Sie sind auf Seite 1von 72

Specialized Toxicity

Studies:
Reproductive and
Developmental Toxicity1

Himanshu Gupta
Why This Testing
• All life depends on reproduction and development
• Individuals can live without reproducing but population cannot survive without
individuals reproducing
• Primary question - What effects this process and harms a child’s potential?
• Function of reproductive system is not tested in standard pre-clinical studies
• Micro-pathology may point towards adverse effects upon testis/ovaries and
reproductive accessory organs
2
Human Reproductive Testing

• There are no follow-up tests in humans to check on reproductive function.

• Effects seen in humans are likely to be retrospective and may not be detected until
considerable damage has been done - Thalidomide.

• Human reproductive processes are considerably less efficient than in animal models.

3
Ancient Awareness
• Many ancient cultures had fertility goddess
• Many ancient documentation of malformations
• Malformations rich aspect of mythology
• 6500 BC Turkey - figurine of conjoined twins
• 4000-5000 BC Australia drawings of twins
• 2000 BC – Tablet of Nineveh describes 62 malformations and predicts the future

4
Historical Awareness
• 15-16th centuries malformations caused by the devil, mother and child killed
• 1830s – Etienne Geoffroy Saint Hilaire experimented with chicken eggs
• 1900s began acceptance of malformations related to genetics
• 1940s – Josef Warkany opined that environmental factors affect rat development

5
Historical Events

1941 Human malformations linked to rubella virus

1960s – Thalidomide found to cause malformations

1970s Alcohol related to developmental effects


Fetal Alcohol Syndrome (FAS) 6
Reproductive/developmental toxicity
• Any adverse effect on any aspect of male or female sexual structure or function,
• or on the conceptus or
• on lactation,
• which would interfere with the production or development of normal offspring
capable in turn of reproducing the species.

7
Types

• Two broad toxicity categories:-

• Reproductive -refers to structural and functional alterations that affect


reproductive competence in sexually mature males and females.

• Developmental -refers to adverse effects on the developing organism that


result from exposure prior to conception, during the prenatal period, or
postnatally up to the time of sexual maturity.
8
Reproductive toxicity include: Developmental toxicity include:

• Male fertility • Mortality

• Female fertility • Dysmorphogenesis (structural abnormalities)

• Parturition • Alterations to growth

• Lactation • Functional impairment

9
Teratology
1. The study of malformations or
serious deviations from the normal type in
organisms
2. The branch of science concerned
with the production, development, anatomy, and
classification of malformed fetuses.
3. Teratogen
• Any agent that causes a birth defect
• After Greek monster creating

A conjoined goddess (6500 BC) from Anatolia 10


Defining a Teratogen
• To be considered teratogenic, a candidate substance or process should
• Result in characteristic set of malformation indicating selectivity for certain
target organs
• Exert its effects at a particular stage of foetal development
• Show a dose dependent incidence
Teratogenic effects are not limited to only major malformations, but also include
IUGR and stillbirth (smoking), miscarriage and neurocognitive delay (alcohol)

11
Stage Humans Main Cellular Affected by
Process(es)
Blastocyst formation 0-16 days Cell division Cytotoxic drugs, alcohol

Organogenesis 17-60 days approx. Division Teratogens


Migration Teratogens
Differentiation Teratogens
Death Teratogens
Histogenesis and 60 days to term As above Miscellaneous drugs (eg
functional maturation alcohol, nicotine, antithyroid
drugs, steroids)
12
Critical Periods of Human Development

14

*Katzung, 13th ed
Trimester Drug Effects
Carbamazepine First Neural Tube Defects
Metronidazole First May be mutagenic
Warfarin First Hypoplastic nasal bridge, chondrodysplasia
Second CNS malformations
Third Risk of bleeding
Androgens Second Masculinization of female foetus
Lithium First, Second Ebstein’s anomaly
ACE Inhibitors Esp Second, Third Renal damage, hypocalvaria
Tricyclic antidepressants Third Neonatal withdrawal symptoms
SSRIs Third Neonatal abstinence syndrome, pulmonary HTN
Diazepam All Long term use – dependence
Phenytoin All Foetal hydantoin syndrome
Tetracycline All Discoloration of teeth, altered bone growth
Ethanol All Fetal alcohol syndrome, neurodevelopmental defects
15
Pregnancy Risk Categories - USFDA
Category Description
A Adequate, well controlled studies in pregnant women have not shown an increased risk of foetal
abnormalities
B Animal studies have revealed no evidence of harm to the foetus, however, there are no adequate and well-
controlled studies inToo
pregnant
manywomen
in an unhelpful category !!!
OR… Animal studies have shown an adverse effect, but adequate and well-controlled studies in pregnant
women have failed to demonstrate a risk to the foetus
C Animal studies have shown an adverse effect, or no animal studies have been conducted, and there are no
adequate and well-controlled studies in pregnant women
Background incidence of defects in human approx 3%
D Adequate, well-controlled or observational studies in pregnant women have demonstrated a risk to the
foetus. However, the benefits of therapy may outweigh the potential risk
X Adequate, well-controlled or observational studies in animals or pregnant women have demonstrated
positive
Oftenevidence of foetal
difficult to getabnormalities.
enough The useto
data of tell
the product
if realishuman
contraindicated
risk inorwomen
not who are or
may become pregnant. 16
Pregnancy and Lactation Labelling Regulations
Subsection Subheadings Description
Pregnancy Exposure Information (inc contact information) regarding a pregnancy
Registry (if applicable) exposure registry that monitors pregnancy outcomes

Risk Summary Statement that the drug is contraindicated during pregnancy


Risk statement based on human and animal data
Background birth defects and miscarriage rates
8.1 Pregnancy
Clinical considerations (if Disease associated maternal and/or embryo-foetal risk
applicable) Dose adjustment during pregnancy and PP period
Maternal, foetal/neonatal adverse reactions
Data Data on which risk summary and clinical considerations are
based
17
Pregnancy and Lactation Labelling Regulations
Subsection Subheadings Description
8.2 Lactation Risk Summary State that the drug is contraindicated during lactation
Presence of drug in human milk
Effects of drug on milk production/excretion and child
Clinical considerations Minimizing exposure to the breastfed infant
(if applicable) Monitoring infant for adverse reactions
Data (if applicable) Data on which Risk Summary and Clinical considerations are based

8.3 Females Pregnancy testing Recommendations or requirements for pregnancy testing before, during
and Males of and after drug therapy
Reproductive Contraception Recommendations or requirements for contraception use before, during
Potential and after drug therapy
Risk Summary Human and/or animal data suggesting drug-associated effects on fertility
18
6 areas of Reproduction Recognized…1/2
A. Premating to conception: adult male and female reproductive functions,
development and maturation of gametes, mating behavior, fertilization

B. Conception to implantation: adult female reproductive functions,


preimplantation development, implantation

C. Implantation to closure of the hard palate: adult female reproductive


functions, embryonic development, major organ formation
19
6 areas of Reproduction Recognized…2/2
D. Closure of the hard palate to the end of pregnancy: adult female reproductive
functions, fetal development and growth, organ development and growth

E. Birth to weaning: adult female reproductive functions, neonate adaptation to


extrauterine life, preweaning development and growth

F. Weaning to sexual maturity: postweaning development and growth, adaptation


to independent life, attainment of full sexual function 20
Basic Tests for Pharmaceuticals
• Fertility - “Segment I” covers phases A and B, both sexes combined or assessed
separately

• Embryo – foetal – “Segment II” covers phase C

• Pre and post-natal study – “Segment III” covers phases C through to F

21
Timing of studies with Clinical Trials

22
Prerequisites…1/3

• Animal species: rodent (rat) and non-rodent (rabbit)

• Housing and feeding conditions:

• Room temp- 22 (±3 °C) for rodents and 18 (±3 °C) rabbits.

• Humidity - 50-60%

• Artificial lighting-12 hours light, 12 hours dark


23
Prerequisites…2/3

• Unlimited supply of drinking water

• Preparation of the animals: Healthy animals acclimated to laboratory conditions for


at least 5 days and have not been subjected to previous experimental procedures.

• Females should be mated with males of the same species and strain and mating of
siblings should be avoided.

24
Prerequisites…3/3
• At least three dose levels should be used.
• The highest dose- induce some developmental and/or maternal toxicity (clinical
signs or a decrease in body weight) but not death or severe suffering.
• One intermediate dose level - minimal observable toxic effects.
• The lowest dose level- not produce any evidence of either maternal or
developmental toxicity.

25
Principle of the Tests…1/2

• The test chemical is administered in graduated doses to several groups of males and
females.
• Males should be dosed for a minimum of four weeks and up to and including the
day before scheduled kill
• Pre-mating dosing period in males, fertility may not be a particular sensitive
indicator of testicular toxicity.
• Therefore, a detailed histological examination of the testes is essential.
26
Principle of the Tests…2/2
• Histopathology of the male gonads, is considered sufficient to enable detection of
the majority of effects on male fertility and spermatogenesis.
• Females should be dosed throughout the study - This includes mating, the duration
of pregnancy and including the day before scheduled kill
• Duration of study, following acclimatization and pre-dosing oestrous cycle
evaluation, is dependent on the female performance and is approximately 63 day

27
What are we Looking for
• Adverse effects of treatment
• Dose relationships
• Evidence of different sensitivity of reproductive organ systems compared to other
systems
• Differential effects in mother and offsprings
• Safe “NOAEL” values

28
How do we Look…1/3
Clinical Observation Every day at same time Behaviour, signs of difficult parturition and signs
of toxicity including mortality
Body weight Weighed day 1 and then weekly During Pregnancy – D 0, 7, 14, 20, within 24 hours
of parturition and D 4 and 13 post partum
food/water consumption weekly
Offspring parameters Number, gender, Still birth, live birth, gross abnormalities, runts
Weight within 24 hours and atleast on D4 and D 13 post partum
Clinical Biochemistry Blood samples from defined site taken:
- from at least two pups per litter on day 4 after birth,
- from all dams and at least two pups* per litter at termination on day 13,
- from all adult males*, at termination
*Samples from day 13 pups and adult males are assessed for thyroid hormones (T4 and TSH) 29
How do we Look…2/3
Gross Necropsy At time of Adult:
sacrifice/death Macroscopically for any abnormalities or pathological changes with
special attention to reproductive system.
Number of implantation sites should be recorded.
Vaginal smears is examined in the morning on the day of necropsy to
determine the stage of the oestrous cycle
Weight of testes and epididymides
Dead or killed on D13 Pups:
Examined for gross abnormalities and of external reproductive genitals
for signs of altered development
Thyroid from 1 male and 1 female pup preserved.
Ovaries, testes, accessory sex organs (uterus and cervix, epididymides, prostate, seminal vesicles plus coagulating
glands), thyroid and all organs showing macroscopic lesions of all adult animals should be preserved 30
How do we Look…3/3

Histopathology Detailed analysis of ovaries, testes and epididymides (with special emphasis on
stages of spermatogenesis and histopathology of interstitial testicular cell
structure) of the animals of the highest dose group and the control group.
The other preserved organs including thyroid from pups and adult animals may
be examined when necessary.
Examinations should be extended to the animals of other dosage groups when
changes are seen in the highest dose group.

31
Reproductive Toxicity

• Male Fertility

• Female Reproduction & Developmental toxicity

• Female Fertility

• Teratogenicity

• Perinatal development
32
Male Fertility Study

• One rodent species (preferably rat)

• 3 dose groups and a control group:

• Dose selection : results of previous 14 or 28-day toxicity study

• Highest dose : showing minimal toxicity in systemic studies

• 6 adult male animals per group


33
Test substance by intended route of use
for minimum 28 days and 70 days max

Paired with female animals of proven


fertility in a ratio of 1:2

Drug treatment of male animals


continues during pairing

Pairing continued till detection of Sperm


in vagina or 10 days, whichever is earlier
34
Pregnant females examined after day 13 of
gestation

Males sacrificed at the end of the study

Weights of each testis and epididymis


recorded

Sperms from one epididymis examined for


motility and morphology

Other epididymis and both testes examined


for histology
35
Female Reproduction and Developmental
Toxicity Studies
• For all drugs proposed for women of child bearing age

• Segment I : Female fertility

• Segment II : Teratogenicity

• Segment III : Perinatal development

• Albino mice or rats used, segment II also includes albino rabbits as 2nd species
36
Female Fertility Study (Segment I)

• One rodent species (rat preferred)

• 3 graded doses

• Highest dose : doesn’t affect general health of parent animals (usually the MTD
from previous systemic studies)

• At least 15 males and 15 females per dose group

• Route of administration same as intended for therapeutic use 37


Drug administration : 28 days (males) & 14 days (females) before mating

Drug treatment continued during mating and gestation period

Females allowed to litter, medication continued till weaning of pups


Body weight, Food Clinical signs of Reproduction and Pathology Gross &
intake intoxication Parturition Micro

Pups : Physiology, Behavior, Pathology (sex-wise distribution noted)


38
Teratogenicity Study (Segment II)

• Rodent (preferably rat) and Non-rodent (rabbit)

• Drug administered throughout organogenesis

• 3 dose levels and a Control group:

• Highest dose: minimum maternal toxicity and lowest as proposed clinical dose or its multiple

• Route of administration : same as intended for humans

• At least 20 pregnant rats (or mice) and 12 rabbits, for each dose
39
40
Observation parameters
General Reproductive General Pathology

Uterus, Number Gross (all)


Signs of
intoxication Ovaries

Foetuses
Females

Gender Visceral
Products of (half)
Body weight
conception Length
Skeletal
(half)
Food intake Weight
41
Perinatal Study (Segment III)
• Specially if
• Drug to pregnant / nursing mothers for long periods
• Indications of possible adverse effects on fetus
• One rodent species (preferably rat)
• At least 4 groups (including control), 15 females/group
• Drug throughout last trimester (from day 15 of gestation)
• Dose causing low fetal loss continued throughout weaning
• Dosing comparable to multiples of human dose and route 42
F1 litter 1 male and 1 female from each group
(total 15 males and 15 females in each group)

Test substance given

Throughout growth to sexual life

Mating performance and fertility of F1: F2


generation

F2 generation growth parameters monitored till


weaning
43
Observation Parameters

• body weight, food intake, general signs of


Dams intoxication, progress of gestation/ parturition
periods and gross pathology.

• clinical signs, sex-wise distribution in dose groups,


body weight, growth parameters, gross examination,
Pups
survival and autopsy and where necessary,
histopathology.

44
Extended One Generation Reproductive
Toxicity Study
• Males and females are exposed 4 and 2 weeks premating respectively, and females
are exposed throughout the mating period, pregnancy and weaning of their pups.
• Male exposure is continued for 10 weeks to cover the entire spermatogenic cycle,
followed by necropsy.
• F1 animals are kept until adulthood and developmental landmarks are assessed.
• F1 are divided into up to three cohorts, one to assess their reproductive capacity,
one for developmental immune toxicity (DIT) assessment, and one for
developmental neurotoxicity (DNT) assessment
45
Developmental Neurotoxicity Study
• Provides information on the potential functional and morphological effects on
developing nervous system of the offspring of repeated exposure to a substance
during in utero and early postnatal development
• Can be conducted as a separate study, incorporated into a reproductive toxicity
and/or adult neurotoxicity study (e.g., Test Guidelines 415, 416, 424) or added onto
a prenatal developmental toxicity study (e.g., Test Guideline 414)
• Test substance is administered daily, generally orally, to mated females (rats are
preferred) from the time of implantation (GD 6) throughout lactation (PND 21).
46
• At least three dose levels and a concurrent control should be used and a total of 20
litters are recommended at each dose level.

• Dams are tested to assess effects in pregnant and lactating females.


• Offspring are randomly selected from within litters for neurotoxicity evaluation.
• All dams and all offspring should be carefully observed at least once daily with
respect to their health, including morbidity and mortality.
47
• Evaluation:
• gross neurological and behavioral abnormalities
• brain weights
• neuropathology during postnatal development and adulthood.
• Report should include the body weight, the food/water consumption; detailed
clinical observations, necropsy findings, number of animals at the start and at the
end of the study and the toxic response data by sex and dose level
48
Female-specific endpoints of reproductive
toxicity
Organ weights Ovary, uterus, vagina, pituitary
Histopathology and visual exam Ovary, uterus, vagina, pituitary, oviduct
Estrous (mentrual) cycle normality Vaginal smear cytology
Sexual Behavior Lordosis, time to mating, vaginal plugs
Hormone levels LH, FSH, estrogen, progesterone
Lactation Offspring growth, milk quantity and quality
Development Normality of external genitalia, vaginal opening, vaginal
smear cytology
Senescence Vaginal smear cytology, ovarian histology (menopause) 49
Male specific endpoints of reproductive
toxicity

50
Animal tests (OECD guidelines)
Type of study Test OECD
Developmental toxicity Prenatal developmental toxicity test TG 414
Developmental neurotoxicity study TG 426
Reproductive toxicity Reproductive/developmental toxicity TG 421
screening assay
One-generation reproductive toxicity study TG 415
Two-generation reproductive toxicity study TG 416
Extended one generation reproductive TG 443
toxicity study
51
Alternative methods…1/3
1. Developmental Toxicity
• Whole Embryo Tests
a) The Rodent Whole Embryo Culture Test
b) Zebrafish Embryo Teratogenicity Assay
c) Frog Embryo Teratogenesis Assay Xenopus (FETAX)
d) The Chicken Embryotoxicity Screening Test (CHEST)
• The Micromass Test
• Pluripotent Stem Cell Based in vitroTests
52
Alternative methods…2/3
2. Placental Toxicity and Transport
a) The Placental Perfusion Assay
b) Trophoblast Cell Assay
3. Preimplantation Toxicity
• Male Fertility
a) Computer Assisted Sperm Analysis
b) Leydig Cell Assay
c) Sertoli Cell Assay
d) ReProComet Assay 53
Alternative methods…3/3
• Female Fertility
a) Follicle Culture Bioassay (FBA)
b) In vitro Bovine Oocyte Maturation Assay (bIVM)
c) In vitro Bovine Fertilisation Test (bIVF)
4. In vitro Tests for Assessing Effects on the Endocrine System
a) Ishikawa Cell Test
b) Cell Proliferation Based Assays for Testing Estrogen Activity
c) Receptor Binding Assays
d) Transcriptional Tests 54
Rodent Whole Embryo Culture Test
• Evaluates drug effects after a 48-hour incubation period during mid-stage embryo
development.
• Embryos are typically removed from the uterus on gestation day 9 (early somite
stage), and the decidua and Reichert’s membrane are dissected away.
• They are placed in bottles with medium (primarily rat serum containing various
concentrations of the compound being tested) gassed with 5% oxygen.

55
Rodent Whole Embryo Culture Test
• These bottles are placed on a rotator and maintained at 37°C in an incubator for up
to 48 hours, with increasing oxygen concentrations appropriate for the
developmental stage.
• End points- All tissues receive a score dependent on their developmental stage, and
all scores added up give the Total Morphological Score (TMS), IC50mal
(concentration at which 50% of embryos are malformed) are calculated.

56
57
Advantages Disadvantages

• reduced animal use • technical skill required

• reduced cost and • restricted culture duration

• allows the direct and controlled • artificial route of administration of test


addition of small amounts of compounds

compounds is possible. • absence of the maternal compartment


58
Mouse Embryonic Stem Cell (ESC)
• The ability of mouse ESCs to shift from a pluripotent to differentiated state makes
it an excellent candidate as a screening tool for hazard identification.
• ESC lines are established from the inner cell mass of the 3 - day mouse blastocyst
• Compound is added to the culture media for up to 10 days.
• The number of days of treatment to measure assay endpoints varies

59
Mouse Embryonic Stem Cell (ESC)
• Endpoints: inhibition of growth (cytotoxicity), differentiation (e.g., beating
cardiomyocytes, differentiated neuronal cells), or transcriptomics to enable a more
detailed effect assessment on molecular endpoints (i.e., pluripotency and cell lineage
markers), which are expressed when ESCs differentiate along a specific lineage.
• Differentiation of these cell types in ESC cultures largely recapitulates some
embryonic processes that occur in vivo.
• Good value in predicting in-vivo developmental toxicity.

60
Mouse Embryonic Stem Cell (ESC)
Advantages Disadvantages
• After the initial isolation, there is no animal usage • No maternal component
• Reduced costs. • Low biological complexity
• Assesses early embryonic development when
ESCs begin to differentiate along lineages.
• It can be used as a high throughput tool for
screening of drugs because of the short duration
of the assay, small resource requirements, and its
amenability to automation.
• Amount of compound required to perform the
assay is small
61
Zebrafish Embryo Teratogenicity Assay
• Development of the zebrafish embryo is very similar to embryogenesis in higher
vertebrates, including humans and many molecular pathways are evolutionary
conserved between zebrafish and humans
• Fertilized fish eggs are exposed to different concentrations of a test substance
• At different time points, exposed developing fish embryos are observed and scored
for lethal, embryotoxic and/or teratogenic effects
• Used not only as a screening tool for teratogenicity but also as a means of
investigating specific mechanisms related to the teratogenic potential of certain
substances
62
Zebrafish Embryo Teratogenicity Assay

C. A 24-hpf embryo has been removed


from the chorion to facilitate viewing
B. Late blastula and
to illustrate
early gastrula how rapidly
period embryos embryonic
(approx.
A. Embryos are collected from egg traps shortly after
4-6 hpf) are development occurs; early eye, brain,
spawning (egplaced
1 hpf),into multiwell
rinsed culture
and selected forplates
use filled
with buffer heart,
(embryo somite, and
medium) containing notochord
several
concentrationsdevelopment are easily viewed in
of the test compound
transparent embryos
E. A typical
D. Hatching
developmental
is complete
toxicity
and the
assay
embryonic
with zebrafish
periodinvolves
ends at evaluation
3 dpf of
viability, growth, and morphology of early stage larvae (eg 5 dpf)
63
Zebrafish Embryo Teratogenicity Assay
Advantages
• Concordance between mammalian and zebrafish assays is strong and may be higher
than 80%
• Especially strong as a tool for mechanistic studies in developmental toxicology
• There are many experimental tools available that work particularly well in zebrafish,
especially molecular and imaging techniques. Examples include functional genomics
approaches, transgenic or mutant lines, microinjection, and fluorescent dyes.
64
Frog Embryo Teratogenesis Assay Xenopus
(FETAX)
• Whole embryo screening assay, based on the South African clawed frog Xenopus
laevis to identify substances that may pose a developmental hazard in humans.
• fertilized eggs in the mid- to late-blastula stage are incubated in media containing
the test substance for 96 h.
• The embryos are scored for lethality, growth retardation and malformations at
different timepoints.
• FETAX encompasses organogenesis and does not include later events of
development.
65
Chicken Embryotoxicity Screening Test
(CHEST)
• First described in 1976 by Jelinek et al. fast and cheap teratogenicity test.
• Two phases of testing:
• CHEST I determines toxic dose range in early administration time (24 hours)
• CHEST II- determines the teratogenic dose range and covers late effects on the
embryo development (days 2, 3 and 4).
• Endpoints - mortality, malformations, embryo development, blood vessel
development and blood vessel coloration.
• Disadvantages- not able to distinguish general toxicity from specific developmental
effects and the absence of mammalian maternal-fetal relations. 66
Micromass Test
• Make use of cell cultures of the limb bud and/or neuronal cells
• Cells are isolated from the limb or the cephalic tissues of mid-organogenesis
embryos
• After preparing a single cell solution the cells are seeded in a high density and
undergo differentiation into chondrocytes and neurons without additional
stimulation
• The differentiation after exposure to test chemicals is analysed by using defined
toxicological endpoints
67
Other Tests

68
Test Part of mechanism covered Area of application
Placenta perfusion Human ex vivo model to assess Hazard identification that
system Transplacental transfer indicating foetal might lead to fertility impairments
exposure and metabolism of test
chemicals

Trophoblast cell Toxicity assessment during embryo Hazard identification that


assay implantation might lead to fertility impairments

Mouse embryo Fertilisation, first cleavage embryo Hazard identification that


bioassay development might lead to fertility impairments

Follicle bioassay completion of oocyte meiosis up to the Hazard identification that


(FBA) metaphase II Oogenesis: oocyte yield; might lead to fertility impairments
diameter; nuclear maturation
69
Test Part of mechanism covered Area of application

Bovine completion of oocyte meiosis up to the Hazard identification that


maturation test metaphase II might lead to fertility
impairments

Bovine Formation of female and male pronuclei Hazard identification that


fertilisation test after penetration of capacitated bull might lead to fertility
spermatozoa into impairments
matured oocytes
ReProComet DNA strand breaks and alkali labile sites in Hazard identification that
assay bull sperm might lead to fertility
impairments

CASA test multiple leading to impairments in Hazard identification that


motility, and viability as well as might lead to fertility
morphology of spermatozoa impairments
70
Conclusion
• Reproductive and developmental toxicity aims to predict effects of medicines and
chemicals on the ability of a man to reproduce by assessing effects in animals.
• Study designs divide the total process into a series of manageable units which can
provide answers to specific challenges to the reproductive processes.
• No real prospects of replacing animals in these studies.
• Challenge is to build on alternative tests in generating a testing strategy in which the
most sensitive end points are combined in a well-informed selection of alternative
assays.
71
References
• Schedule Y of Drugs & Cosmetics Act.
• OECD Guideline for the Testing of Chemicals. Available
from:http://www.oecd.org/dataoecd/20/52/37622194.pdf.
• Chapter 5 - Reproductive Toxicity, Compiled by Workgroup 5-14 July 2010.
• ICH harmonised tripartite guideline. Detection of toxicity to reproduction for medicinal
products & toxicity to male fertility S5(R2).
• Guidance for Industry Reproductive and Developmental Toxicities — Integrating Study
Results to Assess Concerns.
• Parasuraman S. Toxicological screening. J Pharmacol Pharmacother. 2011;2(2):74–79.
72
THANK YOU

73

Das könnte Ihnen auch gefallen