10162/77cf7d29-ba63- 4901-aded-59cf75536e06 Page 58 of 94 ------- 1566 1567 1568 1569 1570 1571 1572 1573 1574 1575 1576 1577 1578 1579 1580 1581 1582 1583 1584 1585 1586 1587 1588 1589 1590 1591 1592 1593 1594 1595 1596 1597 1598 1599 1600 1601 1602 1603 1604 1605 1606 1607 1608 1609 1610 1611 1612 1613 1614 PUBLIC RELEASE DRAFT December 2024 ECHA. (2017a). Annex to the Background document to the Opinion on the Annex XV dossier proposing restrictions on four phthalates (DEHP, BBP, DBP, DIBP). (ECHA/RAC/RES-O- 0000001412-86-140/F; ECHA/SEAC/RES-O-OOOOOO1412-86- 154/F). https://heronet.epa.gov/heronet/index.cfm/reference/download/reference id/10328892 ECHA. (2017b). Opinion on an Annex XV dossier proposing restrictions on four phthalates (DEHP, BBP, DBP, DIBP). (ECHA/RAC/RES-0-0000001412-86-140/F). Helsinki, Finland. https://echa.europa.eu/documents/10162/e39983ad-lbf6-f402-7992-8a032b5b82aa Elsisi- AE; Carter. DE; Sipes. IG. (1989). Dermal absorption of phthalate diesters in rats. Fundam Appl Toxicol 12: 70-77. http://dx.doi.org/10.1016/0272-0590(89)90063-8 Faustman. EM: Allen. BC: Kavlock. RJ; Kimmel. CA. (1994). Dose-response assessment for developmental toxicity: I characterization of data base and determination of no observed adverse effect levels. Fundam Appl Toxicol 23: 478-486. http://dx.doi.Org/10.1006/faat.1994.l 132 Foster. PMD. (2005). Mode of action: Impaired fetal Leydig cell function - Effects on male reproductive development produced by certain phthalate esters [Review], Crit Rev Toxicol 35: 713-719. http://dx.doi.org/10.1080/104084405910Q7395 Foster. PMD: Lake. BG: Cook. MW; Thomas. LV; Gangolli. SD. (1982). Structure-activity requirements for the induction of testicular atrophy by butyl phthalates in immature rats: Effect on testicular zinc content. Adv Exp Med Biol 136: 445-452. http://dx.doi.org/10.10Q7/978-l- 4757-0674-1 33 Foster. PMD: Mylchreest. E; Gaido. KW: Sar. M. (2001). Effects of phthalate esters on the developing reproductive tract of male rats [Review], Hum Reprod Update 7: 231-235. http://dx.doi.Org/10.1093/humupd/7.3.231 Fromme. H; Gruber. L; Seckin. E; Raab. U: Zimmermann. S: Kiranoglu. M; Schlummer. M; Schwegler. U: Smolic. S: Volkel. W. (2011). Phthalates and their metabolites in breast milk - Results from the Bavarian Monitoring of Breast Milk (BAMBI). Environ Int 37: 715-722. http: //dx. doi. or g/10.1016/i. envint .2011.02.008 Furr. JR; Lambright. CS: Wilson. VS: Foster. PM; Gray. LE. Jr. (2014). A short-term in vivo screen using fetal testosterone production, a key event in the phthalate adverse outcome pathway, to predict disruption of sexual differentiation. Toxicol Sci 140: 403-424. http://dx.doi.org/10.1093/toxsci/kfu081 Gray. LE: Furr. J: Tatum-Gibbs. KR; Lambright. C: Sampson. H; Hannas. BR: Wilson. VS: Hotchkiss. A: Foster. PM. (2016). Establishing the Biological Relevance of Dipentyl Phthalate Reductions in Fetal Rat Testosterone Production and Plasma and Testis Testosterone Levels. Toxicol Sci 149: 178-191. http://dx.doi.org/10.1093/toxsci/kfv224 Gray. LE: Lambright. CS: Conlev. JM; Evans. N: Furr. JR: Hannas. BR: Wilson. VS: Sampson. H; Foster. PMD. (2021). Genomic and Hormonal Biomarkers of Phthalate-Induced Male Rat Reproductive Developmental Toxicity Part II: A Targeted RT-qPCR Array Approach That Defines a Unique Adverse Outcome Pathway. Toxicol Sci 182: 195-214. http://dx.doi.org/10.1093/toxsci/kfab053 Hall. AP; Elcombe. CR: Foster. JR: Harada. T; Kaufmann. W: Knippel. A: Kiittler. K: Malarkev. DE: Maronpot. RR: Nishikawa. A: Nolte. T; Schulte. A: Strauss. V: York. MJ. (2012). Liver hypertrophy: A review of adaptive (adverse and non-adverse) changesConclusions from the 3rd International ESTP Expert Workshop [Review], Toxicol Pathol 40: 971-994. http://dx.doi.org/10.1177/0192623312448935 Hallmark. N: Walker. M: McKinnell. C: Mahood. IK: Scott. H: Bavne. R: Coutts. S: Anderson. RA: Greig. I; Morris. K: Sharpe. RM. (2007). Effects of monobutyl and di(n-butyl) phthalate in vitro on steroidogenesis and Leydig cell aggregation in fetal testis explants from the rat: Comparison with effects in vivo in the fetal rat and neonatal marmoset and in vitro in the human. Environ Health Perspect 115: 390-396. http://dx.doi.org/10.1289/ehp.9490 Page 59 of 94 ------- 1615 1616 1617 1618 1619 1620 1621 1622 1623 1624 1625 1626 1627 1628 1629 1630 1631 1632 1633 1634 1635 1636 1637 1638 1639 1640 1641 1642 1643 1644 1645 1646 1647 1648 1649 1650 1651 1652 1653 1654 1655 1656 1657 1658 1659 1660 1661 1662 1663 PUBLIC RELEASE DRAFT December 2024 Hannas. BR; Lambright. CS; Furr. J; Evans. N; Foster. PMD; Gray. EL; Wilson. VS. (2012). Genomic biomarkers of phthalate-induced male reproductive developmental toxicity: A targeted RT-PCR array approach for defining relative potency. Toxicol Sci 125: 544-557. http://dx.doi.org/10.1093/toxsci/kfr315 Hannas. BR; Lambright. CS; Furr. J; Howdeshell. KL; Wilson. VS; Gray. LE. (2011). Dose-response assessment of fetal testosterone production and gene expression levels in rat testes following in utero exposure to diethylhexyl phthalate, diisobutyl phthalate, diisoheptyl phthalate, and diisononyl phthalate. Toxicol Sci 123: 206-216. http://dx.doi.org/10.1093/toxsci/kfrl46 Harlev. KG; Berger. KP; Kogut. K; Parra. K; Lustig. RH; Greenspan. LC; Calafat. AM; Ye. X; Eskenazi. B. (2019). Association of phthalates, parabens and phenols found in personal care products with pubertal timing in girls and boys. Hum Reprod 34: 109-117. http://dx.doi. org/10.1093/humrep/dev3 3 7 Hartle. JC; Cohen. RS; Sakamoto. P; Barr. DB; Carmichael. SL. (2018). Chemical contaminants in raw and pasteurized human milk. J Hum Lact 34: 340-349. http://dx.doi.org/10.1177/0890334418759308 Health Canada. (2018a). Supporting documentation: Evaluation of epidemiologic studies on phthalate compounds and their metabolites for effects on behaviour and neurodevelopment, allergies, cardiovascular function, oxidative stress, breast cancer, obesity, and metabolic disorders. Ottawa, ON. Health Canada. (2018b). Supporting documentation: Evaluation of epidemiologic studies on phthalate compounds and their metabolites for hormonal effects, growth and development and reproductive parameters. Ottawa, ON. Heger. NE; Hall. SJ; Sandrof. MA; McDonnell. EV; Henslev. JB; McDowell. EN; Martin. KA; Gaido. KW; Johnson. KJ; Boekelheide. K. (2012). Human fetal testis xenografts are resistant to phthalate-induced endocrine disruption. Environ Health Perspect 120: 1137-1143. http://dx.doi.org/10.1289/ehp. 1104711 Heggeseth. BC; Holland. N; Eskenazi. B; Kogut. K; Harlev. KG. (2019). Heterogeneity in childhood body mass trajectories in relation to prenatal phthalate exposure. Environ Res 175: 22-33. http://dx.doi.Org/10.1016/i.envres.2019.04.036 Hogberg. J; Hanberg. A; Berglund. M; Skerfving. S; Remberger. M; Calafat. AM; Filipsson. AF; Jansson. B; Johansson. N; Appelgren. M; Hakansson. H. (2008). Phthalate diesters and their metabolites in human breast milk, blood or serum, and urine as biomarkers of exposure in vulnerable populations. Environ Health Perspect 116: 334-339. http://dx.doi.org/10.1289/ehp.10788 Howdeshell. KL; Hotchkiss. AK; Gray. LE. (2017). Cumulative effects of antiandrogenic chemical mixtures and their relevance to human health risk assessment [Review], Int J Hyg Environ Health 220: 179-188. http://dx.doi.Org/10.1016/i.iiheh.2016.ll.007 Howdeshell. KL; Rider. CV; Wilson. VS; Furr. JR; Lambright. CR; Gray. LE. (2015). Dose addition models based on biologically relevant reductions in fetal testosterone accurately predict postnatal reproductive tract alterations by a phthalate mixture in rats. Toxicol Sci 148: 488-502. http://dx.doi.org/10.1093/toxsci/kfvl96 Howdeshell. KL; Wilson. VS; Furr. J; Lambright. CR; Rider. CV; Blystone. CR; Hotchkiss. AK; Gray. LE. Jr. (2008). A mixture of five phthalate esters inhibits fetal testicular testosterone production in the Sprague-Dawley rat in a cumulative, dose-additive manner. Toxicol Sci 105: 153-165. http://dx.doi.org/10.1093/toxsci/kfn077 Jensen. TK; Frederiksen. H; Kyhl. HB; Lassen. TH; Swan. SH; Bornehag. CG; Skakkebaek. NE; Main. KM; Lind. DV; Husbv. S; Andersson. AM. (2016). Prenatal exposure to phthalates and anogenital distance in male infants from a low-exposed Danish cohort (2010-2012). Environ Health Perspect 124: 1107-1113. http://dx.doi.org/10.1289/ehp.150987Q Page 60 of 94 ------- 1664 1665 1666 1667 1668 1669 1670 1671 1672 1673 1674 1675 1676 1677 1678 1679 1680 1681 1682 1683 1684 1685 1686 1687 1688 1689 1690 1691 1692 1693 1694 1695 1696 1697 1698 1699 1700 1701 1702 1703 1704 1705 1706 1707 1708 1709 1710 PUBLIC RELEASE DRAFT December 2024 Johnson. KJ; Heger. NE; Boekelheide. K. (2012). Of mice and men (and rats): phthalate-induced fetal testis endocrine disruption is species-dependent [Review], Toxicol Sci 129: 235-248. http://dx.doi.org/10.1093/toxsci/kfs206 Johnson. KJ: McDowell EN: Viereck. MP: Xia. JO. (2011). Species-specific dibutyl phthalate fetal testis endocrine disruption correlates with inhibition of SREBP2-dependent gene expression pathways. Toxicol Sci 120: 460-474. http://dx.doi.org/10.1093/toxsci/kfr020 Kim. JH; Kim. D; Moon. SM; Yang. EJ. (2020). Associations of lifestyle factors with phthalate metabolites, bisphenol A, parabens, and triclosan concentrations in breast milk of Korean mothers. Chemosphere 249: 126149. http://dx.doi.org/10.1016/i.chemosphere.2020.126149 Kim. S: Eom. S: Kim. HJ; Lee. JJ; Choi. G: Choi. S: Kim. S: Kim. SY; Cho. G: Kim. YD: Suh. E; Kim. SK; Kim. S: Kim. GH; Moon. HB; Park. J: Kim. S: Choi. K; Eun. SH. (2018). Association between maternal exposure to major phthalates, heavy metals, and persistent organic pollutants, and the neurodevelopmental performances of their children at 1 to 2 years of age-CHECK cohort study. Sci Total Environ 624: 377-384. http://dx.doi.Org/10.1016/i.scitotenv.2017.12.058 Kim. S: Lee. J: Park. J: Kim. HJ: Cho. G: Kim. GH: Eun. SH: Lee. JJ: Choi. G: Suh. E; Choi. S: Kim. S: Kim. YD: Kim. SK: Kim. SY: Kim. S: Eom. S: Moon. HB: Kim. S: Choi. K. (2015). Concentrations of phthalate metabolites in breast milk in Korea: estimating exposure to phthalates and potential risks among breast-fed infants. Sci Total Environ 508: 13-19. http: //dx. doi. or g/10.1016/i. scitotenv .2014.11.019 Koch. HM; Haller. A: Weifi. T; Kafferlein. HU: Stork. J: Briining. T. (2012). Phthalate exposure during cold plastisol application - A human biomonitoring study. Toxicol Lett 213: 100-106. http://dx.doi.Org/10.1016/i.toxlet.2011.06.010 Lambrot. R: Muczynski. V: Lecureuil. C: Angenard. G: Coffigny. H: Pairault. C: Moison. D: Frydman. R: Habert. R: Rouiller-Fabre. V. (2009). Phthalates impair germ cell development in the human fetal testis in vitro without change in testosterone production. Environ Health Perspect 117: 32- 37. http://dx.doi.org/10.1289/ehp.11146 Latini. G: Wittassek. M: Del Vecchio. A: Presta. G: De Felice. C: Angerer. J. (2009). Lactational exposure to phthalates in Southern Italy. Environ Int 35: 236-239. http://dx.doi.Org/10.1016/i.envint.2008.06.002 Lee. KY: Shibutani. M: Takagi. H: Kato. N: Takigami. S: Unevama. C: Hirose. M. (2004). Diverse developmental toxicity of di-n-butyl phthalate in both sexes of rat offspring after maternal exposure during the period from late gestation through lactation. Toxicology 203: 221-238. http://dx.doi.Org/10.1016/i.tox.2004.06.013 Lin. S: Ku. H: Su. P; Chen. J: Huang. P; Angerer. J: Wang. S. (2011). Phthalate exposure in pregnant women and their children in central Taiwan. Chemosphere 82: 947-955. http://dx.doi.Org/10.1016/i.chemosphere.2010.10.073 Machtinger. R: Mansur. A: Baccarelli. AA: Calafat. AM: Gaskins. AJ: Racowsky. C: Adir. M: Hauser. R. (2018). Urinary concentrations of biomarkers of phthalates and phthalate alternatives and IVF outcomes. Environ Int 111: 23-31. http://dx.doi.Org/10.1016/i.envint.2017.l 1.011 MacLeod. DJ: Sharpe. RM: Welsh. M: Fisken. M: Scott. HM: Hutchison. GR: Drake. AJ: van Den Driesche. S. (2010). Androgen action in the masculinization programming window and development of male reproductive organs. Int J Androl 33: 279-287. http://dx.doi.org/10.1111/i. 1365-2605.2009.01005.X Mitchell. RT: Childs. AJ: Anderson. RA: van Den Driesche. S: Saunders. PTK: McKinnell. C: Wallace. WHB: Kelnar. CJH: Sharpe. RM. (2012). Do phthalates affect steroidogenesis by the human fetal testis? Exposure of human fetal testis xenografts to di-n-butyl phthalate. J Clin Endocrinol Metab 97: E341-E348. http://dx.doi.org/10.1210/ic.2011-2411 Page 61 of 94 ------- 1711 1712 1713 1714 1715 1716 1717 1718 1719 1720 1721 1722 1723 1724 1725 1726 1727 1728 1729 1730 1731 1732 1733 1734 1735 1736 1737 1738 1739 1740 1741 1742 1743 1744 1745 1746 1747 1748 1749 1750 1751 1752 1753 1754 1755 1756 1757 1758 PUBLIC RELEASE DRAFT December 2024 NASEM. (2017). Application of systematic review methods in an overall strategy for evaluating low- dose toxicity from endocrine active chemicals. In Consensus Study Report. Washington, D.C.: The National Academies Press, http://dx.doi.org/10.17226/24758 NICNAS. (2008a). Existing chemical hazard assessment report: Diisobutyl phthalate. Sydney, Australia: National Industrial Chemicals Notification and Assessment Scheme. https://www.nicnas.gov.au/ data/assets/pdf file/0006/4965/DIBP-hazard-assessment.pdf NICNAS. (2008b). Phthalates hazard compendium: A summary of physicochemical and human health hazard data for 24 ortho-phthalate chemicals. Sydney, Australia: Australian Department of Health and Ageing, National Industrial Chemicals Notification and Assessment Scheme. https://www.regulations.gov/document/EPA-HQ-OPPT-2010-0573-00Q8 NICNAS. (2016). C4-6 side chain transitional phthalates: Human health tier II assessment. Sydney, Australia: Australian Department of Health, National Industrial Chemicals Notification and Assessment Scheme. https://www.industrialchemicals.gov.au/sites/default/files/C4- 6%20side%20chain%20transitional%20phthalates Human%20health%20tier%20II%20assessm ent.pdf NTP. (2015). Handbook for conducting a literature-based health assessment using OHAT approach for systematic review and evidence integration. Research Triangle Park, NC: U.S. Deptartment of Health and Human Services, National Toxicology Program, Office of Health Assessment and Translation, https://ntp.niehs.nih.gov/ntp/ohat/pubs/handbookian2015 508.pdf ODPHP. (2023a). Healthy People 2030 - Social determinants of health literature summaries: Neighborhood and built environment [Website], https://health.gov/healthypeople/prioritv- areas/social-determinants-health/literature-summaries#neighborhood ODPHP. (2023b). Healthy People 2030 - Social determinants of health literature summaries: Poverty [Website], https://health.gov/healthvpeople/prioritv-areas/social-determinants-health/literature- summaries/poverty ODPHP. (2023c). Healthy People 2030 - Social determinants of health literature summaries: Social and community context [Website], https://health.gov/healthvpeople/prioritv-areas/social- determinants-health/literature-summaries#social Oishi. S: Hiraga. K. (1980). Testicular atrophy induced by phthalic acid monoesters: Effects of zinc and testosterone concentrations. Toxicology 15: 197-202. http://dx.doi.org/10.1016/030Q- 483X(80)90053-0 Pan. Y; Wang. X: Yeung. LWY; Sheng. N: Cut 0: Cut R; Zhang. H; Dai. J. (2017). Dietary exposure to di-isobutyl phthalate increases urinary 5-methyl-2'-deoxycytidine level and affects reproductive function in adult male mice. J Environ Sci 61: 14-23. http://dx.doi.Org/10.1016/i.ies.2017.04.036 Parks. LG: Ostbv. JS: Lambright. CR; Abbott. BP: Klinefelter. GR; Barlow. NJ: Gray. LE. Jr. (2000). The plasticizer diethylhexyl phthalate induces malformations by decreasing fetal testosterone synthesis during sexual differentiation in the male rat. Toxicol Sci 58: 339-349. http://dx.doi.Org/10.1093/toxsci/58.2.339 Radke. EG: Braun. JM; Meeker. JD; Cooper. GS. (2018). Phthalate exposure and male reproductive outcomes: A systematic review of the human epidemiological evidence [Review], Environ Int 121: 764-793. http://dx.doi.Org/10.1016/i.envint.2018.07.029 Radke. EG: Braun. JM: Nachman. RM; Cooper. GS. (2020a). Phthalate exposure and neurodevelopment: A systematic review and meta-analysis of human epidemiological evidence [Review], Environ Int 137: 105408. http://dx.doi.Org/10.1016/i.envint.2019.105408 Radke. EG: Galizia. A: Thayer. KA; Cooper. GS. (2019a). Phthalate exposure and metabolic effects: A systematic review of the human epidemiological evidence [Review], Environ Int 132: 104768. http: //dx. doi. or g/10.1016/i. envint .2019.04.040 Page 62 of 94 ------- 1759 1760 1761 1762 1763 1764 1765 1766 1767 1768 1769 1770 1771 1772 1773 1774 1775 1776 1777 1778 1779 1780 1781 1782 1783 1784 1785 1786 1787 1788 1789 1790 1791 1792 1793 1794 1795 1796 1797 1798 1799 1800 1801 1802 1803 1804 1805 1806 PUBLIC RELEASE DRAFT December 2024 Radke. EG; Glenn. BS; Braun. JM; Cooper. GS. (2019b). Phthalate exposure and female reproductive and developmental outcomes: A systematic review of the human epidemiological evidence [Review], Environ Int 130: 104580. http://dx.doi.Org/10.1016/i.envint.2019.02.003 Radke. EG: Yost. EE: Roth. N: Sathyanaravana. S: Whalev. P. (2020b). Application of US EPA IRIS systematic review methods to the health effects of phthalates: Lessons learned and path forward [Editorial], Environ Int 145: 105820. http://dx.doi.org/10.1016/i.envint.2020.105820 Saillenfait. AM: Sabate. JP; Denis. F; Antoine. G: Robert. A: Roudot. AC: Ndiave. D; Eliarrat. E. (2017). Evaluation of the effects of a-cypermethrin on fetal rat testicular steroidogenesis. Reprod Toxicol 72: 106-114. http://dx.doi.Org/10.1016/i.reprotox.2017.06.133 Saillenfait. AM: Sabate. JP: Gallissot. F. (2006). Developmental toxic effects of diisobutyl phthalate, the methyl-branched analogue of di-n-butyl phthalate, administered by gavage to rats. Toxicol Lett 165: 39-46. http://dx.doi.Org/10.1016/i.toxlet.2006.01.013 Saillenfait. AM: Sabate. JP: Gallissot. F. (2008). Diisobutyl phthalate impairs the androgen-dependent reproductive development of the male rat. Reprod Toxicol 26: 107-115. http://dx.doi.Org/10.1016/i.reprotox.2008.07.006 Schlumpf. M; Kypke. K; Wittassek. M; Angerer. J; Mascher. H; Mascher. D; Yokt. C: Birchler. M; Lichtensteiger. W. (2010). Exposure patterns of UV filters, fragrances, parabens, phthalates, organochlor pesticides, PBDEs, and PCBs in human milk: correlation of UV filters with use of cosmetics. Chemosphere 81: 1171-1183. http://dx.doi.Org/10.1016/i.chemosphere.2010.09.079 Schwartz. CL; Christiansen. S: Hass. U; Ramhai. L; Axelstad. M; Lobl. NM; Svingen. T. (2021). On the use and interpretation of areola/nipple retention as a biomarker for anti-androgenic effects in rat toxicity studies [Review], Front Toxicol 3: 730752. https://heronet.epa.gov/heronet/index.cfm/reference/download/reference id/10492323 Sedha. S: Gautam. AK; Verma. Y; Ahmad. R; Kumar. S. (2015). Determination of in vivo estrogenic potential of Di-isobutyl phthalate (DIBP) and Di-isononyl phthalate (DINP) in rats. Environ Sci Pollut Res Int 22: 18197-18202. http://dx.doi.org/10.1007/sll356-015-5021-6 Shin. HM; Bennett. DH; Barkoski. J; Ye. X; Calafat. AM; Tancredi. D; Hertz-Picciotto. I. (2019). Variability of urinary concentrations of phthalate metabolites during pregnancy in first morning voids and pooled samples. Environ Int 122: 222-230. http://dx.doi.Org/10.1016/i.envint.2018.l 1.012 Spade. DJ; Hall. SJ; Saffarini. C; Huse. SM; McDonnell. EV; Boekelheide. K. (2014). Differential response to abiraterone acetate and di-n-butyl phthalate in an androgen-sensitive human fetal testis xenograft bioassay. Toxicol Sci 138: 148-160. http://dx.doi.org/10.1093/toxsci/kft266 Sterne. JAC; Hernan. MA; Reeves. BC; Savovic. J; Berkman. ND; Viswanathan. M; Henry. D; Altman. DG; Ansari. MT; Boutron. I; Carpenter. JR; Chan. AW; Churchill. R; Peeks. JJ; Hrobiartsson. A; Kirkham. J; Jtini. P; Loke. YK; Pigott. TP; Ramsay. CR; Regidor. D; Rothstein, HR; Sandhu, L; Santaguida, PL; Schiinemann, HJ; Shea, B; Shrier, I; Tugwell, P; Turner, L; Valentine, JC; Waddington, H; Waters, E; Wells, GA; Whiting, PF; Higgins, JPT. (2016). ROBINS-I: A tool for assessing risk of bias in non-randomised studies of interventions. BMJ 355: i4919. http://dx.doi. org/10.113 6/bmi ,i4919 Swan. SH. (2008). Environmental phthalate exposure in relation to reproductive outcomes and other health endpoints in humans [Review], Environ Res 108: 177-184. http://dx.doi.Org/10.1016/i.envres.2008.08.007 Swan. SH; Sathyanaravana. S; Barrett. ES; Janssen. S; Liu. F; Nguyen. RH; Redmon. JB; Team. TS. (2015). First trimester phthalate exposure and anogenital distance in newborns. Hum Reprod 30: 963-972. http://dx.doi.org/10.1093/humrep/deu363 Thompson. CJ; Ross. SM; Henslev. J; Liu. K; Heinze. SC; Young. SS; Gaido. KW. (2005). Pifferential steroidogenic gene expression in the fetal adrenal gland versus the testis and rapid and dynamic Page 63 of 94 ------- 1807 1808 1809 1810 1811 1812 1813 1814 1815 1816 1817 1818 1819 1820 1821 1822 1823 1824 1825 1826 1827 1828 1829 1830 1831 1832 1833 1834 1835 1836 1837 1838 1839 1840 1841 1842 1843 1844 1845 1846 1847 1848 1849 1850 1851 1852 1853 1854 1855 PUBLIC RELEASE DRAFT December 2024 response of the fetal testis to di(n-butyl) phthalate. Biol Reprod 73: 908-917. http://dx.doi.org/10.1095/biolreprod.105.042382 U.S. CPSC. (2011). Toxicity review of diisobutyl phthalate (DiBP, CASRN 84-69-5). Bethesda, MD: U.S. Consumer Product Safety Commission, https://www.cpsc.gov/s3fs- publi c/T oxi cityRevi ewOfDiBP. pdf U.S. CPSC. (2014). Chronic Hazard Advisory Panel on Phthalates and Phthalate Alternatives (with appendices). Bethesda, MD: U.S. Consumer Product Safety Commission, Directorate for Health Sciences. https://www.cpsc.gov/s3fs-public/CHAP-REPORT-With-Appendices.pdf U.S. EPA. (1991). Guidelines for developmental toxicity risk assessment. Fed Reg 56: 63798-63826. U.S. EPA. (1993). Reference Dose (RfD): description and use in health risk assessments background document 1A, March 15, 1993. Washington, DC: U.S. Environmental Protection Agency, Integrated Risk Information System, https://www.epa.gov/iris/reference-dose-rfd-description- and-use-health-risk-assessments U.S. EPA. (1994). Methods for derivation of inhalation reference concentrations and application of inhalation dosimetry [EPA Report], (EPA600890066F). Research Triangle Park, NC. https://cfpub.epa.gov/ncea/risk/recordisplav.cfm?deid=71993&CFID=51174829&CFTOKEN=2 5006317 U.S. EPA. (1996). Guidelines for reproductive toxicity risk assessment [EPA Report], (EPA/630/R- 96/009). Washington, DC: U.S. Environmental Protection Agency, Risk Assessment Forum. https://nepis.epa. gov/Exe/ZvPURL.cgi?Dockev=3 0004YQB.txt U.S. EPA. (2002a). Hepatocellular hypertrophy. HED guidance document #G2002.01 [EPA Report], Washington, DC. U.S. EPA. (2002b). A review of the reference dose and reference concentration processes. (EPA630P02002F). Washington, DC. https://www.epa.gov/sites/production/files/2014- 12/documents/rfd-final.pdf U.S. EPA. (201 la). Exposure factors handbook: 2011 edition [EPA Report], (EPA/600/R-090/052F). Washington, DC: U.S. Environmental Protection Agency, Office of Research and Development, National Center for Environmental Assessment. https://nepis.epa. gov/Exe/ZvPURL.cgi?Dockev=P100F2QS.txt U.S. EPA. (201 lb). Exposure factors handbook: 2011 edition (final) (EPA/600/R-090/052F). Washington, DC. http://cfpub.epa.gov/ncea/cfm/recordisplav.cfm?deid=236252 U.S. EPA. (2011c). Recommended use of body weight 3/4 as the default method in derivation of the oral reference dose. (EPA100R110001). Washington, DC. https://www.epa.gov/sites/production/files/2013-09/documents/recommended-use-of-bw34.pdf U.S. EPA. (2012). Benchmark dose technical guidance [EPA Report], (EPA100R12001). Washington, DC: U.S. Environmental Protection Agency, Risk Assessment Forum. https://www.epa.gov/risk/benchmark-dose-technical-guidance U.S. EPA. (2020a). Draft Scope of the Risk Evaluation for Di-isobutyl Phthalate (1,2- Benzenedicarboxylic acid, l,2-bis(2-methylpropyl) ester) CASRN 84-69-5. (EPA-740-D-20- 018). https://www.epa.gov/sites/production/files/2020-Q4/documents/casrn-84-69- 5 diisobutyl phthalate draft scope 4-15-2020.pdf U.S. EPA. (2020b). Final scope of the risk evaluation for di-isobutyl phthalate (1,2-benzenedicarboxylic acid, l,2-bis(2-methylpropyl) ester); CASRN 84-69-5 [EPA Report], (EPA-740-R-20-018). Washington, DC: Office of Chemical Safety and Pollution Prevention. https://www.epa.gov/sites/default/files/2020-09/documents/casrn 84-69-5 di- isobutyl phthalate final scope.pdf U.S. EPA. (2021). Draft systematic review protocol supporting TSCA risk evaluations for chemical substances, Version 1.0: A generic TSCA systematic review protocol with chemical-specific methodologies. (EPA Document #EPA-D-20-031). Washington, DC: Office of Chemical Safety Page 64 of 94 ------- 1856 1857 1858 1859 1860 1861 1862 1863 1864 1865 1866 1867 1868 1869 1870 1871 1872 1873 1874 1875 1876 1877 1878 1879 1880 1881 1882 1883 1884 1885 1886 1887 1888 1889 1890 1891 1892 1893 1894 1895 1896 1897 1898 1899 1900 1901 1902 1903 1904 PUBLIC RELEASE DRAFT December 2024 and Pollution Prevention. https://www.regulations.gov/document/EPA-HQ-OPPT-2021-0414- 0005 U.S. EPA. (2022). ORD staff handbook for developing IRIS assessments [EPA Report], (EPA 600/R- 22/268). Washington, DC: U.S. Environmental Protection Agency, Office of Research and Development, Center for Public Health and Environmental Assessment. https://cfpub.epa.gov/ncea/iris drafts/recordisplav.cfm?deid=356370 U.S. EPA. (2023a). Draft Proposed Approach for Cumulative Risk Assessment of High-Priority Phthalates and a Manufacturer-Requested Phthalate under the Toxic Substances Control Act. (EPA-740-P-23-002). Washington, DC: U.S. Environmental Protection Agency, Office of Chemical Safety and Pollution Prevention. https://www.regulations.gov/document/EPA-HQ- QPPT-2022-0918-0009 U.S. EPA. (2023b). Science Advisory Committee on Chemicals meeting minutes and final report, No. 2023-01 - A set of scientific issues being considered by the Environmental Protection Agency regarding: Draft Proposed Principles of Cumulative Risk Assessment (CRA) under the Toxic Substances Control Act and a Draft Proposed Approach for CRA of High-Priority Phthalates and a Manufacturer-Requested Phthalate. (EPA-HQ-OPPT-2022-0918). Washington, DC: U.S. Environmental Protection Agency, Office of Chemical Safety and Pollution Prevention. https://www.regulations.gov/document/EPA-HQ-OPPT-2022-0918-0Q67 U.S. EPA. (2024a). Draft Data Extraction Information for Environmental Hazard and Human Health Hazard Animal Toxicology and Epidemiology for Diisobutyl Phthalate (DIBP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2024b). Draft Data Quality Evaluation Information for Human Health Hazard Animal Toxicology for Diisobutyl Phthalate (DIBP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2024c). Draft Data Quality Evaluation Information for Human Health Hazard Epidemiology for Diisobutyl Phthalate (DIBP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2024d). Draft Meta-Analysis and Benchmark Dose Modeling of Fetal Testicular Testosterone for Di(2-ethylhexyl) Phthalate (DEHP), Dibutyl Phthalate (DBP), Butyl Benzyl Phthalate (BBP), Diisobutyl Phthalate (DIBP), and Dicyclohexyl Phthalate (DCHP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2024e). Draft Non-cancer Human Health Hazard Assessment for Butyl benzyl phthalate (BBP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2024f). Draft Non-cancer Human Health Hazard Assessment for Dibutyl Phthalate (DBP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2024g). Draft Non-Cancer Human Health Hazard Assessment for Dicyclohexyl Phthalate (DCHP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2024h). Draft Non-cancer Human Health Hazard Assessment for Diethylhexyl Phthalate (DEHP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2024i). Draft Non-cancer Human Health Hazard Assessment for Diisobutyl phthalate (DIBP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2024j). Draft Risk Evaluation for Diisobutyl Phthalate (DIBP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2024k). Draft Systematic Review Protocol for Diisobutyl Phthalate (DIBP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (20241). Draft Technical Support Document for the Cumulative Risk Analysis of Di(2- ethylhexyl) Phthalate (DEHP), Dibutyl Phthalate (DBP), Butyl Benzyl Phthalate (BBP), Diisobutyl Phthalate (DIBP), Dicyclohexyl Phthalate (DCHP), and Diisononyl Phthalate (DINP) Under the Toxic Substances Control Act (TSCA). Washington, DC: Office of Chemical Safety and Pollution Prevention. Page 65 of 94 ------- 1905 1906 1907 1908 1909 1910 1911 1912 1913 1914 1915 1916 1917 1918 1919 1920 1921 1922 1923 1924 1925 1926 1927 1928 1929 1930 1931 1932 1933 1934 1935 1936 1937 1938 1939 1940 1941 1942 1943 1944 PUBLIC RELEASE DRAFT December 2024 U.S. EPA. (2024m). Science Advisory Committee on Chemicals Meeting Minutes and Final Report No. 2024-2, Docket ID: EPA-HQ-OPPT-2024-0073: For the Draft Risk Evaluation for Di-isodecyl Phthalate (DIDP) and Draft Hazard Assessments for Di-isononyl Phthalate (DINP). Washington, DC: U.S. Environmental Protection Agency, Science Advisory Committee on Chemicals. U.S. EPA. (2025a). Draft Cancer Human Health Hazard Assessment for Di(2-ethylhexyl) Phthalate (DEHP), Dibutyl Phthalate (DBP), Butyl Benzyl Phthalate (BBP), Diisobutyl Phthalate (DIBP), and Dicyclohexyl Phthalate (DCHP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2025b). Draft Consumer and Indoor Dust Exposure Assessment for Diisobutyl phthalate (DIBP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2025c). Draft Environmental Release and Occupational Exposure Assessment for Diisobutyl phthalate (DIBP). Washington, DC: Office of Pollution Prevention and Toxics. U.S. EPA. (2025d). Non-Cancer Human Health Hazard Assessment for Diisononyl Phthalate (DINP) Washington, DC: Office of Pollution Prevention and Toxics. University of Rochester. (1953). One month feeding tests of di-isobutyl phthalate with cover letter [TSCA Submission], (OTS0205995. 878212229. TSCATS/017429). Confidential. https://ntrl.ntis.gov/NTRL/dashboard/searchResults/titleDetail/QTS0205995.xhtml van Den Driesche, S; McKinnell, C; Calarrao, A; Kennedy, L; Hutchison, GR; Hrabalkova, L; Jobling, MS; Macpherson, S; Anderson, RA; Sharpe, RM; Mitchell, RT. (2015). Comparative effects of di(n-butyl) phthalate exposure on fetal germ cell development in the rat and in human fetal testis xenografts. Environ Health Perspect 123: 223-230. http://dx.doi.org/10.1289/ehp. 1408248 Wang, X; Sheng, N; Cui, R; Zhang, H; Wang, J; Dai, J. (2017). Gestational and lactational exposure to di-isobutyl phthalate via diet in maternal mice decreases testosterone levels in male offspring. Chemosphere 172: 260-267. http://dx.doi.org/10.1016/i.chemosphere.2017.01.011 Welsh, M; Saunders, PTK; Fisken, M; Scott, HM; Hutchison, GR; Smith, LB; Sharpe, RM. (2008). Identification in rats of a programming window for reproductive tract masculinization, disruption of which leads to hypospadias and cryptorchidism. J Clin Invest 118: 1479-1490. http: //dx. doi. or g/10.1172/i ci34241 Wenzel, AG; Bloom, MS; Butts, CD; Wineland, RJ; Brock, JW; Cruze, L; Unal, ER; Kucklick, JR; Somerville, SE; Newman, RB. (2018). Influence of race on prenatal phthalate exposure and anogenital measurements among boys and girls. Environ Int 110: 61-70. http://dx.doi.Org/10.1016/i.envint.2017.10.007 Yang, TC; Peterson, KE; Meeker, JD; Sanchez, BN; Zhang, Z; Cantoral, A; Solano, M; Tellez-Rojo, MM. (2018). Exposure to Bisphenol A and phthalates metabolites in the third trimester of pregnancy and BMI trajectories. Pediatr Obes 13: 550-557. http://dx.doi.org/10. Ill 1/ijpo. 12279 Yost, EE; Euling, SY; Weaver, JA; Beverly, BEJ; Keshava, N; Mudipalli, A; Arzuaga, X; Blessinger, T; Dishaw, L; Hotchkiss, A; Makris, SL. (2019). Hazards of diisobutyl phthalate (DIBP) exposure: A systematic review of animal toxicology studies [Review], Environ Int 125: 579-594. http://dx.doi.Org/10.1016/i.envint.2018.09.038 Page 66 of 94 ------- PUBLIC RELEASE DRAFT December 2024 1945 1946 1947 1948 1949 1950 1951 APPENDICES Appendix A Existing Assessments of DIBP The available existing assessments of DIBP are summarized in Table Apx A-l, which includes details regarding external peer-review, public consultation, and systematic review protocols that were used. Table Apx A-l. Summary of Peer-review, Public Comments, and Systematic Review for Existing Assessments of DIBP Agency Assessment(s) (Reference) External Peer- Review? Public Consultation? Systematic Review Protocol Employed? Remarks U.S. EPA (Publications by the Center for Public Health and Environmental Assessment [CPHEA] within the Office of Research and Development [ORD]) Phthalate exposure and male reproductive outcomes: A systematic review of the human epidemiological evidence (Radke et al.. 2018) No No Yes Phthalate exposure andfemale reproductive and developmental outcomes: A systematic review of the human epidemiological evidence (Radke et al.. 2019b) Phthalate exposure and metabolic effects: A systematic review of the human epidemiological evidence (Radke et al.. 2019a) Phthalate exposure and neurodevelopment: A systematic review and meta-analysis of human epidemiological evidence (Radke et al.. 2020a). Hazards of diisobutylphthalate (DIBP Exposure): A systematic - Publications were subjected to peer- review prior to being published in a special issue of the journal Environment International - Publications employed a systematic review process that included literature search and screening, study evaluation, data extraction, and evidence synthesis. The full systematic review protocol is available as a supplemental file associated with each publication. Page 67 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Agency Assessment(s) (Reference) External Peer- Review? Public Consultation? Systematic Review Protocol Employed? Remarks review of animal toxicology studies (Yost et al., 2019) U.S. CPSC Toxicity review of diisobutyl phthalate (DiBP, CASRN 84-69- 5) (U.S. CPSC. 2011) Yes Yes No - Peer-reviewed by panel of four experts. Peer review report available at: httDs://www.cDsc.sov/s3fs-Dublic/Peer- Chronic Hazard Advisory Panel on Phthalates and Phthalate Alternatives (U.S. CPSC, 2014) Review-Report-Comments.pdf -Public comments available at: https://www.cpsc.sov/chap - No formal systematic review protocol employed. - Details regarding CPSC's strategy for identifying new information and literature are provided on case 12 of (U.S. CPSC, 2014) NASEM Application of systematic review methods in an overall strategy for evaluating low-dose toxicity fi'om endocrine active chemicals mASEM. 2017) Yes No Yes - Draft report was reviewed by individuals chosen for their diverse perspectives and technical expertise in accordance with the National Academies peer-review process. See Acknowledgements section of (NASEM, 2017) for more details. - Employed NTP's Office of Heath Assessment and Translation (OHAT) systematic review method Health Canada State of the science report: Phthalate substance grouping: Medium-chain phthalate esters: Chemical Abstracts Service Registry Numbers: 84-61-7; 84- Yes Yes No (Animal studies) - Ecological and human health portions of the screening assessment report (ECCC/HC, 2020) were subiect to external review and/or consultation. See Page 68 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Agency Assessment(s) (Reference) External Peer- Review? Public Consultation? Systematic Review Protocol Employed? Remarks 64-0; 84-69-5; 523-31-9; 5334- 09-8; 16883-83-3; 27215-22-1; 27987-25-3; 68515-40-2; 71888- 89-6 (EC/HC. 2015b) Supporting documentation: Evaluation of epidemiologic studies on phthalate compounds and their metabolites for hormonal effects, growth and development and reproductive parameters (Health Canada, Yes (Epidemiologic studies) case 2 of (ECCC/HC, 2020) for additional details. - State of the science report (EC/HC, 2015 a) and draft screening assessment report for the phthalate substance group subjected to 60-day public comment periods. Summaries of received public comments available at: httos ://www. Canada, ca/ en/heal th- canada/ servi ce s/chemi cal - substances/substance-srouDinss- 2018b) Supporting documentation: Evaluation of epidemiologic studies on phthalate compounds and their metabolites for effects on behaviour and neurodevelopment, allergies, cardiovascular function, oxidative stress, breast cancer, obesity, and metabolic disorders (Health Canada, 2018a) Screening Assessment - Phthalate Substance Grouping (ECCC/HC, 2020) initi ative/phthal ate. html#a 1 - No formal systematic review protocol employed to identify or evaluate experimental animal toxicology studies. - Details regarding Health Canada's strategy for identifying new information and literature are provided in Section 1 of (EC/HC. 2015a) and (ECCC/HC. 2020) - Human epidemiologic studies evaluated usins Downs and Black Method (Health Canada, 2018a, b) NICNAS Existing chemical hazard assessment report: Diisobiityl phthalate (NICNAS, 2008a) No Yes No - No details regarding peer-review are provided. - No formal systematic review protocol employed. Page 69 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Agency Assessment(s) (Reference) External Peer- Review? Public Consultation? Systematic Review Protocol Employed? Remarks - No details regarding how NICNAS identified literature for inclusion in its assessment are provided. ECHA Committee for Risk Assessment (RAC) Opinion on an Annex XV dossier proposing restrictions on four vhthalates (ECHA, 2012b) Committee for Risk Assessment (RAC) Committee for Socio- economic Analysis (SEAC): Background document to the Opinion on the Annex XV dossier proposing restrictions on four phthalates (ECHA, 2012a) Opinion on an Annex XV dossier proposing restrictions on four phthalates (DEHP, BBP, DBF, DIBP) (ECHA. 2017b) Annex to the Background document to the Opinion on the Annex XV dossier proposing restrictions on four phthalates (DEHP, BBP, DBP, DIBP) (ECHA. 2017a) Yes Yes No - Peer-reviewed by ECHA's Committee for Risk Assessment (RAC) - Subject to public consultation - No formal systematic review protocol employed. 1952 Page 70 of 94 ------- 1953 1954 1955 1956 1957 1958 1959 1960 1961 1962 1963 1964 1965 1966 1967 1968 1969 1970 1971 1972 1973 1974 1975 1976 1977 1978 1979 PUBLIC RELEASE DRAFT December 2024 Appendix B Fetal Testicular Testosterone as an Acute Effect No studies of experimental animal models are available that investigate the antiandrogenic effects of DIBP following single dose, acute exposures. However, there are studies of its isomer, dibutyl phthalate (DBP) available that indicate a single acute exposure during the critical window of development (i.e., GD 15.5 to GD 18.5 in rats) can reduce fetal testicular testosterone production and disrupt testicular steroidogenic gene expression. Two studies were identified that demonstrate single doses of 500 mg/kg DBP can reduce fetal testicular testosterone and steroidogenic gene expression. Johnson et al. (2012; 2011) gavaged pregnant SD rats with a single dose of 500 mg/kg DBP on GD 19 and observed reductions in steroidogenic gene expression in the fetal testes three (Cypl7al) to six (Cypllal, StAR) hours post-exposure, while fetal testicular testosterone was reduced starting 18 hours post-exposure. Similarly, Thompson et al. (2005) reported a 50 percent reduction in fetal testicular testosterone 1-hour after pregnant SD rats were gavaged with a single dose of 500 mg/kg DBP on GD 19, while changes in steroidogenic gene expression occurred 3 (StAR) to 6 (Cypllal, Cypl7al, Scarbl) hours post-exposure, and protein levels of these genes were reduced 6 to 12 hours post-exposure. Additionally, studies by Carruthers et al. (2005) further demonstrate that exposure to as few as two oral doses of 500 mg/kg DBP on successive days between GDs 15 to 20 can reduce male pup AGD, cause permanent nipple retention, and increase the frequency of reproductive tract malformations and testicular pathology in adult rats that received two doses of DBP during the critical window. Studies of DBP provide evidence to support use of effects on fetal testosterone and the developing male reproductive system consistent with phthalate syndrome as an acute effect. However, the database is limited to just a few DBP studies that test relatively high (500 mg/kg) single doses of DBP. Although there are no single exposure studies of DIBP that evaluate anti androgenic effects on the developing male reproductive system, there are three studies that have evaluated effects on fetal testicular testosterone production and steroidogenic gene expression following daily gavage doses of 100 to 900 mg/kg-day DIBP from GDs 14 to 18 (5 total doses) (Gray et al.. 2021; Furr et al.. 2014; Hannas et al.. 2012; Hannas et al.. 2011). all of which consistently report anti androgenic effects at 300 mg/kg-day DIBP. Page 71 of 94 ------- 1980 1981 1982 1983 1984 1985 1986 1987 1988 1989 1990 1991 1992 1993 1994 1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016 2017 2018 2019 2020 2021 2022 2023 2024 2025 2026 PUBLIC RELEASE DRAFT December 2024 Appendix C Calculating Daily Oral Human Equivalent Doses and Human Equivalent Concentrations For DIBP, all data considered for PODs are obtained from oral animal toxicity studies in rats or mice. Because toxicity values for DIBP are from oral animal studies, EPA must use an extrapolation method to estimate HEDs. The preferred method would be to use chemical-specific information for such an extrapolation. However, no PBPK models or chemical-specific information was identified for DIBP to support a quantitative extrapolation. In the absence of such data, EPA relied on the guidance from U.S. EPA (2011c). which recommends scaling allometrically across species using the three-quarter power of body weight (BW34) for oral data. Allometric scaling accounts for differences in physiological and biochemical processes, mostly related to kinetics. For application of allometric scaling in risk evaluations, EPA uses dosimetric adjustment factors (DAFs), which can be calculated using EquationApx C-l. EquationApx C-l. Dosimetric Adjustment Factor /BWa\1/4 Where: DAF = Dosimetric adjustment factor (unitless) BWa = Body weight of species used in toxicity study (kg) BWh = Body weight of adult human (kg) U.S. EPA (2011c). presents DAFs for extrapolation to humans from several species. However, because those DAFs used a human body weight of 70 kg, EPA has updated the DAFs using a human body weight of 80 kg for the DIBP risk evaluation (U.S. EPA. 2011a). EPA used the body weights of 0.025 and 0.25 kg for mice and rats, respectively, as presented in U.S. EPA (2011c). The resulting DAFs for mice and rats are 0.133 and 0.236, respectively. Use of allometric scaling for oral animal toxicity data to account for differences among species allows EPA to decrease the default intraspecies UF (UFa) used to set the benchmark MOE; the default value of 10 can be decreased to 3, which accounts for any toxicodynamic differences that are not covered by use of BW34. Using the appropriate DAF from EquationApx C-l, EPA adjusts the POD to obtain the HED using Equation Apx C-2: Equation Apx C-2. Daily Oral Human Equivalent Dose Where: HEDDaily PODDaily DAF HEDDaily PODDaiiy X DAF Human equivalent dose assuming daily doses (mg/kg-day) Oral POD assuming daily doses (mg/kg-day) Dosimetric adjustment factor (unitless) For this draft risk evaluation, EPA assumes similar absorption for the oral and inhalation routes, and no adjustment was made when extrapolating to the inhalation route. For the inhalation route, EPA extrapolated the daily oral HEDs to inhalation HECs using a human body weight and breathing rate relevant to a continuous exposure of an individual at rest, as follows: Equation Apx C-3. Extrapolating from Oral HED to Inhalation HEC Page 72 of 94 ------- 2027 2028 2029 2030 2031 2032 2033 2034 2035 2036 2037 2038 2039 2040 2041 2042 2043 2044 2045 2046 2047 2048 2049 2050 2051 2052 2053 2054 2055 2056 2057 2058 2059 2060 2061 2062 2063 2064 2065 2066 2067 PUBLIC RELEASE DRAFT December 2024 jirn r BWh HECDaily,continuous ~ H E D Daily ^ ^JR * ED ^ Where: HECDaily, continuous = Inhalation HEC based on continuous daily exposure (mg/m3) HEDDaiiy = Oral HED based on daily exposure (mg/kg-day) BWh = Body weight of adult humans (kg) = 80 IRr = Inhalation rate for an individual at rest (m3/hr) = 0.6125 EDc = Exposure duration for a continuous exposure (hr/day) = 24 Based on information from U.S. EPA (2011a). EPA assumes an at rest breathing rate of 0.6125 m3/hr. Adjustments for different breathing rates required for individual exposure scenarios are made in the exposure calculations, as needed. It is often necessary to convert between ppm and mg/m3 due to variation in concentration reporting in studies and the default units for different OPPT models. Therefore, EPA presents all PODs in equivalents of both units to avoid confusion and errors. EquationApx C-4 presents the conversion of the HEC from mg/m3 to ppm. Equation Apx C-4. Converting Units for HECs (mg/m3 to ppm) mg 24.45 X ppm = Y 5- x m3 MW Where: 24.45 = Molar volume of a gas at standard temperature and pressure (L/mol), default MW = Molecular weight of the chemical (MW of DIBP = 278.35 g/mol) C.l DIBP Non-cancer HED and HEC Calculations for Acute, Intermediate, and Chronic Duration Exposures The acute, intermediate, and chronic duration non-cancer POD is based on a BMDLs of 24 mg/kg-day and the critical effect is decreased fetal testicular testosterone. The POD was derived from three gestational exposure studies of rats (Gray et al.. 2021: Hannas et al.. 2011: Howdeshell et al.. 2008). This non-cancer POD is considered protective of effects observed following acute, intermediate, and chronic duration exposures to DIBP. EPA used Equation Apx C-l to determine a DAF specific to rats (0.236), which was in turn used in the following calculation of the daily HED using Equation Apx C-2: mq mq 5.66 = 24- X 0.236 kg day kg day EPA then calculated the continuous HEC for an individual at rest using Equation Apx C-3: mq mq 80 kq 30.9 ^ = 5.66 2- x ( = ) m kg day 0.6125* 24 hr hr Equation Apx C-4 was used to convert the HEC from mg/m3 to ppm: Page 73 of 94 ------- PUBLIC RELEASE DRAFT December 2024 ma 24.45 2068 2.71 ppm = 30.9 - X m3 278.35 g/mol Page 74 of 94 ------- 2069 2070 2071 2072 2073 2074 2075 2076 2077 2078 2079 2080 2081 2082 2083 2084 2085 2086 2087 2088 2089 2090 2091 2092 2093 2094 2095 2096 2097 2098 2099 2100 2101 2102 2103 2104 2105 2106 2107 2108 2109 2110 2111 2112 PUBLIC RELEASE DRAFT December 2024 Appendix D Considerations for Benchmark Response (BMR) Selection for Reduced Fetal Testicular Testosterone D.l Purpose EPA has conducted an updated meta-analysis and benchmark dose modeling (BMD) analysis of decreased fetal rat testicular testosterone (U.S. EPA. 2024d). During the July 2024 Science Advisory Committee on Chemicals (SACC) peer-review meeting of the draft risk evaluation of diisodecyl phthalate (DIDP) and draft human health hazard assessments for diisononyl phthalate (DINP), the SACC recommended that EPA should clearly state its rational for selection of benchmark response (BMR) levels evaluated for decreases in fetal testicular testosterone relevant to the single chemical assessments (U.S. EPA. 2024m). This appendix describes EPA's rationale for evaluating BMRs of 5, 10, and 40 percent for decreases in fetal testicular testosterone. {Note: EPA will assess the relevant BMR for deriving relative potency factors to be used in the draft cumulative risk assessment separately fi'om this analysis.) D.2 Methods As described in EPA's Benchmark Dose Technical Guidance (U.S. EPA. 2012). "Selectinga BMR(s) involves making judgments about the statistical and biological characteristics of the dataset and about the applications for which the resulting BMDs BMDLs will be used. " For the updated meta-analysis and BMD modeling analysis of fetal rat testicular testosterone, EPA evaluated BMR values of 5, 10, and 40 percent based on both statistical and biological considerations (U.S. EPA. 2024d). In 2017, NASEM (2017) modeled BMRs of 5 and 40 percent for decreases in fetal testicular testosterone. NASEM did not provide explicit justification for selection of a BMR of 5 percent. However, justification for the BMR of 5 can be found elsewhere. As discussed in EPA's Benchmark Dose Technical Guidance (U.S. EPA. 2012). a BMR of 5 percent is supported in most developmental and reproductive studies. Comparative analyses of a large database of developmental toxicity studies demonstrated that developmental NOAELs are approximately equal to the BMDLs (Allen et al.. 1994a. b; Faustman et al.. 1994). EPA also evaluated a BMR of 10 percent as part of the updated BMD analysis. BMD modeling of fetal testosterone conducted by NASEM (2017) indicated that BMDs estimates are below the lowest dose with empirical testosterone data for several of the phthalates (e.g., DIBP). As discussed in EPA's Benchmark Dose Technical Guidance (U.S. EPA. 2012) "For some datasets the observations may correspond to response levels far in excess of a selected BMR and extrapolation sufficiently below the observable range may be too uncertain to reliably estimate BMDsBMDLs for the selected BMR. " Therefore, EPA modelled a BMR of 10 percent because datasets for some of the phthalates may not include sufficiently low doses to support modeling of a 5 percent response level. NASEM (2017) also modeled a BMR of 40 percent using the following justification: "previous studies have shown that reproductive-tract malformations were seen in male rats when fetal testosterone production was reduced by about 40% ^Grav et al.. 2016; Howdeshell et al.. 2015)." Further description of methods and results for the updated meta-analysis and BMD modeling analysis that evaluated BMRs of 5, 10, and 40 percent for decreased fetal testicular testosterone are provided in EPA's Draft Meta-Analysis and Benchmark Dose Modeling of Fetal Testicular Testosterone for Di (2- Page 75 of 94 ------- 2113 2114 2115 2116 2117 2118 2119 2120 2121 2122 2123 2124 2125 2126 2127 2128 2129 2130 2131 2132 2133 2134 2135 2136 2137 2138 2139 2140 2141 2142 2143 2144 2145 2146 2147 2148 2149 2150 2151 2152 2153 2154 2155 2156 2157 2158 PUBLIC RELEASE DRAFT December 2024 ethylhexyl) Phthalate (DEHP), Dibutyl Phthalate (DBP), Butyl Benzyl Phthalate (BBP), Diisobutyl Phthalate (DIBP), andDicyclohexylPhthalate (DCHP) (U.S. EPA. 2024cT). D.3 Results BMD estimates, as well as 95 percent upper and lower confidence limits, for decreased fetal testicular testosterone for the evaluated BMRs of 5, 10, and 40 percent are shown in TableApx D-l. BMDs estimates ranged from 8.4 to 74 mg/kg-day for DEHP, DBP, DCHP, and DINP; however, a BMDs estimate could not be derived for BBP or DIBP. Similarly, BMDio estimates ranged from 17 to 152 for DEHP, DBP, DCHP, DIBP and DINP; however, a BMDio estimate could not be derived for BBP. BMD40 estimates were derived for all phthalates (i.e., DEHP, DBP, DCHP, DIBP, BBP, DINP) and ranged from 90 to 699 mg/kg-day. In the mode of action (MOA) for phthalate syndrome, which is described elsewhere (U.S. EPA. 2023a) and in Section 3.1.2 of this document, decreased fetal testicular testosterone is an early, upstream event in the MOA that precedes downstream apical outcomes such as male nipple retention, decrease anogenital distance, and reproductive tract malformations. Decreased fetal testicular testosterone should occur at lower or equal doses than downstream apical outcomes associated with a disruption of androgen action. Because the lower 95 percent confidence limit on the BMD, or BMDL, is used for deriving a point of departure (POD), EPA compared BMDL estimates at the 5, 10, and 40 percent response levels for each phthalate (DEHP, DBP, DCHP, DIBP, BBP, DINP) to the lowest identified apical outcomes associated with phthalate syndrome to determine which response level is protective of downstream apical outcomes. Table Apx D-l provides a comparison of BMD and BMDL estimates for decreased fetal testicular testosterone at BMRs of 5, 10, and 40 percent, the lowest LOAEL(s) for apical outcomes associated with phthalate syndrome, and the POD selected for each phthalate for use in risk characterization. As can be seen from Table Apx D-l, BMDL40 values for DEHP, DBP, DIBP, BBP, DCHP, and DINP are all well above the PODs selected for use in risk characterization for each phthalate by 3X (for BBP) to 25 .4X (for DEHP). Further, BMDL40 values for DEHP, DBP, DIBP, BBP, and DCHP, but not DINP, are above the lowest LOAELs identified for apical outcomes on the developing male reproductive system. These results clearly demonstrate that a BMR of 40 percent is not appropriate for use in human health risk assessment. As can be seen from Table Apx D-l, BMDL10 values for DBP (BMDL10, POD, LOAEL = 20, 9, 30 mg/kg-day, respectively) and DCHP (BMDL10, POD, LOAEL = 12, 10, 20 mg/kg-day, respectively) are slightly higher than the PODs selected for use in risk characterization and slightly less than the lowest LOAELs identified based on apical outcomes associated with the developing male reproductive system. This indicates that a BMR of 10% may be protective of apical outcomes evaluated in available studies for both DBP and DCHP. BMDL10 values could not be derived for DIBP or BBP (Table Apx D-l). Therefore, no comparisons to the POD or lowest LOAEL for apical outcomes could be made for either of these phthalates at the 10 percent response level. For DEHP, the BMDL10 is greater than the POD selected for use in risk characterization by 5X (BMDL10 and POD = 24 and 4.8 mg/kg-day, respectively) and is greater than the lowest LOAEL identified for apical outcomes on the developing male reproductive system by 2.4X (BMDL10 and LOAEL = 24 and 10 mg/kg-day, respectively). This indicates that a BMR of 10 percent for decreased fetal testicular testosterone is not health protective for DEHP. For DEHP, the BMDLs (11 mg/kg-day) is Page 76 of 94 ------- 2159 2160 2161 2162 2163 2164 2165 2166 2167 2168 2169 2170 2171 2172 2173 2174 2175 2176 2177 2178 2179 2180 2181 2182 2183 2184 2185 2186 2187 2188 2189 2190 2191 2192 2193 2194 2195 2196 2197 2198 2199 2200 2201 2202 PUBLIC RELEASE DRAFT December 2024 similar to the selected POD (NOAEL of 4.8 mg/kg-day) and the lowest LOAEL identified for apical outcomes on the developing male reproductive system (10 mg/kg-day). D.4 Weight of Scientific Evidence Conclusion As discussed elsewhere (U.S. EPA. 2023a). DEHP, DBP, BBP, DIBP, DCHP, and DINP are toxicologically similar and induce effects on the developing male reproductive system consistent with a disruption of androgen action. Because these phthalates are toxicologically similar, it is more appropriate to select a single BMR for decreased fetal testicular testosterone to provide a consistent basis for dose response analysis and for deriving PODs relevant to the single chemical assessments. EPA has reached the preliminary conclusion that a BMR of 5 percent is the most appropriate and health protective response level for evaluating decreased fetal testicular testosterone when sufficient dose- response data are available to support modeling of fetal testicular testosterone in the low-end range of the dose-response curve. This conclusion is supported by the following weight of scientific evidence considerations. For DEHP, the BMDLio estimate is greater than the POD selected for use in risk characterization by 5X and is greater than the lowest LOAEL identified for apical outcomes on the developing male reproductive system by 2.4X. This indicates that a BMR of 10 percent is not protective for DEHP. The BMDL5 estimate for DEHP is similar to the selected POD and lowest LOAEL for apical outcomes on the developing male reproductive system. BMDLio estimates for DBP (BMDLio, POD, LOAEL = 20, 9, 30 mg/kg-day, respectively) and DCHP (BMDLio, POD, LOAEL = 12, 10, 20 mg/kg-day, respectively) are slightly higher than the PODs selected for use in risk characterization and slightly less than the lowest LOAELs identified based on apical outcomes associated with the developing male reproductive system. This indicates that a BMR of 10 percent may be protective of apical outcomes evaluated in available studies for both DBP and DCHP. However, this may be a reflection of the larger database of studies and wider range of endpoints evaluated for DEHP, compared to DBP and DCHP. NASEM (2017) modeled a BMR of 40 percent using the following justification: "previous studies hcn'e shown that reproductive-tract malformations were seen in male rats when fetal testosterone production was reduced by about 40% ^Grav et al.. 2016; Howdeshell et al.. 2015)." However, publications supporting a 40 percent response level are relatively narrow in scope and assessed the link between reduced fetal testicular testosterone in SD rats on GD 18 and later life reproductive tract malformations in F1 males. More specifically, Howdeshell et al. (2015) found reproductive tract malformations in 17 to 100 percent of F1 males when fetal testosterone on GD 18 was reduced by approximately 25 to 72 percent, while Gray et al. (2016) found dose-related reproductive alterations in F1 males treated with dipentyl phthalate (a phthalate not currently being evaluated under TSCA) when fetal testosterone was reduced by about 45 percent on GD 18. Although NASEM modeled a BMR of 40 percent based on biological considerations, there is no scientific consensus on the biologically significant response level and no other authoritative or regulatory agencies have endorsed the 40 percent response level as biologically significant for reductions in fetal testosterone. BMDL40 values for DEHP, DBP, DIBP, BBP, DCHP, and DINP are above the PODs selected for use in risk characterization for each phthalate by 3X to 25.4X (Table Apx D-l). BMDL40 values for DEHP, DBP, DIBP, BBP, and DCHP, but not DINP, are above the lowest LOAELs Page 77 of 94 ------- PUBLIC RELEASE DRAFT December 2024 2203 identified for apical outcomes on the developing male reproductive system. These results clearly 2204 demonstrate that a BMR of 40 percent is not health protective. Page 78 of 94 ------- PUBLIC RELEASE DRAFT December 2024 2205 2206 TableApx D-l. Comparison of BMD/BMDL Values Across BMRs of 5%, 10%, and 40% with PODs and LOAELs for Apical Outcomes for DEHP, DBP, DIBP, BBP, DCHP, and DINP Phthalate POD (mg/kg-day) Selected for use in Risk Characterization (Effect) Lowest LOAEL(s) (mg/kg-day) for Apical Effects on the Male Reproductive System BMDS Estimate" (mg/kg-day) [95% CI] BMDio Estimate" (mg/kg-day) [95% CI] BMD40 Estimate" (mg/kg-day) [95% CI] Reference For Further Details on the Selected POD and Lowest Identified LOAEL, DEHP NOAEL = 4.8 (t male RTM in F1 and F2 males) 10 to 15 (NR, | AGD, RTMs) 17 [11, 31] 35 [24, 63] 178 [122, 284] (U.S. EPA. 2024h) DBP BMDL5 = 9 (J, fetal testicular testosterone) 30 (t Testicular Pathology) 14 [9, 27] 29 [20, 54] 149 [101,247] (U.S. EPA. 2024f) DIBP BMDL5 = 24 (J, fetal testicular testosterone) 125 (t Testicular Pathology) _b 55 [NA, 266f 279 [136, 517] (U.S. EPA. 2024i) BBP NOAEL = 50 (phthalate syndrome-related effects) 100 (IAGD) _b _b 284 [150, 481] (U.S. EPA. 2024e) DCHP NOAEL = 10 (phthalate syndrome-related effects) 20 (t Testicular Pathology) 8.4 [6.0, 14] 17 [12, 29] 90 [63, 151] (U.S. EPA. 2024a) DINP BMDL5 = 49 (J, fetal testicular testosterone) 600 (J, sperm motility) 74 [47, 158] 152 [97, 278] 699 [539, 858] (U.S. EPA. 2025d) Abbreviations: AGD = anogenital distance; BMD = benchmark dose; BMDL = lower 95% confidence limit on BMD; CI = 95% confidence interval; LOAEL = lowest observable-adverse-effect level; NOAEL = no observable-adverse-effect level; POD = point of departure; RTM = reproductive tract malformations " The linear-quadratic model provided the best fit (based on lowest AIC) for DEHP, DBP, DIBP, BBP, DCHP, and DINP. h BMD and/or BMDL estimate could not be derived. 2207 Page 79 of 94 ------- 2208 2209 2210 2211 2212 2213 2214 2215 2216 2217 2218 2219 2220 2221 2222 2223 2224 2225 2226 2227 2228 2229 2230 2231 2232 2233 2234 2235 2236 2237 2238 2239 2240 2241 2242 2243 2244 2245 2246 2247 2248 2249 2250 PUBLIC RELEASE DRAFT December 2024 Appendix E BENCHMARK DOSE MODELING OF FETAL TESTICULAR TESTOSTERONE EPA conducted benchmark dose (BMD) modeling of ex vivo fetal testicular testosterone data from three gestational exposure studies of DIBP (Gray et al.. 2021; Hannas et al.. 2011; Howdeshell et al.. 2008). The BMD modeling for continuous data was conducted with the EPA's BMD software (BMDS 3.3.2). All standard BMDS 3.3.2 continuous models that use maximum likelihood (MLE) optimization and profile likelihood-based confidence intervals were used in this analysis. Standard forms of these models (defined below) were run so that auto-generated model selection recommendations accurately reflect current EPA model selection procedures EPA's benchmark Dose Technical Guidance (U.S. EPA. 2012). BMDS 3.3.2 models that use Bayesian fitting procedures and Bayesian model averaging were not applied in this work. Standard BMDS 3.3.2 Models Applied to Continuous Endpoints: Exponential 3-restricted (exp3-r) Exponential 5-restricted (exp5-r) Hill-restricted (hil-r) Polynomial Degree 3-restricted (ply3-r Polynomial Degree 2-restricted (ply2-r) Power-restricted (pow-r) Linear-unrestricted (lin-ur) EPA evaluated benchmark response (BMR) levels of 1 control standard deviation (1 SD) and 5, 10, and 40% relative deviation. Model fit was judged consistent with EPA's benchmark Dose Technical Guidance (U.S. EPA. 2012). An adequate fit was judged based on the %2 goodness-of-fit p-value (p > 0.1), magnitude of the scaled residuals in the vicinity of the BMR, and visual inspection of the model fit. In addition to these three criteria forjudging adequacy of model fit, a determination was made as to whether the variance across dose groups was constant. If a constant variance model was deemed appropriate based on the statistical test provided in BMDS (i.e., Test 2; p-value > 0.05 [note: this is a change from previous versions of BMDS, which required variance p-value > 0.10 for adequate fit]), the final BMD results were estimated from a constant variance model. If the test for homogeneity of variance was rejected (i.e., p-value < 0.05), the model was run again while modeling the variance as a power function of the mean to account for this nonconstant variance. If this nonconstant variance model did not adequately fit the data (i.e., Test 3; p-value < 0.05), the data set was considered unsuitable for BMD modeling. Among all models providing adequate fit, the lowest BMDL was selected if the BMDLs estimated from different adequately fitting models varied >3-fold; otherwise, the BMDL from the model with the lowest AIC was selected. If no model adequately fit the data set using the approach described above, EPA removed the highest dose group and modelled the data again using the approach described above. Table Apx E-l summarizes BMD modeling results for reduced ex vivo fetal testicular testosterone data, while more detailed BMD model results are provided in Appendices E.l through E.3. Page 80 of 94 ------- PUBLIC RELEASE DRAFT December 2024 2251 TableApx E-l. Summary of BMD Model Results for Decreased Ex Vivo Fetal Testicular 2252 Testosterone Data set BMR Best-Fit Model (Variance) BMD (mg/kg- day) BMDL (mg/kg- day) Notes Appendix Containing Results (Grav et al.. 2021) 5% Exponential 3 (Constant) 63 24 E.l (Howdeshell et al., 2008) 5% Hill (Constant) 103 52 E.2 (Hannas et al., 2011) 5% No models adequately fit the data set E.3 2253 2254 E.l BMD Model Results (Gray et al. 2021) 2255 Table Apx E-2. Ex Vivo Feta Rat Testicular Testosterone Data (Gray et al. 2021) Dose (mg/kg-day) N (# of litters) Mean Standard Deviation Notes 0 3 7.972888889 1.465303963 Data for Block 67 rats reported in Supplementary Data file associated with (Gray et al., 2021) 100 3 7.727111111 1.105751094 300 3 5.247777778 1.429576563 600 2 2.082416667 0.659141371 900 2 1.705333333 0.145192592 2257 Page 81 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Table Apx E-3. BM1 ) Mode Results Ex Vivo Fetal Testicular Testosterone (Gray et al. 2021 Models3 Restriction15 Variance BMR = 5% BMR = 10% BMR = 40% BMR = 1 SD P Value AIC BMDS Recommends BMDS Recommendation Notes BMD BMDL BMD BMDL BMD BMDL BMD BMDL Exponentia 13 Restricted Constant 63.12026 24.43019 106.4244 50.18702 334.6495 242.737 124.0844 57.8285 0.4081261 44.84273867 Viable - Recommended Lowest AIC BMDL 3x lower than lowest non-zero dose Exponential 5 Restricted Constant 117.9931 36.28242 161.5201 69.23603 329.678 257.1545 173.525 79.69645 0.9645746 45.05235319 Viable - Alternate BMD/BMDL ratio > 3 Hill Restricted Constant 159.6043 38.88005 195.2869 72.20498 326.5199 255.0622 205.6406 83.34761 0.8074988 45.10974788 Viable - Alternate BMD/BMDL ratio > 3 Polynomial Degree 3 Restricted Constant 50.44101 43.48323 100.882 86.96657 403.528 347.8659 143.1312 102.5143 0.1694486 46.08266114 Viable - Alternate Polynomial Degree 2 Restricted Constant 50.4376 43.48294 100.8752 86.96588 403.5008 347.8635 143.1079 102.5161 0.1694491 46.08265471 Viable - Alternate Power Restricted Constant 50.42151 43.48131 100.843 86.96262 403.3721 347.8505 143.04 102.5174 0.1694501 46.08264079 Viable - Alternate Linear Unrestricted Constant 50.42151 43.48125 100.843 86.96262 403.3721 347.8505 143.04 102.5173 0.1694501 46.08264079 Viable - Alternate AIC = Akaike information criterion; BMD = benchmark dose; BMDL =benchmark dose lower limit; NA = Not Applicable. a Selected Model (bolded and shaded gray). b Restrictions defined in the BMDS 3.3 User Guide. 2259 Page 82 of 94 ------- PUBLIC RELEASE DRAFT December 2024 12 Frequentist Exponential Degree 3 Model with BMR of 0.05 Added Risk for the BMD and 0.95 Lower Confidence Limit for the BMDL 10 2260 2261 2262 2263 £ o +-< LO 3 u Estimated Probability Response at BMD O Data BMD BMDL 100 200 300 400 500 mg/kg-d 600 700 800 900 User Input Info Model frequentist Ewponential degree 3 Model Restriction Restricted Dataset Name Gray et al. (2021) - DIBP Testosterone User notes [Add user notes here] Dose-Response Modi M[dose] = a" ewp(±11 (b1 dose)"d) Variance Model Var[i] = alpha Model Options BMR Type Rel. Dev. BMRF 0.05 Tail Probability - Confidence Level 0.95 Distribution Type Normal Variance Type Constant Model Data Dependent Variable mg/kg-d Independent Variable [Custom] Total # of Observation 5 Adverse Direction Automatic Page 83 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Model Results Benchmark Dose BMD 63.12026361 BMDL 24.43018547 BMDU 136.7436224 AIC 44.84273867 Test4P-value 0.40812612 0.0. F. 2 Model Parameters # of Parameters 4 Variable Estimate Std Error Lower Conf Upper Conf a 8.176712147 0.50528127 7.186379 9.167045 b 0.00183526 2.35E-04 0.001375 0.002296 d 1.377943821 3.25E-01 0.741889 2.013999 log-alpha -0.003820249 1.44E-01 -0.28666 0.279017 Goodness of Fit Dose Size Estimated Median Calc'd Median Observed Mean Estimated 3D Calc'd 3D Observe d 3D Scaled Residual 8.17671215 7.972889 7.972889 0.998092 1.4659 1.4659 -0.959707 100 7.42306676 7.727111 7.727111 0.998092 1.1058 1.10575 0.5276271 900 5.26924478 5.247778 5.247778 0.998092 1.4296 1.42958 -0.037253 600 2.60984715 2.082417 2.082417 0.998092 0.6591 0.65914 -0.747325 900 1.11027296 1.705333 1.705333 0.998092 0.1452 0.14519 0.8431514 Likelihoods of Interest #of Model Log Likelihood" Parameters AIC A1 -17.5251903 6 47.05038 A2 -13.06218817 10 46.12438 A3 -17.5251903 6 47.05038 fitted -18.42136934 4 44.84274 R -31.49407383 2 66.98815 ' Includes additive constant of -11.9462. This constant was not included in the LL derivation prior to BMDS 3.0. 2264 Tests of Interest Test 2'Log(Likeliho od Ratio) Test df p-value 1 36.86377131 6 <0.0001 2 8.926004259 4 0.062976 3 8.926004259 4 0.062976 4 1.792358067 2 0.408126 2265 2266 2267 E.2 BMP Model Results (Howdeshell et al. 2008) Table Apx E-4. Ex Vivo Feta Rat Testicular Testosterone Data (Howdeshell et al. 2008) Dose (mg/kg-day) N (# of litters) Mean Standard Deviation Notes 0 5 5.7 0.290688837 Data from Table 6 in (Howdeshell et al., 2008) 100 8 5.44 0.537401154 300 5 3.4 0.626099034 600 5 2.31 0.782623792 Page 84 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Dose (mg/kg-day) N (# of litters) Mean Standard Deviation Notes 900 2 2.09 1.286934342 2268 Page 85 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Table Apx E-5. BM1 ) Mode Results Ex Vivo Fetal Testicular Testosterone (Howdes iell et al. 2008) Models3 Restriction15 Variance BMR = 5% BMR = 10% BMR = 40% BMR = 1 SD P Value AIC BMDS Recommends BMDS Recommendation Notes BMD BMDL BMD BMDL BMD BMDL BMD BMDL Exponential 3 Restricted Constant 37.18733 28.33677 74.74254 58.22005 345.5016 282.4184 82.49155 57.22709 0.0322184 57.43690601 Questionable Goodness of fit p-value <0.1 BMDL 3x lower than lowest non-zero dose Modeled control response std. dev. >1.5 actual response std. dev. Exponential 5 Restricted Constant 101.588 45.31085 139.2085 77.87386 298.0878 246.6513 139.0035 75.64862 0.6618655 52.75773749 Viable - Alternate Modeled control response std. dev. > 1.5 actual response std. dev. Hill Restricted Constant 102.9819 52.24216 136.2697 82.27878 297.6961 236.3744 135.9319 80.08333 0.9596039 52.56903757 Viable - Recommended Lowest AIC Modeled control response std. dev. >|1.5| actual response std. dev. Polynomial Degree 3 Restricted Constant 56.3685 49.44861 112.737 98.89673 450.9479 395.5888 149.7341 115.3485 0.0035229 62.15461672 Questionable Goodness of fit p-value <0.1 Modeled control response std. dev. >1.5 actual response std. dev. Polynomial Degree 2 Restricted Constant 56.36571 49.44887 112.7314 98.89766 450.9255 395.5908 149.7243 115.3486 0.0035229 62.15461883 Questionable Goodness of fit p-value <0.1 Modeled control response std. dev. >1.5 actual response std. dev. Power Restricted Constant 56.37483 49.44799 112.7497 98.89597 450.9986 395.5839 149.7562 115.3481 0.0035229 62.15461483 Questionable Goodness of fit p-value <0.1 Modeled control response std. dev. >1.5 actual response std. dev. Linear Unrestricted Constant 56.37483 49.448 112.7497 98.89599 450.9986 395.584 149.7562 115.3481 0.0035229 62.15461483 Questionable Goodness of fit p-value <0.1 Modeled control response std. dev. >1.5 actual response std. dev. AIC = Akaike information criterion; BMD = benchmark dose; BMDL =benchmark dose lower limit; NA = Not Applicable. a Selected Model (bolded and shaded gray). b Restrictions defined in the BMDS 3.3 User Guide. 2270 Page 86 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Frequentist Hill Model with BMR of 0.05 Added Risk for the BMD and 0.95 Lower Confidence Limit for the BMDL Estimated Probability Response at BMD O Data BMD BMDL mg/kg-d 2271 2272 User Input Info Model frequentist Hill Model Restriction Restricted Dataset Name Howdeshell (£008) - OIBP Testosterone User notes [Add user notes here] Dose-Response Modi M[dose] = q + v"dose*n/(k*n + dose'n) Variance Model Var[i] = alpha Model Options BMR Type Rel. Dev. BMRF 0.05 Tail Probability - Confidence Level 0.95 Distribution Type Normal Variance Type Constant Model Data Dependent Variable mg/kg-d Independent Variable [Custom] Total # of Observation 5 Adverse Direction Automatic 2274 Page 87 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Model Results Benchmark Dose BMD 102.981862 BMDL 52.24216337 BMDU 185.1065152 AIC 52.56903757 Test4P-value 0.959603873 D.O.F. 1 Model Parameters # of Parameters 5 Variable Estimate Std Error Lower Conl Upper Conl 3 5.702356605 0.24846946 5.215365 6.189348 Y -3.700369546 0.54289795 -4.76443 -2.63631 k 251.0918424 37.2929652 177.999 324.1847 n 2.786041669 1.15019349 0.531704 5.04038 alpha 0.321384979 2.92E-02 0.264127 0.378643 Goodness of Fit Dose Size Estimated Calc'd Observed Estimated Calc'd Observe Scaled Median Median Mean 3D 3D d 3D Residual 0 5 5.7023566 5.7 5.7 0.566908 0.2907 0.29069 -0.009295 100 8 5.43804842 5.44 5.44 0.566908 0.5374 0.5374 0.0097369 300 5 3.40266882 3.4 3.4 0.566908 0.6261 0.6261 -0.010527 bUU b Z. JUZZ 3bb4 Z.'Jl Z.J1 U.bbbSUH U. (bZb 0. {'6ZtiZ 0. U JU6Z04 900 2 2.10465068 2.09 2.09 0.566908 1.2869 1.28693 -0.036548 2275 2276 Likelihoods of Interest Model Log Likelihood" #of Parameters AIC A1 -21.28323604 6 54.56647 A2 -18.36479748 10 56.72959 A3 -21.28323604 6 54.56647 fitted -21.28451878 5 52.56904 R -46.73498878 2 97.46998 ' Includes additive constant of -22.97346. This constant w a; Tests of Interest Test 2"Log(Likeliho od Ratio) Test df p-ualue 1 56.74038261 8 <0.0001 2 5.83687712 4 0.211666 3 5.83687712 4 0.211666 4 0.002565492 1 0.959604 2277 2278 2279 E.3 BMP Model Results (Hannas et al. 2011) Table Apx E-6. Ex Vivo Feta Rat Testicular Testosterone Data (Hannas et al. 2011) Dose (mg/kg-day) N (# of litters) Mean Standard Deviation Notes 0 3 5.19 1.195115057 Data from Table 1 in (Hannas et al., 2011) 100 3 5.7 0.294448637 300 3 2.27 1.420281662 Page 88 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Dose (mg/kg-day) N (# of litters) Mean Standard Deviation Notes 600 3 1.05 0.692820323 900 3 0.65 0.173205081 2280 Page 89 of 94 ------- PUBLIC RELEASE DRAFT December 2024 2281 Table Apx E-7. BMP Mode Results Ex Vivo Fetal Testicular Testosterone (All Dose Groups) (Hannas et al. 2011) Models" Restrictionb Variance BMR = 5% BMR = 10% BMR = 40% BMR = 1 SD P Value AIC BMDS Recommends BMDS Recommendation Notes BMD BMDL BMD BMDL BMD BMDL BMD BMDL Exponential 3 Restricted Constant 53.19681 17.80873 86.18426 36.58139 248.2968 173.5584 121.3679 58.47676 0.0435151 47.56574178 Questionable Constant variance test failed (Test 2 p-value < 0.05) Goodness of fit p-value <0.1 BMDL 3x lower than lowest non-zero dose Exponential 5 Restricted Constant 253.0245 60.82534 263.8179 89.98152 289.7556 204.3087 269.3968 109.1352 0.2886587 44.42231369 Questionable Constant variance test failed (Test 2 p-value < 0.05) BMD/BMDL ratio > 3 Hill Restricted Constant 168.4655 62.74761 190.5248 161.2484 259.3454 187.7035 202.9592 104.7965 0.2934707 44.40007786 Questionable Constant variance test failed (Test 2 p-value < 0.05) Polynomial Degree 3 Restricted Constant 44.5932 38.08458 89.1864 76.16924 356.7457 304.6767 191.342 137.0539 0.0060429 51.72768824 Questionable Constant variance test failed (Test 2 p-value < 0.05) Goodness of fit p-value <0.1 Polynomial Degree 2 Restricted Constant 44.61656 38.08266 89.23315 76.16526 356.9326 304.661 191.5359 137.0482 0.0060428 51.72769929 Questionable Constant variance test failed (Test 2 p-value < 0.05) Goodness of fit p-value <0.1 Power Restricted Constant 44.60136 38.08385 89.20272 76.1677 356.8109 304.6708 191.4181 137.0529 0.0060429 51.72768347 Questionable Constant variance test failed (Test 2 p-value < 0.05) Goodness of fit p-value <0.1 Linear Unrestricted Constant 44.60135 38.08385 89.20271 76.16769 356.8109 304.6725 191.4181 137.0514 0.0060429 51.72768347 Questionable Constant variance test failed (Test 2 p-value < 0.05) Goodness of fit p-value <0.1 Exponential 3 Restricted Non- Constant 27.99886 15.98305 53.77395 32.81671 224.9883 159.1025 150.5282 68.44409 0.1963944 44.46367306 Questionable Non-constant variance test failed (Test 3 p-value < 0.05) BMD 3x lower than lowest non-zero dose BMDL 3x lower than lowest non-zero dose Exponential 5 Restricted Non- Constant 54.67479 14.05239 87.27415 28.92156 246.6568 142.7024 167.4876 74.84092 0.1504498 45.2760973 Questionable Non-constant variance test failed (Test 3 p-value < 0.05) BMD/BMDL ratio > 3 BMDL 3x lower than lowest non-zero dose Hill Restricted Non- Constant 89.27453 18.68063 119.5257 35.15881 243.712 156.1378 171.9022 88.60382 0.2773613 44.38838686 Questionable Non-constant variance test failed (Test 3 p-value < 0.05) BMD/BMDL ratio > 3 Page 90 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Models" Restrictionb Variance BMR = 5% BMR = 10% BMR = 40% BMR = 1 SD P Value AIC BMDS Recommends BMDS Recommendation Notes BMD BMDL BMD BMDL BMD BMDL BMD BMDL BMDL 3x lower than lowest non-zero dose Polynomial Degree 3 Restricted Non- Constant 52.39019 47.32479 104.7804 94.65019 419.1216 378.5982 500.2151 240.1444 0.0315801 48.04250119 Questionable Non-constant variance test failed (Test 3 p-value < 0.05) Goodness of fit p-value <0.1 Modeled control response std. dev. >1.5 actual response std. dev. Polynomial Degree 2 Restricted Non- Constant 52.38006 47.32657 104.7601 94.65252 419.0404 378.615 499.0547 240.1565 0.0315806 48.04246336 Questionable Non-constant variance test failed (Test 3 p-value < 0.05) Goodness of fit p-value <0.1 Modeled control response std. dev. >1.5 actual response std. dev. Power Restricted Non- Constant 51.70133 47.10326 103.4027 94.20652 413.6106 376.8261 434.4408 236.6546 0.0300659 48.15086454 Questionable Non-constant variance test failed (Test 3 p-value < 0.05) Goodness of fit p-value <0.1 Modeled control response std. dev. >1.5 actual response std. dev. Linear Unrestricted Non- Constant 52.37973 47.32659 104.7594 94.65314 419.0377 378.6051 499.0851 240.1585 0.0315806 48.04246244 Questionable Non-constant variance test failed (Test 3 p-value < 0.05) Goodness of fit p-value <0.1 Modeled control response std. dev. >1.5 actual response std. dev. AIC = Akaike information criterion; BMD = benchmark dose; BMDL =benchmark dose lower limit; NA = Not Applicable. " Selected Model (bolded and shaded gray). b Restrictions defined in the BMDS 3.3 User Guide. 2282 2283 Table Apx E-8. BMP Mode Results Ex Vivo Fetal Testicular Testosterone (Highest Dose Group Removed) (Hannas et al. 2011) Models" Restrictionb Variance BMR = 5% BMR = 10% BMR = 40% BMR = 1 SD P Value AIC BMDS Recommends BMDS Recommendation Notes BMD BMDL BMD BMDL BMD BMDL BMD BMDL Exponential 3 Restricted Constant 256.2599 17.37356 266.7157 35.68542 291.1642 166.2802 276.4071 63.30013 0.0329607 41.77330522 Questionable Goodness of fit p-value <0.1 BMD/BMDL ratio > 3 BMDL 3x lower than lowest non-zero dose Page 91 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Models" Restrictionb Variance BMR = 5% BMR = 10% BMR = 40% BMR = 1 SD P Value AIC BMDS Recommends BMDS Recommendation Notes BMD BMDL BMD BMDL BMD BMDL BMD BMDL Exponential 5 Restricted Constant 252.0464 55.61988 262.9164 83.58491 289.2605 194.5526 270.1764 108.3206 NA 39.79519328 Questionable BMD/BMDL ratio > 3 d.f.=0, saturated model (Goodness of fit test cannot be calculated) Hill Restricted Constant 244.6114 207.076 255.1814 217.9335 284.1949 174.1718 262.508 107.2089 0.450376 37.79519327 Viable - Recommended Lowest AIC EPA Notes: Poor visual fit. No model selected for this data set. Polynomial Degree 3 Restricted Constant 34.81702 28.89754 69.63404 57.79509 278.5362 231.18 134.3093 92.80279 0.0401532 41.65559526 Questionable Goodness of fit p-value <0.1 BMDL 3x lower than lowest non-zero dose Polynomial Degree 2 Restricted Constant 34.81929 28.89742 69.63859 57.79485 278.5543 231.1794 134.3216 92.80369 0.0401532 41.65559589 Questionable Goodness of fit p-value <0.1 BMDL 3x lower than lowest non-zero dose Power Restricted Constant 34.81603 28.89757 69.63207 57.79515 278.5283 231.1806 134.3029 92.80444 0.0401532 41.65559519 Questionable Goodness of fit p-value <0.1 BMDL 3x lower than lowest non-zero dose Linear Unrestricted Constant 34.81604 28.89758 69.63208 57.79515 278.5283 231.1806 134.3029 92.80386 0.0401532 41.65559519 Questionable Goodness of fit p-value <0.1 BMDL 3x lower than lowest non-zero dose AIC = Akaike information criterion; BMD = benchmark dose; BMDL =benchmark dose lower limit; NA = Not Applicable. " Selected Model (bolded and shaded gray). b Restrictions defined in the BMDS 3.3 User Guide. 2284 Page 92 of 94 ------- PUBLIC RELEASE DRAFT December 2024 Frequentist Hill Model with BMR of 0.05 Added Risk for the BMD and 0.95 Lower Confidence Limit for the BMDL Estimated Probability Response at BMD O Data BMD BMDL 600 2285 2286 User Input Info Model frequentist Hill Model Restriction Restricted Dataset Name as (2011.1 - DIBP Testosterone (high dose removed) User notes [Add user notes here] Dose-Response Modi M[dose] = q + v"doseYif(k*n + dose"n) Variance Model Var[i] = alpha Model Options BMR Type Rel. Dev. BMRF 0.05 Tail Probability - Confidence Level 0.95 Distribution Type Normal Variance Type Constant Model Data Dependent Variable mgfkg-d Independent Variable [Custom] Total # of Observation 4 Adverse Direction Automatic 2288 Page 93 of 94 ------- PUBLIC RELEASE DRAFT December 2024 2289 2290 2291 Model Results Benchmark Dose BMD 244.6114226 EiMDL 207.075989 BMDU 256.7691509 AIC 37.79519327 Test 4 P-ualue 1 0.450375979 D.O.F. 1 Model Parameters # of Parameters 5 Variable Estimate Std Error Lower Coni Upper Uonl 9 5.445000004 0.34186044 4.774966 6.115034 V -4.395006471 0.59212145 -5.55554 -3.23447 k 284.475258 10.8295577 263.2497 305.7008 n Bounded NA MA NA alpha 0.701212507 0.20072773 0.307793 1.094632 Goodness of Fit Dose Size Estimated Calc'd Observed Estimated Calc'd Observe Scaled Median Median Mean SD 3D d 3D Residual 0 3 5.445 5.19 5.19 0.837384 1.1951 1.19512 -0.527444 100 3 5.44499997 5.7 5.7 0.837384 0.2944 0.29445 0.5274436 300 3 2.27000003 2.27 2.27 0.837384 1.4203 1.42028 -6.12E-08 bUU J iLwyyyyyr1 lUb lUb U.tiJ (by 4 U. bbLlHZ b.Zbbt-Ub Likelihoods of Interest #of Model Leg Likelihood" Parameters AIC A1 -14.6127439 5 39.22549 A2 -11.4128589 8 38.82572 A3 -14.6127439 5 39.22549 fitted -14.89759663 4 37.79519 R -26.01000707 2 56.02001 " Includes additive constant of -11.02726. This constant was not included in the LL derivation prior to BMDS 3.0. Tests of Interest Test 2'Log(Likeliho od Ratio) Test df p-value 1 29.19429634 6 <0.0001 2 6.399769998 3 0.0937 3 6.399769998 3 0.0937 4 0.569705469 1 0.450376 Page 94 of 94 ------- |