Draft Report on Alternative (Non-Animal) Methods for Cosmetics Testing: current status and future prospects – 2010 doc

45 688 0
Draft Report on Alternative (Non-Animal) Methods for Cosmetics Testing: current status and future prospects – 2010 doc

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

Working Group 5: Reproductive Toxicity 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 DRAFT FOR CONSULTATION 14.07.10 Draft Report on Alternative (Non-Animal) Methods for Cosmetics Testing: current status and future prospects – 2010: Chapter Reproductive Toxicity Compiled by Workgroup 14 July 2010 Sarah Adler1, Thomas Broschard2, Susanne Bremer3, Mark Cronin4, George Daston5, Elise Grignard3, Aldert Piersma6, Guillermo Repetto7 and Michael Schwarz8 Centre for Documentation and Evaluation of Alternatives to Animal Experiments (ZEBET), Federal Institute for Risk Assessment (BfR), Berlin, Germany; Merck KGaA, Darmstadt, Germany; Institute for Health & Consumer Protection, Joint Research Centre, European Commission, Ispra, Italy; School of Pharmacy and Chemistry Liverpool, John Moores University, Liverpool, England; Miami Valley Innovation Center, The Procter and Gamble Company, Cincinnati, USA; Laboratory for Health Protection Research, National Institute for Public Health and the Environment RIVM, Bilthoven, The Netherlands; National Institute of Toxicology and Forensic Sciences, University Pablo de Olavide, Sevilla, Spain; Institute of Pharmacology und Toxicology, University of Tuebingen, Germany Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 34 Executive Summary 35 In the last decades, significant efforts have been undertaken to develop alternative methods to 36 assess reproductive toxicity However, despite the impressive number of alternative tests that 37 have been published and are listed in this report, the majority of these tests have not yet 38 gained regulatory acceptance There are a number of reasons for the relatively slow progress 39 in the implementation of alternative methods for reproductive toxicity safety evaluations, 40 these include: the lengthy research and development phase; a lack of understanding of the 41 mode of actions of reproductive toxicants; and the huge number of physiological mechanisms 42 involved in mammalian reproduction which can be affected by xenobiotics Among the 43 various stages in the reproductive cycle, embryo-foetal development is considered as one of 44 the most critical steps Substantial effort has been spent in the development of promising in 45 vitro assays, such as the Zebrafish embryo test and pluripotent embryonic stem cell models, to 46 allow for the detection of the teratogenic potential of substances However, besides their 47 current role as mechanistic support and screening tools, the role of alternative methods as part 48 of integrated testing strategies for regulatory toxicity evaluations has to be defined further 49 The complexity of mammalian reproduction requires integrated testing strategies to fulfil all 50 needs for hazard identification and risk assessment A promising way forward is the use of 51 recently established comprehensive databases in which toxicological information derived 52 from standardised animal experimentations is collected These databases will allow for the 53 identification of the most sensitive targets of reproductive toxicants This priority setting of 54 sensitive endpoints is the first step to obtain a detailed understanding of the toxicological 55 relevance of the in vitro tests described in this report and how they can be used in integrated 56 testing strategies Furthermore, this mapping exercise will also support the identification of 57 information gaps where further efforts in test development are necessary to design specific 58 alternative methods covering identified sensitive endpoints 59 According to the Cosmetics Directive 76/768/EEC only alternatives leading to full 60 replacement of animal experiments are of relevance for safety evaluations of cosmetic 61 ingredients Regardless, the retrospective analysis of available in vivo data allowing the 62 detection of the most sensitive endpoints, the definition of a tool-box of alternative methods 63 as well as the eventual need to develop additional alternatives to cover the missing building 64 blocks in the testing strategy will need more than 10 years to complete 65 66 67 68 Working Group 5: Reproductive Toxicity 69 70 DRAFT FOR CONSULTATION 14.07.10 Introduction 2.1 Complexity of the Reproductive Cycle 71 Reproductive toxicity refers to a wide variety of toxicological effects that may occur in 72 different phases within the reproductive cycle (figure 1) This includes effects on fertility, 73 sexual behaviour, embryo implantation, embryonic/foetal development, parturition, postnatal 74 adaptation, and subsequent growth and development into sexual maturity An enormous 75 variety of mechanisms at the molecular, cellular and tissue levels cooperate in a concerted and 76 genetically programmed way to regulate these processes The sensitivity to chemical insults 77 may differ extensively between processes In addition, different temporal windows of 78 sensitivity have been observed for different processes As an example, neural tube closure 79 occurs early in pregnancy, and most effects on this process can only be determined after 80 exposure during this critical period of time 81 82 83 Sexual maturation Growth and development Gamete production Postnatal development Fertilisation Transport of the zygote Birth Fetogenesis Implantation Embryogenesis 84 85 86 87 88 Figure 1: The main stages in the mammalian reproductive cycle 89 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 90 91 92 2.2 Alternatives for Reproductive Toxicity Testing 93 Over the last two decades, a wealth of ex vivo and in vitro assays have been proposed as 94 alternative test systems for testing toxic effects on the various processes in reproduction and 95 development Individual in vitro models are reductionistic in nature and are therefore unable to 96 cover all aspects of the reproductive cycle since reproduction requires a complex interplay of 97 integrated functions However, parts of the reproductive cycle can be mimicked by in vitro 98 systems and it is conceivable that a panel of well-designed and validated in vitro tests could 99 replace a substantial proportion of in vivo testing procedures This chapter gives an inventory 100 of the current state of development of alternative test systems for reproductive toxicity hazard 101 assessment 102 Although not applicable for cosmetic ingredients, reduction of animal studies is a more 103 feasible goal than replacement, one example being the current OECD activity towards an 104 extended-1-generation study protocol, which, if it would replace the current 2-generation 105 study, would reduce animal use by roughly 40% in each study [1] The addition of relevant 106 parameters to this novel study protocol represents a good example of refined testing 107 108 Information Requirements for the Safety Assessment of Cosmetic 109 Within the EU the safety of cosmetic products is regulated by the Cosmetics Product 110 Directive 76/768/EEC [2] which will be replaced stepwise by the new EU Cosmetics 111 Regulation 1223/2009 According to Article of Directive 76/768/EEC, a “cosmetic product 112 put on the market must not cause damage to human health when applied under normal or 113 reasonably foreseeable conditions of use” In addition, Article 7a of the same Directive states 114 that the safety evaluation of a finished product should be based on the general toxicological 115 profile, the chemical structure and the level of exposure of each ingredient This implies that 116 a quantitative risk assessment is required for each single ingredient of a cosmetic product 117 Being responsible for the safety of its cosmetic product, the producer performs a risk 118 assessment based on the data of all ingredients used Therefore, a pre-market approval is not 119 necessary for most ingredients used for cosmetics However, certain ingredients listed in 120 positive lists of the Cosmetics Directive such as colorants (Annex IV), preservatives (Annex 121 VI), UV filters (Annex VII) and, most recently, hair dyes require approval of their safety prior 122 to marketing by the EU commission which require the submission of a full dossier[3] 123 Specific requirements for the evaluation of the safety of a cosmetic ingredient are not further Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 124 specified in the Directive either with regard to reproductive toxicity or to any other 125 toxicological endpoint However, information on data requirements with regard to the safety 126 evaluation of cosmetic ingredients are provided in the Notes of Guidance of the former SCCP 127 (now SCCS) [4]: For substances which are submitted for inclusion in the positive lists of the 128 Cosmetics Directive, a comprehensive dossier must be provided for evaluation by the SCCS 129 The dossier includes data on acute toxicity (if available), dermal and mucous membrane 130 irritation, dermal penetration, skin sensitization, repeated dose toxicity, genotoxicity, and 131 phototoxicity (if the cosmetic product is intended to be used on sunlight-exposed skin) 132 Further, it is stated that when considerable oral intake is expected, or when dermal penetration 133 data suggest a significant systemic absorption, information on toxicokinetics, carcinogenicity 134 and reproductive toxicity “may become necessary” Additional recommendations on specific 135 in vivo or in vitro reproductive toxicity studies to be submitted with a dossier are not described 136 in the Notes on Guidance From the SCCS/SCCP opinions published within recent years 137 (2000 – 2009) (http://ec.europa.eu/health/ph_risk/committees/04_sccs/sccs_opinions_en.htm) it can 138 be concluded that in most cases an in vivo developmental toxicity study in the rat (OECD TG 139 414) - submitted by the manufacturer as the only study on reproductive toxicity - was 140 considered sufficient by the SCCS In only a few cases additional data from a 1- or 2- 141 generation study (OECD TG 415 and 416) were included in a dossier [5] 142 For substances, which are not listed in one of the Annexes of the Cosmetic Directive, data on 143 reproductive toxicity are not explicitly asked for in the Notes of Guidance However, some 144 indications of adverse effects on the fertility could be obtained e.g from repeated dose 145 toxicity studies, if available (e.g histopathologic effects on reproductive organs, effects on the 146 endocrine system) 147 148 149 150 Inventory of Animal Test Methods Currently Used for the Evaluation of 151 Developmental and Reproductive Toxicity 152 In the following, OECD test guidelines for the regulatory investigation of the developmental 153 and reproductive toxicity of chemicals are described The list comprises an inventory of the 154 main study protocols However, not all of them are used for testing cosmetic ingredients 155 156 157 4.1 OECD Test Guideline 414: Prenatal Development Toxicity Study for the Testing of Chemicals 158 This Guideline provides general information concerning the effects of prenatal exposure on 159 the pregnant test animal and on the developing organism; this may include assessment of Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 160 maternal effects as well as death, structural abnormalities, or altered growth in the foetus The 161 guideline is not intended to examine solely the period of organogenesis, (e.g days 5-15 in the 162 rodent, and days 6-18 in the rabbit) but also effects from preimplantation, when appropriate, 163 through the entire period of gestation to the day before caesarean section Functional deficits, 164 although an important part of development, are not a part of this Guideline They may be 165 tested for in a separate study or as an adjunct to this study using the Guideline for 166 developmental neurotoxicity 167 The test substance is normally administered to pregnant animals at least from implantation to 168 one day prior to the day of scheduled kill, which should be as close as possible to the normal 169 day of delivery The Guideline is intended for use with rodent (preferably rat) and non-rodent 170 (preferably rabbit) Each test and control group should contain a sufficient number of females 171 to result in approximately 20 female animals with implantation sites at necropsy Three 172 concentrations, at least, should be used The test substance or vehicle is usually administered 173 orally by intubation A limit test may be performed if no effects are expected at a dose of 174 1000 mg/kg bw/d The study includes measurements (weighing) and clinical daily 175 observations One day prior to the expected day of delivery the females are killed, the uterine 176 contents are examined, and the foetuses are evaluated for soft tissue and skeletal changes In 177 any study which demonstrates an absence of toxic effects, further investigation to establish 178 absorption and bioavailability of the test substance should be considered [6] 179 180 4.2 OECD Test Guideline 415: One-Generation Reproduction Toxicity Study 181 This Test Guideline provides general information concerning the effects of a test substance on 182 male and female reproductive performance, such as gonadal function, oestrous cycle, mating 183 behaviour, conception, parturition, lactation and weaning The study may also provide 184 preliminary information about developmental toxic effects of the test substance, such as 185 neonatal morbidity, mortality, behaviour and teratogenesis and to serve as a guide for 186 subsequent tests The test substance is administered orally in graduated doses to several 187 groups of males and females 188 Males should be dosed during growth and for at least one complete spermatogenic cycle; 189 females of the Parent generation should be dosed for at least two complete oestrous cycles 190 The animals are then mated The test substance is administered to both sexes during the 191 mating period and thereafter only to females during pregnancy and for the duration of the 192 nursing period This Test Guideline is intended primarily for use with the rat or mouse Each 193 test and control group should contain a sufficient number of animals to yield about 20 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 194 pregnant females at, or near, term Three test groups, at least, should be used It is 195 recommended that the test substance be administered in the diet or drinking water A limit test 196 may be performed if no effects would be expected at a dose of 1000 mg/kg bw/d The results 197 of this study include measurements (weighing, food consumption) and daily and detailed 198 observations, each day preferably at the same time, as well as gross necropsy and 199 histopathology The findings of a reproduction toxicity study should be evaluated in terms of 200 the observed effects, necropsy and microscopic findings [7] 201 202 4.3 OECD Test Guideline 416: Two-Generation Reproduction Toxicity 203 This Guideline provides general information concerning the effects of a substance on the 204 integrity and performance of the male and female reproductive systems, and on the growth 205 and development of the offspring, including gonadal function, the oestrus cycle, mating 206 behaviour, conception, gestation, parturition, lactation, and weaning, and the growth and 207 development of the offspring The study may also provide information about the effects on 208 neonatal morbidity, mortality, and preliminary data on prenatal and postnatal developmental 209 toxicity as well as serving as a guide for subsequent tests In addition to studying growth and 210 development of the F1 generation, this Guideline is also intended to assess the integrity and 211 performance of the male and female reproductive systems as well as growth and development 212 of the F2 generation For further information on developmental toxicity and functional 213 deficiencies, either additional study segments can be incorporated into this protocol, utilising 214 the Guidelines for developmental toxicity and/or developmental neurotoxicity, or these 215 endpoints could be studied in separate studies 216 The test substance is administered daily in graduated doses to several groups of males and 217 females Males and females of the Parent generation (5-9 weeks old) should be dosed during 218 growth, their mating, the resulting pregnancies and through the weaning of their first 219 generation offspring The administration of the substance is continued to first generation 220 offspring during their growth into adulthood, mating and production of a second generation 221 (until the weaning) The rat is the preferred species for testing Each test and control group 222 should contain a sufficient number of animals to yield preferably not less than 20 pregnant 223 females at or near parturition At least three dose levels and a concurrent control shall be used 224 A limit test may be performed if no effects would be expected at a dose of 1000 mg/kg bw/d 225 The results of this study include: measurements (weighing, sperm parameters, oestrus cycle 226 parameters and offspring parameters), clinical daily observations, as well as gross necropsy 227 and histopathology The findings of this two-generation reproduction toxicity study should be Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 228 evaluated in terms of the observed effects including necropsy and microscopic findings A 229 properly conducted reproductive toxicity test should provide a satisfactory estimation of a no- 230 effect level and an understanding of adverse effects on reproduction, parturition, lactation, 231 postnatal development including growth and sexual development [8] 232 233 4.4 OECD Test Guideline 421: Reproduction/Developmental Toxicity Screening Test 234 This Guideline generates limited information concerning the effects of a substance on male 235 and female reproductive performance such as gonadal function, mating behaviour, 236 conception, development of the conceptus and parturition It is not an alternative to, nor does 237 it replace the existing Test Guidelines 414, 415 and 416 This Screening Test Guideline can 238 be used to provide initial information on possible effects on reproduction and/or development 239 This test does not provide complete information on all aspects of reproduction and 240 development In particular, it offers only limited means of detecting post-natal manifestations 241 of prenatal exposure, or effects that may be induced during post-natal exposure Due (amongst 242 other reasons) to the relatively small numbers of animals in the dose groups, the selectivity of 243 the end points, and the short duration of the study, this method will not provide evidence for 244 definite claims of no effects However, positive results are useful for initial hazard assessment 245 and contribute to decisions with respect to the necessity and timing of additional testing 246 247 The test substance is administered in graduated doses to several groups of male and female 248 rats Males should be dosed for a minimum of four weeks Females should be dosed 249 throughout the study, so approximately 54 days It is recommended that each group be started 250 with at least 10 animals of each sex Generally, at least three test groups and a control group 251 should be used Dose levels may be based on information from acute toxicity tests or on 252 results from repeated dose studies The test substance is administered orally and daily The 253 limit test corresponds to one dose level of at least 1000 mg/kg body weight The results of this 254 study include measurements (weighing, food/water consumption) and daily and detailed 255 observations, preferably each day at the same time, as well as gross necropsy and 256 histopathology The findings of this toxicity study should be evaluated in terms of the 257 observed effects, necropsy and microscopic findings Because of the short period of treatment 258 of the male, the histopathology of the testis and epididymus must be considered, along with 259 the fertility data, when assessing male reproductive effects [9] 260 261 262 4.5 OECD Test Guideline 422: Combined Repeated Dose Toxicity Study with the Reproduction/Developmental Toxicity Screening Test Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 263 The test may be particularly useful as part of the initial screening for the assessment of 264 chemicals for which little or no toxicological information is available and can serve as an 265 alternative to conducting two separate tests for repeated dose toxicity (Guideline 407) and 266 reproduction/developmental toxicity (Guideline 421), respectively It can also be used as a 267 dose range finding study for more extensive reproduction/developmental studies, or when 268 otherwise considered relevant 269 The method comprises the basic repeated dose toxicity study that may be used for chemicals 270 on which a 90-day study is not warranted (e.g when the production volume does not exceed 271 certain limits) or as a preliminary study to a long-term study It further comprises a 272 reproduction/developmental toxicity screening test and, therefore, can also be used to provide 273 initial information on possible effects on male and female reproductive performance such as 274 gonadal function, mating behaviour, conception, development of the conceptus and 275 parturition, either at an early stage of assessing the toxicological properties of chemicals This 276 test does not provide complete information on all aspects of reproduction and development In 277 particular, it offers only limited means of detecting postnatal manifestations of prenatal 278 exposure, or effects that may be induced during postnatal exposure Due (amongst other 279 reasons) to the selectivity of the endpoints and the short duration of the study, this method 280 will not provide evidence for definite claims of no reproduction/developmental effects 281 Although, as a consequence, negative data not indicate absolute safety with respect to 282 reproduction and development, this information may provide some reassurance if actual 283 exposures were clearly less than the dose related to the No Observed Adverse Effect Level 284 (NOAEL) The Guideline also places emphasis on neurological and immunological effects 285 The test substance is administered in graduated doses to several groups of male and female 286 rats Males should be dosed for a minimum of four weeks; females should be dosed 287 throughout the study (approximately 54 days) Normally, matings of "one male to one female" 288 should be used in this study It is recommended that the test substance be administered orally 289 by gavage Each group should be started with at least 10 animals of each sex Generally at 290 least three test groups and a control group should be used Dose levels should be selected 291 taking into account any existing toxicity and (toxico-) kinetic data available The limit test 292 corresponds to one dose level of at least 1000 mg/kg body weight The results of this study 293 include measurements (weighing, food/water consumption) and daily detailed observations 294 (including sensory reactivity to stimuli), preferably each day at the same time, as well as gross 295 necropsy and histopathology The findings of this toxicity study should be evaluated in terms 296 of the observed effects, necropsy and microscopic findings The evaluation will include the Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 297 relationship between the dose of the test substance and the presence or absence of 298 observations Because of the short period of treatment of the male, the histopathology of the 299 testis and epididymus must be considered along with the fertility data, when assessing male 300 reproduction effects [10] 301 302 4.6 OECD Test Guideline 426: Developmental Neurotoxicity Study 303 The developmental neurotoxicity study provides information on the potential functional and 304 morphological effects on the developing nervous system of the offspring of repeated exposure 305 to a substance during in utero and early postnatal development 306 A developmental neurotoxicity study can be conducted as a separate study, incorporated into 307 a reproductive toxicity and/or adult neurotoxicity study (e.g., Test Guidelines 415, 416, 424), 308 or added onto a prenatal developmental toxicity study (e.g., Test Guideline 414) When the 309 developmental neurotoxicity study is incorporated within or attached to another study, it is 310 imperative to preserve the integrity of both study types 311 The test substance is administered daily, generally orally, to mated females (rats are preferred) 312 from the time of implantation (GD 6) throughout lactation (PND 21) At least three dose 313 levels and a concurrent control should be used and a total of 20 litters are recommended at 314 each dose level Dams are tested to assess effects in pregnant and lactating females and may 315 also provide comparative information Offspring are randomly selected from within litters for 316 neurotoxicity evaluation All dams and all offspring should be carefully observed at least once 317 daily with respect to their health, including morbidity and mortality The evaluation consists 318 of observations to detect gross neurological and behavioural abnormalities, and the evaluation 319 of brain weights and neuropathology during postnatal development and adulthood The report 320 should include the body weight, the food/water consumption; the detailed clinical 321 observations, the necropsy findings, a detailed description of all behavioural, the number of 322 animals at the start and at the end of the study and the toxic response data by sex and dose 323 level [11] 324 325 326 4.7 OECD Test Guideline 440: Uterotrophic Bioassay in Rodents: A short-term screening test for oestrogenic properties 327 The Uterotrophic Bioassay is an in vivo short-term screening test It evaluates the ability of a 328 chemical to elicit biological endocrine disruption activities consistent with agonists or 329 antagonists of natural oestrogens (e.g 17ß-estradiol) It is based on the increase in uterine 330 weight or uterotrophic response The uterus responds to oestrogens in two ways An initial 331 response is an increase in weight due to water imbibition This response is followed by a 10 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 Covering different aspects of the steroid production incl gonadotrophinreceptors mediated toxicity Hazard identification and mechanistic studies Inhibition of androgens aromatisation Mechanistic studies Binding assays and reporter gene assays Mechanistic studies Optimisation > years Induction/inhibition of reporter gene product Mechanistic studies Optimisation 3-5 years OECD TG 416, Interaction with OECD TG 415 estrogen OECD TG 414 receptors OECD TG 440 Estrogen receptor binding, Induction of reporter gene product Mechanistic studies Validation and regulatory acceptance ongoing OECD TG 416, Interaction with OECD TG 415 Progesterone OECD TG 414 receptor Receptor binding, reporter gene assays Mechanistic studies Optimization 3-5 years OECD TG 416, Progesterone OECD TG 415 production OECD TG 414 Cell viability, progesterone production, histology Mechanistic studies R&D > years OECD TG 416, OECD TG 415 Other Steroidogenesis Assays OECD TG 416, OECD TG 415 Placental aromatase assay OECD TG 416, Thyroid OECD TG 415 receptor interactions OECD TG 416, Interaction with OECD TG 415 the aryl OECD TG 414 hydrocarbon receptor R&D > years Validation ongoing Different tests are based on various cell lines driving from rat, mouse and human tissues, as well as genetically engineered cells and primary leydig cells Different cellular assays as well as microsomal tests are in development 31 Working Group 5: Reproductive Toxicity OECD TG 416, Interaction with OECD TG 415 androgen OECD TG 414 receptor OECD TG 441 DRAFT FOR CONSULTATION 14.07.10 Binding and induction/inhibition of reporter gene product Mechanistic studies Optimization 3-5 years OECD TG 416, OECD TG 415 Gonadotrophin mediated effects release of gonadotrophins or receptor binding Human ex vivo model to assess transplacental Placenta transfer indicting perfusion system foetal exposure and metabolism of test chemicals OECD TG 416, Toxicity assessment Trophoblast cell OECD TG 415 during embryo assay OECD TG 414 implantation OECD TG 416, OECD TG 415 OECD TG 414 OECD TG 416, OECD TG 415 OECD TG 416, OECD TG 415 Mechanistic studies R&D Hazard identification that Optimisation might lead to fertility impairments R&D > years Several celluar tests based on primary cells/explants or cell lines covering FSH, GnRH, LH, induced effects are described 3-5 years > years Fertilisation, first Mouse embryo cleavage embryo bioassay development Hazard identification that Optimisation might lead to fertility impairments 3-5 years completion of oocyte meiosis up to the Follicle bioassay metaphase II (FBA) Oogenesis: oocyte yield; diameter; nuclear maturatio Hazard identification that Optimisation might lead to fertility impairments 3-5 years 32 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 Bovine maturation test completion of oocyte meiosis up to the metaphase II Hazard identification that Optimisation might lead to fertility impairments Bovine fertilisation test OECD TG 416, OECD TG 415 Formation of female and male pronuclei after penetration of capacitated bull spermatozoa into matured oocytes Hazard identification that Optimisation might lead to fertility impairments 3-5 years DNA strand breaks and alkali labile sites in bull sperm Hazard identification that Optimisation might lead to fertility impairments 3-5 years Hazard identification that Optimisation might lead to fertility impairments 3-5 years Hazard identification that R&D might lead to fertility impairments > years OECD TG 416, OECD TG 415 OECD TG 416, OECD TG 415 ReProComet assay OECD TG 416, OECD TG 415 CASA test OECD TG 416, OECD TG 415 Sertoli cell tests multiple leading to impairments in motility, and viability as well as morphology of spermatozoa Secretion of Inhibin B by (1) primary rat Sertoli cells and (2) cells of line SerW3; cytotoxicity 3-5 years 33 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 OECD TG 414 Stem cell based tests for assessing embryotoxicity OECD TG 414 OECD TG 414 OECD TG 414 OECD TG 414 Interaction with cell differentiation into neural, cartilage, bone and cardiac cells, chemical effects on signalling pathways Whole rat embryo culture Chicken embryo culture The Zebrafish Embryo Teratogenicity Assay Frog Embryo Teratogenesis Assay Xenopus (FETAX) OECD TG 416, Hamster Egg OECD TG 415 Penetration Test/ Hypoosmotic Swelling Test Assessment of lethality, malformations and growth retardation Assessment of lethality, malformations and growth retardation Functional status of the sperm: -sperm quality -fertilizing capacity Hazard identification of embryotoxicants Hazard identification of embryotoxicants Hazard identification of embryotoxicants Screening test for the hazard identification of developmental toxicants Screening test for the hazard identification of developmental toxicants Hazard identification leading to female and/or male toxicity Optimisation At least 3-5 years Stem cell based systems are difficult to optimized due to the challenges in stem cell differentiation protocols Validated Optimisation At least 3-5 years Optimisation At least 3-5 years Optimisation At least 3-5 years Optimisation A common standard protocol is needed to start prevalidation 3-5 years 951 34 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 952 953 Identified Areas with no Alternative Methods Available and Related Scientific/ 954 Technical Difficulties 955 6.1 Approaches for alternative Testing 956 A significant number of alternative assays have been developed as described in chapter 957 However, their implementation in regulatory toxicity testing has not yet been achieved As 958 stated in the introduction to this chapter, the reproductive cycle combines a highly diverse 959 multitude of biological processes and mechanisms, each of which has their own time-related 960 sensitivity to xenobiotic exposures It is therefore a significant challenge to mimic all aspects 961 of the reproductive cycle with in vitro and in silico assays, which may be considered necessary 962 in order that reproductive toxicity can be predicted reliably on the basis of alternative assays 963 alone The classical aim of “one to one” replacement of in vivo protocols by alternative tests is 964 clearly not feasible for the complex reproductive and developmental toxicity animal study 965 protocols Alternative approaches are required in which a limited array of most sensitive 966 endpoints are reproduced by a set of alternative assays which, in combination, could provide 967 sufficient background for hazard identification and risk assessment other endpoints might be 968 identified for which alternative methods not yet exist These endpoints may include 969 amongst others ADME, spermatogenesis, sperm maturation, HPG axis and maternally 970 mediated effects 971 972 6.2 General limitations of in vitro methods for reproductive toxicity testing 973 It is generally recognised that in vitro methods represent only a very simplified picture of 974 reality, i.e of living organisms In the case of reproductive toxicity each in vitro model 975 encompasses only a small part of the complex reproductive cycle and not all steps of 976 reproduction are covered by those assays so far Moreover, many in vitro assays, e.g the 977 receptor binding assays, investigate rather a cellular mechanism which finally might or might 978 not result in an adverse effect in vivo Most in vitro assays not consider the various aspects 979 of absorption, distribution, metabolism and excretion (ADME), which have a tremendous 980 impact on the toxicological profile of a substance In addition, the distinction between general 981 toxicity and specific developmental effects is difficult to evaluate even with more complex in 982 vitro studies such as the whole embryo assays In general, the influence of the maternal 983 organism including maternal toxicity is not covered by in vitro studies Finally, technical 984 difficulties may occur such as low water solubility of test materials, which have to be 985 considered in the design of each individual in vitro assay 986 35 Working Group 5: Reproductive Toxicity 987 6.3 DRAFT FOR CONSULTATION 14.07.10 The Testing Strategy as the Future Driving Force 988 Classically, many in vitro alternatives have been developed based on relatively simple 989 endpoints that were deemed representative of the wider context of e.g embryotoxicity 990 Intrinsically reductionistic assays such as rodent whole embryo culture (WEC), the embryonic 991 stem cell test (EST), and the limb bud micromass (MM) were validated for their prediction of 992 the entire embryotoxicity endpoint This approach presumes that the endpoints represented in 993 these tests are actually among the critical ones for embryotoxicity prediction in general 994 Recent experience with the EST has shown that this is an oversimplification, leading to false 995 positives and false negatives and thus limited predictability, showing that the applicability 996 domain of the assay was more limited than anticipated This has led to the insight that rather 997 than focussing exclusively on individual assays and their relevance, it might be more 998 productive, in view of the regulatory implementation of alternative assays, to start from the 999 different direction, which is their anticipated role in testing strategies for chemical risk 1000 assessment 1001 1002 6.4 Retrospective Analyses to Select Critical Endpoints 1003 A wealth of in vivo data has been collected over the past 30 years since the introduction of 1004 OECD test guidelines for reproductive and developmental toxicity testing Databases 1005 collecting past experience are being built and allow detailed analysis to assess the relative 1006 sensitivity of endpoints in existing animal protocols [103] The combination of the most 1007 sensitive endpoints should be able to detect nearly all reproductive and developmental 1008 toxicants Current thinkings evolving towards designing alternative assays limited to covering 1009 each of these most sensitive endpoints only In addition, these assays should be fitted 1010 appropriately within the entire testing strategy either tiered, or as a battery, or in some 1011 combination of the two, in a phase where the information can be used optimally for hazard 1012 and risk assessment in order to preclude as much as possible any ultimate animal 1013 experimentation [104] 1014 1015 6.5 Towards the Definition of Novel Testing Paradigms 1016 The OECD conceptual framework for reproductive toxicity testing provides an outline of such 1017 an approach Starting from in silico (non-testing) information such as physico-chemical 1018 characteristics, structure-activity relationships and read-across methods, and followed by in 1019 vitro assays, it should be possible to restrict in vivo testing to an essential minimum Crucial 1020 steps in the process of designing the testing strategy on the basis of alternative approaches are 1021 the identification of most sensitive end points, evaluation of existing alternative tests 36 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 1022 containing these end points, design of novel assays for sensitive endpoints not already 1023 addressed by current assays and optimisation of the testing strategy through the 1024 combination of assays It should be realized that the entire reproductive cycle in a living 1025 animal is, by definition, more than the sum of any combination of alternative approaches 1026 Therefore, for reproductive toxicity testing, animal studies will remain the last resort for 1027 foreseeable time This is caused by e.g the often long lag time between exposure and 1028 observed adverse effect in the reproductive cycle and also by kinetic aspects, which hamper 1029 ready translation from in vitro effective concentrations to in vivo effective doses In addition, 1030 efforts to build risk assessment solely on human derived data [105] are currently limited by 1031 the scarcity of relevant human toxicological data 1032 1033 6.6 Time Schedule for Phasing out in vivo Reproductive Toxicity Testing 1034 Given current knowledge and a realistic outlook into the future, full replacement of animal 1035 studies for reproductive toxicity hazard assessment is not probable within foreseeable time 1036 (>10 years) However, alternative assays are already being used for priority setting and 1037 screening purposes In addition, alternative assays can make an important contribution to the 1038 mechanistic understanding of reproductive toxicity Such tests may actually be able to give 1039 more specific information on the interference of the test compound with the endpoint 1040 involved than the in vivo study is able to generate The challenge is to build on these 1041 advantages of alternative tests in generating a testing strategy in which the most sensitive end 1042 points are combined in a well-informed selection of alternative assays Applying such a 1043 strategy in a tiered screening situation (cf the OECD conceptual framework) could preclude 1044 the in vivo testing of many reproductive toxicants and thus would considerably refine and 1045 reduce testing for reproductive toxicity 1046 1047 1048 1049 1050 1051 1052 1053 References [1] Cooper RL, Lamb JC, Barlow SM, Bentley K, Brady AM, Doerrer NG, Eisenbrandt DL, Fenner-Crisp PA, Hines RN, Irvine LF, Kimmel CA, Koeter H, Li AA, Makris SL, Sheets LP, Speijers G, Whitby KE: A tiered approach to life stages testing for agricultural chemical safety assessment Crit Rev Toxicol 2006;36:69-98 [2] Anon Council Directive 76/768/EEC of July 1976 on the approximation of the laws of the Member States relating to cosmetic products 169-200 1976 Official J L262 1054 1055 1056 1057 1058 [3] Pauwels M, Rogiers V: Human health safety evaluation of cosmetics in the EU: a legally imposed challenge to science Toxicol Appl Pharmacol 1-3-2010;243:260-274 1059 1060 [4] Scientific Committee on Consumer Products (SCCP): The SCCP's note of guidance for the testing of cosmetic ingredients and their safety evaluation 2006;6th revision:106 37 Working Group 5: Reproductive Toxicity 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 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 DRAFT FOR CONSULTATION 14.07.10 [5] Pauwels M, Rogiers V In preparation [6] Anon OECD Guideline for the testing of chemicals 414 Prenatal Development Toxicity Study 2001 http://www.oecdilibrary.org/oecd/deliver/fulltext/9741401e.pdf?contentType=/ns/Book&itemI d=/content/book/9789264070820en&containerItemId=/content/serial/20745788&accessItemIds=&mimeType=application/pdf, accessed 11 July 2010 [7] Anon OECD Guideline for the testing of chemicals 415 1983 http://www.oecdilibrary.org/oecd/deliver/fulltext/9741501e.pdf?contentType=/ns/Book&itemI d=/content/book/9789264070844en&containerItemId=/content/serial/20745788&accessItemIds=&mimeType=application/pdf, accessed 11 July 2010 [8] Anon OECD Guideline for the testing of chemicals 416 Two-Generation Reproduction Toxicity 2001 http://www.oecdilibrary.org/oecd/deliver/fulltext/9741601e.pdf?contentType=/ns/Book&itemI d=/content/book/9789264070868en&containerItemId=/content/serial/20745788&accessItemIds=&mimeType=application/pdf, acccessed 11 July 2010 [9] Anon OECD Guideline for the testing of chemicals 421 Reproduction/Developmental Toxicity Screening Test 1995 http://www.oecdilibrary.org/oecd/deliver/fulltext/9742101e.pdf?contentType=/ns/Book&itemI d=/content/book/9789264070967en&containerItemId=/content/serial/20745788&accessItemIds=&mimeType=application/pdf, accessed 11 July 2010 [10] Anon OECD Guideline for the testing of chemicals 422 Combined Repeated Dose Toxicity Study with the Reproduction/Developmental Toxicity Screening Test 1996 http://www.oecdilibrary.org/oecd/deliver/fulltext/9742201e.pdf?contentType=/ns/Book&itemI d=/content/book/9789264070981en&containerItemId=/content/serial/20745788&accessItemIds=&mimeType=application/pdf, accessed 11 July 2010 [11] Anon OECD Guideline for the testing of chemicals 426 Developmental Neurotoxicity Study 2007 http://www.oecdilibrary.org/oecd/deliver/fulltext/9742601e.pdf?contentType=/ns/Book&itemI d=/content/book/9789264067394en&containerItemId=/content/serial/20745788&accessItemIds=&mimeType=application/pdf, accessed 11 July 2010 [12] Anon OECD Guideline for the testing of chemicals 440 Uterotrophic Bioassay in Rodents A short-term screening test for oestrogenic properties 2007 http://www.oecdilibrary.org/oecd/deliver/fulltext/9744001e.pdf?contentType=/ns/Book&itemI d=/content/book/9789264067417en&containerItemId=/content/serial/20745788&accessItemIds=&mimeType=application/pdf, accessed 11 July 2010 [13] Anon OECD Guideline for the testing of chemicals 441 Hershberger Bioassay in Rats A Short-term Screening Assay for (Anti)Androgenic Properties 2009 http://www.oecdilibrary.org/oecd/deliver/fulltext/9744101e.pdf?contentType=/ns/Book&itemI d=/content/book/9789264076334en&containerItemId=/content/serial/20745788&accessItemIds=&mimeType=application/pdf, accessed 11 July 2010 38 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 1117 1118 1119 1120 1121 1122 1123 1124 1125 [14] Anon OECD Guideline for the testing of chemicals 455 The Stably Transfected Human Estrogen Receptor-alpha Transcriptional Activation Assay for Detection of Estrogenic Agonist-Activity of Chemicals 2009 http://www.oecdilibrary.org/oecd/deliver/fulltext/9745501e.pdf?contentType=/ns/Book&itemI d=/content/book/9789264076372en&containerItemId=/content/serial/20745788&accessItemIds=&mimeType=application/pdf, accessed 11 July 2010 1126 1127 [15] Brown NA, Fabro S: Quantitation of rat embryonic development in vitro: a morphological scoring system Teratology 1981;24:65-78 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 [16] Anon Embryotoxicity Testing in Post-Implantation Embryo Culture- Method of Piersma INVITTOX no 123 http://ecvamdbalm.jrc.ec.europa.eu/public_view_doc2.cfm?id=15BFE9AC6ECC8ED3D7F6DF03B5D5B BE97180BB0BC12CB10496CDA74B54630A05A3291B895581F634, accessed 13 July 2010 1138 1139 [18] Nagel R: DarT: The embryo test with the Zebrafish Danio rerio a general model in ecotoxicology and toxicology ALTEX 2002;19 Suppl 1:38-48 1140 1141 [19] Zon LI, Peterson RT: In vivo drug discovery in the zebrafish Nat Rev Drug Discov 2005;4:35-44 1142 1143 1144 [20] Brannen KC, Panzica-Kelly JM, Danberry TL, Augustine-Rauch KA: Development of a zebrafish embryo teratogenicity assay and quantitative prediction model Birth Defects Res B Dev Reprod Toxicol 2010;89:66-77 1145 1146 1147 [21] Selderslaghs IW, Van Rompay AR, De CW, Witters HE: Development of a screening assay to identify teratogenic and embryotoxic chemicals using the zebrafish embryo Reprod Toxicol 2009;28:308-320 1148 1149 1150 [22] Ito T, Ando H, Suzuki T, Ogura T, Hotta K, Imamura Y, Yamaguchi Y, Handa H: Identification of a primary target of thalidomide teratogenicity Science 12-3-2010;327:13451350 1151 1152 1153 [23] van Boxtel AL, Kamstra JH, Fluitsma DM, Legler J: Dithiocarbamates are teratogenic to developing zebrafish through inhibition of lysyl oxidase activity Toxicol Appl Pharmacol 154-2010;244:156-161 1154 1155 1156 1157 [24] Busquet F, Nagel R, von Landenberg F, Mueller SO, Huebler N, Broschard TH: Development of a new screening assay to identify proteratogenic substances using zebrafish Danio rerio embryo combined with an exogenous mammalian metabolic activation system (mDarT) Toxicological Sciences 2008;104:177-188 1158 1159 1160 [25] Bantle JA, Finch RA, Fort DJ, Stover EL, Hull M, Kumsher-King M, Gaudet-Hull AM: Phase III interlaboratory study of FETAX Part FETAX validation using 12 compounds with and without an exogenous metabolic activation system J Appl Toxicol 1999;19:447-472 [17] Genschow E, Spielmann H, Scholz G, Seiler A, Brown N, Piersma A, Brady M, Clemann N, Huuskonen H, Paillard F, Bremer S, Becker K: The ECVAM international validation study on in vitro embryotoxicity tests: results of the definitive phase and evaluation of prediction models European Centre for the Validation of Alternative Methods Altern Lab Anim 2002;30:151-176 39 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 1161 1162 1163 1164 1165 1166 [26] Anon American Society for Testing an Materials (ASTM) Standard guide for conducting the frog embryo teratogenesis assay - Xenopus (FETAX), E1439-91 11.05, 826-836 1998 Annual Book of ASTM Standards, ASTM:Philadelphia, PA 1167 1168 [28] Jelinek R: The chick embryotoxicity screening test (CHEST).; in Neubert D, Merker HJ, Kwasigroch TE (eds): Methods in pre-natal toxicology Stuttgart, Thieme, 1977, pp 381-386 1169 1170 1171 1172 1173 1174 1175 [29] Boehn SNE, Kaufmann T, Koch M, Wessa P, Kamp HG, Landsiedel R, van Ravenzwaay B Modification and evaluation of the chicken embryotoxicity screening test (CHEST) as in vitro test system for embryotoxicity 2009 VII World Congress on Alternatives & Animal Use in the Life Sciences, Rome 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 [31] Bernshausen T, Noss A, Boehn SN, Fabian E, Kamp HG, Kaufmann T, Koch M, Wessa P, van Ravenzwaay B, Landsiedel R Metabolic Competences of the Chicken Embryo used in the Toxicity Screening Test (CHEST) as an In Vitro Test System for Teratogenicity and Embryotoxicity 2009 50 Jahrestagung der DGPT, Mainz, Germany 1187 1188 [34] Kucera P, Burnand MB: Routine teratogenicity test that uses chick embryos in vitro Teratog Carcinog Mutagen 1987;7:427-447 1189 1190 [35] Jelinek R, Peterka M, Rychter Z: Chick embryotoxicity screening test 130 substances tested Indian J Exp Biol 1985;23:588-595 1191 1192 [36] Jelinek R, Marhan O: Validation of the Chick Embryotoxicity Screening Test (CHEST) A comparative study Funct Dev Morphol 1994;4:317-323 1193 1194 [37] Gilani SH, Alibhai Y: Teratogenicity of metals to chick embryos J Toxicol Environ Health 1990;30:23-31 1195 1196 1197 [38] Durmus E, Inan O, Celik I, Sur E, Ozkan Y, Acar A, Aydin MF: Use of the fertilized hen's egg in the evaluation of embryotoxicity of dental alloys J Biomed Mater Res B Appl Biomater 15-2-2005;72:322-327 1198 1199 [39] Flint OP: A micromass culture method for rat embryonic neural cells J Cell Sci 1983;61:247262 1200 1201 [40] Flint OP, Orton TC: An in vitro assay for teratogens with cultures of rat embryo midbrain and limb bud cells Toxicol Appl Pharmacol 1984;76:383-395 1202 1203 [41] Brown NA, Spielmann H, Bechter R, Flint OP, Freeman SJ, Jeläinek RJ, Koch E, Nau H, Newall DR, Palmer AK, et al Screening chemicals for reproductive toxicity: the current [27] Fort JD, Paul RRP: Enhancing the Predictive Validity of Frog Embryo Teratogenesis AssayXenopus(FETAX) Journal of Applied Toxycology 2002;22:185-191 [30] Kotwani A: Use of chick embryo in screening for teratogenicity Indian J Physiol Pharmacol 1998;42:189-204 [32] Anon Report of the WHO Scientific Group WHO principles for the testing of drugs for teratogenicity 18, 364 1967 WHO Technical Reports Series [33] Kemper FH, Luepke NP: Toxicity Testing by the Hens Egg Test (Het) Food and Chemical Toxicology 1986;24:647-648 40 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 alternatives; the report and recommendations of an ECVAM/ETS workshop (ECVAM workshop 12) 23[6], 868-82 1995 Altern Lab Anim 1204 1205 1206 1207 1208 [42] Balls M, Hellsten E: Statement of the scientific validity of the embryonic stem cell test (EST) an in Vitro test for embryotoxicity Altern Lab Anim 2002;30:265-268 1209 1210 1211 1212 1213 1214 [43] Marx-Stoelting P, Adriaens E, Ahr HJ, Bremer S, Garthoff B, Gelbke HP, Piersma A, Pellizzer C, Reuter U, Rogiers V, Schenk B, Schwengberg S, Seiler A, Spielmann H, Steemans M, Stedman DB, Vanparys P, Vericat JA, Verwei M, van der Water F, Weimer M, Schwarz M: A review of the implementation of the embryonic stem cell test (EST) The report and recommendations of an ECVAM/ReProTect Workshop Altern Lab Anim 2009;37:313328 1215 1216 1217 [44] Zur Nieden NI, Kempka G, Ahr HJ: Molecular multiple endpoint embryonic stem cell test a possible approach to test for the teratogenic potential of compounds Toxicol Appl Pharmacol 1-2-2004;194:257-269 1218 1219 1220 [45] Buesen R, Genschow E, Slawik B, Visan A, Spielmann H, Luch A, Seiler A: Embryonic stem cell test remastered: comparison between the validated EST and the new molecular FACSEST for assessing developmental toxicity in vitro Toxicol Sci 2009;108:389-400 1221 1222 [46] Zur Nieden NI, Ruf LJ, Kempka G, Hildebrand H, Ahr HJ: Molecular markers in embryonic stem cells Toxicol In Vitro 2001;15:455-461 1223 1224 1225 [47] Peters AK, Wouwer GV, Weyn B, Verheyen GR, Vanparys P, Gompel JV: Automated analysis of contractility in the embryonic stem cell test, a novel approach to assess embryotoxicity Toxicol In Vitro 2008;22:1948-1956 1226 1227 1228 1229 [48] Bremer S, Van Dooren M, Paparella M, Kossolov E, Fleischmann B, Hescheler J: Establishment of an embryotoxicity assay with green fluorescence protein-expressing embryonic cell-derived cardiomyocytes Atla-Alternatives to Laboratory Animals 1999;27:471-484 1230 1231 1232 [49] Uibel F, Muhleisen A, Kohle C, Weimer M, Stummann TC, Bremer S, Schwarz M: ReProGlo: A new stem cell-based reporter assay aimed to predict embryotoxic potential of drugs and chemicals Reprod Toxicol 14-12-2009 1233 1234 1235 [50] Adler S, Lindqvist J, Uddenberg K, Hyllner J, Strehl R: Testing potential developmental toxicants with a cytotoxicity assay based on human embryonic stem cells Altern Lab Anim 2008;36:129-140 1236 1237 1238 [51] Adler S, Pellizzer C, Hareng L, Hartung T, Bremer S: First steps in establishing a developmental toxicity test method based on human embryonic stem cells Toxicol In Vitro 2008;22:200-211 1239 1240 1241 [52] Stummann TC, Bremer S: The possible impact of human embryonic stem cells on safety pharmacological and toxicological assessments in drug discovery and drug development Curr Stem Cell Res Ther 2008;3:118-131 1242 1243 1244 [53] West PR, Weir AM, Smith AM, Donley EL, Cezar GG: Predicting human developmental toxicity of pharmaceuticals using human embryonic stem cells and metabolomics Toxicol Appl Pharmacol 19-5-2010 1245 1246 1247 [54] Mose T, Kjaerstad MB, Mathiesen L, Nielsen JB, Edelfors S, Knudsen LE: Placental passage of benzoic acid, caffeine, and glyphosate in an ex vivo human perfusion system J Toxicol Environ Health A 2008;71:984-991 41 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 1248 1249 1250 [55] Myllynen P, Immonen E, Kummu M, Vahakangas K: Developmental expression of drug metabolizing enzymes and transporter proteins in human placenta and fetal tissues Expert Opin Drug Metab Toxicol 2009;5:1483-1499 1251 1252 1253 [56] Morck TJ, Sorda G, Bechi N, Rasmussen BS, Nielsen JB, Ietta F, Rytting E, Mathiesen L, Paulesu L, Knudsen LE: Placental transport and in vitro effects of Bisphenol A Reprod Toxicol 7-3-2010 1254 1255 1256 1257 1258 [57] Anon The official ReProTect Website http://www.reprotect.eu/, accessed 12 July 2010 1259 1260 [59] Lemeire K, Van M, V, Cortvrindt R: The antibiotic streptomycin assessed in a battery of in vitro tests for reproductive toxicology Toxicol In Vitro 2007;21:1348-1353 1261 1262 1263 [60] Lazzari G, Tessaro I, Crotti G, Galli C, Hoffmann S, Bremer S, Pellizzer C: Development of an in vitro test battery for assessing chemical effects on bovine germ cells under the ReProTect umbrella Toxicol Appl Pharmacol 15-12-2008;233:360-370 1264 1265 1266 1267 [61] Luciano AM, Franciosi F, Lodde V, Corbani D, Lazzari G, Crotti G, Galli C, Pellizzer C, Bremer S, Weimer M, Modina SC: Transferability and inter-laboratory variability assessment of the in vitro bovine oocyte maturation (IVM) test within ReProTect Reprod Toxicol 13-22010 1268 1269 [62] Van M, V, Van WK, Cortvrindt R: In vitro effects of dexamethasone on mouse ovarian function and pre-implantation embryo development Reprod Toxicol 2007;23:32-41 1270 1271 1272 [63] Schaefer WR, Fischer L, Deppert WR, Hanjalic-Beck A, Seebacher L, Weimer M, Zahradnik HP: In vitro-Ishikawa cell test for assessing tissue-specific chemical effects on human endometrium Reprod Toxicol 19-2-2010 1273 1274 [64] Lippman M, Bolan G, Huff K: The effects of estrogens and antiestrogens on hormoneresponsive human breast cancer in long-term tissue culture Cancer Res 1976;36:4595-4601 1275 1276 1277 1278 1279 1280 1281 1282 [65] Anon US-EPA: Integrated summary report for the Validation of an Androgen Receptor Binding Assay as a potential screen in the endocrine disruptor screening program [http://www.epa.gov/endo/pubs/ar_binding_isr.pdf] 2007 assessed on July 2010 1283 1284 1285 1286 1287 1288 1289 1290 1291 [67] Anon US-EPA: Integrated summary report for Validation of an Estrogen Receptor Binding Assay using Rat Uterine Cytosol as Source of Receptor as a Potential Screen in the Endocrine Disruptor Screening Program Tier Battery 2009 http://www.epa.gov/endo/pubs/assayvalidation/er_ruc_isr.pdf, assessed on July 2010 [58] Cordelli E, Fresegna AM, D'Alessio A, Eleuteri P, Spano M, Pacchierotti F, Villani P: ReProComet: a new in vitro method to assess DNA damage in mammalian sperm Toxicol Sci 2007;99:545-552 [66] Hartig PC, Cardon MC, Blystone CR, Gray LE, Jr., Wilson VS: High throughput adjustable 96-well plate assay for androgen receptor binding: a practical approach for EDC screening using the chimpanzee AR Toxicol Lett 26-9-2008;181:126-131 [68] Freyberger A, Wilson V, Weimer M, Tan S, Tran HS, Ahr HJ: Assessment of a robust model protocol with accelerated throughput for a human recombinant full length estrogen receptoralpha binding assay: Protocol optimization and intralaboratory assay performance as initial steps towards validation Reprod Toxicol 13-1-2010 42 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 [69] Battelle Revised AR binding Assay Protocol: Androgen Receptor Competitive Binding Protocol Using Rat Ven,tral Prostate Cytosol, SOP No NHEERL-H/RTD/EB/VW/2002-03000 In Quality Assurance Project Plan for Work Assignement 2-22, Development of Androgen Receptor Binding Data, prepared by Battelle for U.S Environmental Protection Agency, Endocrine Disruptor Screening Program, Contract No 68-W-01-023 2002 1292 1293 1294 1295 1296 1297 1298 1299 [70] Freyberger A, Weimer M, Tran HS, Ahr HJ: Assessment of a recombinant androgen receptor binding assay: Initial steps towards validation Reprod Toxicol 13-10-2009 1300 1301 [71] Attardi BJ, Hild SA, Reel JR: Dimethandrolone undecanoate: a new potent orally active androgen with progestational activity Endocrinology 2006;147:3016-3026 1302 1303 1304 [72] Viswanath G, Halder S, Divya G, Majumder CB, Roy P: Detection of potential (anti)progestagenic endocrine disruptors using a recombinant human progesterone receptor binding and transactivation assay Mol Cell Endocrinol 25-11-2008;295:1-9 1305 1306 1307 [73] Klotz DM, Ladlie BL, Vonier PM, McLachlan JA, Arnold SF: o,p'-DDT and its metabolites inhibit progesterone-dependent responses in yeast and human cells Mol Cell Endocrinol 25-41997;129:63-71 1308 1309 1310 [74] Ishihara A, Rahman FB, Leelawatwattana L, Prapunpoj P, Yamauchi K: In vitro thyroid hormone-disrupting activity in effluents and surface waters in Thailand Environ Toxicol Chem 2009;28:586-594 1311 1312 1313 1314 [75] Witters H, Freyberger A, Smits K, Vangenechten C, Lofink W, Weimer M, Bremer S, Ahr PH, Berckmans P: The assessment of estrogenic or anti-estrogenic activity of chemicals by the human stably transfected estrogen sensitive MELN cell line: Results of test performance and transferability Reprod Toxicol 31-3-2010 1315 1316 1317 1318 [76] van der Burg B, Winter R, Weimer M, Berckmans P, Suzuki G, Gijsbers L, Jonas A, van der Linden S, Witters H, Aarts J, Legler J, Kopp-Schneider A, Bremer S: Optimization and prevalidation of the in vitro ERalpha CALUX method to test estrogenic and antiestrogenic activity of compounds Reprod Toxicol 8-5-2010 1319 1320 1321 [77] Long M, Laier P, Vinggaard AM, Andersen HR, Lynggaard J, Bonefeld-Jorgensen EC: Effects of currently used pesticides in the AhR-CALUX assay: comparison between the human TV101L and the rat H4IIE cell line Toxicology 15-12-2003;194:77-93 1322 1323 [78] Bittner M, Hilscherova K, Giesy JP: In vitro assessment of AhR-mediated activities of TCDD in mixture with humic substances Chemosphere 2009;76:1505-1508 1324 1325 1326 [79] van der Burg B, Winter R, Man HY, Vangenechten C, Berckmans P, Weimer M, Witters H, van der Linden S: Optimization and prevalidation of the in vitro AR CALUX method to test androgenic and antiandrogenic activity of compounds Reprod Toxicol 10-5-2010 1327 1328 1329 1330 [80] Freyberger A, Witters H, Weimer M, Lofink W, Berckmans P, Ahr HJ: Screening for (anti)androgenic properties using a standard operation protocol based on the human stably transfected androgen sensitive PALM cell line First steps towards validation Reprod Toxicol 27-10-2009 1331 1332 1333 [81] Molina-Molina JM, Hillenweck A, Jouanin I, Zalko D, Cravedi JP, Fernandez MF, Pillon A, Nicolas JC, Olea N, Balaguer P: Steroid receptor profiling of vinclozolin and its primary metabolites Toxicol Appl Pharmacol 1-10-2006;216:44-54 1334 1335 1336 [82] Willemsen P, Scippo ML, Kausel G, Figueroa J, Maghuin-Rogister G, Martial JA, Muller M: Use of reporter cell lines for detection of endocrine-disrupter activity Anal Bioanal Chem 2004;378:655-663 43 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 1337 1338 [83] Ghisari M, Bonefeld-Jorgensen EC: Effects of plasticizers and their mixtures on estrogen receptor and thyroid hormone functions Toxicol Lett 25-8-2009;189:67-77 1339 1340 [84] Shen O, Du G, Sun H, Wu W, Jiang Y, Song L, Wang X: Comparison of in vitro hormone activities of selected phthalates using reporter gene assays Toxicol Lett 1-12-2009;191:9-14 1341 1342 1343 [85] Zachow R, Uzumcu M: The methoxychlor metabolite, 2,2-bis-(p-hydroxyphenyl)-1,1,1trichloroethane, inhibits steroidogenesis in rat ovarian granulosa cells in vitro Reprod Toxicol 2006;22:659-665 1344 1345 1346 1347 [86] Lambrot R, Muczynski V, Lecureuil C, Angenard G, Coffigny H, Pairault C, Moison D, Frydman R, Habert R, Rouiller-Fabre V: Phthalates impair germ cell development in the human fetal testis in vitro without change in testosterone production Environ Health Perspect 2009;117:32-37 1348 1349 1350 1351 1352 1353 1354 1355 1356 [87] Anon OECD Guideline for the testing of chemicals draft proposal for a new guideline 4XX The H295R Steroidogenisis Assay 2010 http://www.oecd.org/dataoecd/56/11/44285292.pdf, assessed on July 2010 [88] Anon US-Epa: Integrated Summary Report on Aromatase http://www.epa.gov/endo/pubs/aromatase_isr.pdf, assessed on July 2010 2007 [89] Xu X, Yang W, Li Y, Wang YH: Discovery of estrogen receptor modulators: a review of virtual screening and SAR efforts Expert Opinion on Drug Discovery 2010;5:21-31 [90] Enoch SJ, Cronin MTD, Madden JC, Hewitt M Formation of structural categories to allow for read-across for teratogenicity [28], 696-708 2009 QSAR and Combinatorial Science 1357 1358 1359 1360 1361 1362 1363 1364 1365 [91] Hewitt M, Ellison CM, Enoch SJ, Madden JC, Cronin MTD Integrating (Q)SAR models, expert systems and read-across approaches for the prediction of developmental toxicity [29], 147-160 2010 Reproductive Toxicology 1366 1367 1368 [93] Jensen GE, Niemela JR, Wedebye EB, Nikolov NG: QSAR models for reproductive toxicity and endocrine disruption in regulatory use a preliminary investigation SAR QSAR Environ Res 2008;19:631-641 1369 1370 1371 [94] Madden JC: In silico approaches for predicting ADME properties.; in Puzyn T, Leszczynski J, Cronin MTD (eds): Recent Advances in QSAR Studies – Methods and Applications Springer, Dordrecht, 2010, pp 283-304 1372 1373 [95] Hewitt M, Madden JC, Rowe PH, Cronin MT: Structure-based modelling in reproductive toxicology: (Q)SARs for the placental barrier SAR QSAR Environ Res 2007;18:57-76 1374 1375 1376 [96] Cronin MTD , Hewitt M: In silico models to predict passage through the skin and other barriers.; in Testa B, van de Waterbeemd H (eds): Comprehensive Medicinal Chemistry II Elsevier, Oxford, 2007, vol 5, pp 725-744 1377 [97] Devillers J: Endocrine Disruption Modeling CRC Press, Boca Raton FL, USA, 2009 1378 1379 [98] Vedani A, Smiesko M: In silico toxicology in drug discovery - concepts based on threedimensional models Altern Lab Anim 2009;37:477-496 [92] Cronin MTD, Worth AP: (Q)SARs for predicting effects relating to reproductive toxicity Qsar & Combinatorial Science 2008;27:91-100 44 Working Group 5: Reproductive Toxicity DRAFT FOR CONSULTATION 14.07.10 [99] Pedersen F, de Bruijn J, Munn S & van Leeuwen K Assessment of additional testing needs under REACH – effects of (Q)SARs, risk based testing and voluntary industry initiatives 33 p 2003 http://ecb.jrc.ec.europa.eu/documents/REACH/PUBLICATIONS/REACH_testing_needs_fina l.pdf, JRC Report EUR 20863 1380 1381 1382 1383 1384 1385 1386 1387 1388 1389 [100] Zuang V, Eskes C: Alternative (non-animal) methods for cosmetics testing: Current status and future prospects - A report prepared in the context of the 7th amendment of the cosmetic directive for establishing the timetable for phasing out animal testing - Foreword AtlaAlternatives to Laboratory Animals 2005;33:1-+ 1390 1391 1392 [101] Curren RD, Southee JA, Spielmann H, Liebsch M, Fentem JH, Balls M: The Role of Prevalidation in the Development, Validation and Acceptance of Alternative Methods AtlaAlternatives to Laboratory Animals 1995;23:211-217 1393 1394 1395 [102] Hartung T, Bremer S, Casati S, Coecke S, Corvi R, Fortaner S, Gribaldo L, Halder M, Hoffmann S, Roi AJ, Prieto P, Sabbioni E, Scott L, Worth A, Zuang V: A modular approach to the ECVAM principles on test validity Altern Lab Anim 2004;32:467-472 1396 1397 1398 [103] Martin MT, Judson RS, Reif DM, Kavlock RJ, Dix DJ: Profiling chemicals based on chronic toxicity results from the U.S EPA ToxRef Database Environ Health Perspect 2009;117:392399 1399 1400 1401 [104] Bremer S, Pellizzer C, Hoffmann S, Seidle T, Hartung T: The development of new concepts for assessing reproductive toxicity applicable to large scale toxicological programmes Curr Pharm Des 2007;13:3047-3058 1402 1403 1404 1405 1406 [105] Krewski D, Acosta D, Jr., Andersen M, Anderson H, Bailar JC, III, Boekelheide K, Brent R, Charnley G, Cheung VG, Green S Jr, Kelsey KT, Kerkvliet NI, Li AA, McCray L, Meyer O, Patterson RD, Pennie W, Scala RA, Solomon GM, Stephens M, Yager J, Zeise L: Toxicity testing in the 21st century: a vision and a strategy J Toxicol Environ Health B Crit Rev 2010;13:51-138 45 ... systems, and on the growth 205 and development of the offspring, including gonadal function, the oestrus cycle, mating 206 behaviour, conception, gestation, parturition, lactation, and weaning, and. .. to provide initial information on possible effects on reproduction and/ or development 239 This test does not provide complete information on all aspects of reproduction and 240 development In... Interaction with OECD TG 415 androgen OECD TG 414 receptor OECD TG 441 DRAFT FOR CONSULTATION 14.07.10 Binding and induction/inhibition of reporter gene product Mechanistic studies Optimization 3-5

Ngày đăng: 05/03/2014, 16:20

Từ khóa liên quan

Mục lục

  • 1. Executive Summary

  • 2. Introduction

    • 2.1. Complexity of the Reproductive Cycle

    • 2.2. Alternatives for Reproductive Toxicity Testing

    • 3. Information Requirements for the Safety Assessment of Cosmetic

    • 4. Inventory of Animal Test Methods Currently Used for the Evaluation of Developmental and Reproductive Toxicity

      • 4.1. OECD Test Guideline 414: Prenatal Development Toxicity Study for the Testing of Chemicals

      • 4.2. OECD Test Guideline 415: One-Generation Reproduction Toxicity Study

      • 4.3. OECD Test Guideline 416: Two-Generation Reproduction Toxicity

      • 4.4. OECD Test Guideline 421: Reproduction/Developmental Toxicity Screening Test

      • 4.5. OECD Test Guideline 422: Combined Repeated Dose Toxicity Study with the Reproduction/Developmental Toxicity Screening Tes

      • 4.6. OECD Test Guideline 426: Developmental Neurotoxicity Study

      • 4.7. OECD Test Guideline 440: Uterotrophic Bioassay in Rodents: A short-term screening test for oestrogenic properties

      • 4.8. OECD Test Guideline 441: Hershberger Bioassay in Rats: A Short-term Screening Assay for (Anti-) Androgenic Properties

      • 4.9. OECD Test Guideline 455: The Stably Transfected Human Estrogen Receptor-α Transcriptional Activation Assay for Detection

      • 4.10. Draft OECD Test Guideline Extended One Generation Reproductive Toxicity Study

      • 5. Inventory of Alternative Methods

        • 5.1. Developmental Toxicity

          • 5.1.1. Whole Embryo Tests

            • 5.1.1.1. The Rodent Whole Embryo Culture Test

            • 5.1.1.2. The Zebrafish Embryo Teratogenicity Assay

            • 5.1.1.3. Frog Embryo Teratogenesis Assay Xenopus (FETAX)

            • 5.1.1.4. The Chicken Embryotoxicity Screening Test (CHEST)

            • 5.1.2. The Micromass Test

            • 5.1.3. Pluripotent Stem Cell Based in vitro Tests

Tài liệu cùng người dùng

Tài liệu liên quan