You are on page 1of 91

Non-clinical Studies:

Non-
Requirements for Human
Clinical Studies
Kesara Na-
Na-Bangchang
Ph.D. (Pharmacology)
Graduate Program in Bioclinical Sciences
Chulabhorn International College of Medicine
Thammasat University
Learning Objectives
 To describe the objectives & various types
of non-clinical study requirements for
human clinical studies

 To describe various approaches for


determining first in man (FIM) dose
based on non-clinical data
Outline
 Objectives of non-clinical studies
 Types of non-clinical studies
 Pre-clinical requirements to support
first-in-man Phase I
Development Stage of New Drugs
Quality 2-10 ys (ave. 5ys) Common
30d
Technical

NDA submission
CMC, stability Investigator’s Brochure
Document
Seeds, screening

Post marketing
IND Clinical trials

Pre--clinical
Pre Phase 1

NDA review

Approval
PK/PD Phase 2
Phase 3
Toxicology

What kind of pre-clinical data


are necessary to get approval ?
Types of Non-
Non-clinical Studies

 Full evaluation of the clinical candidates


 Translation from animal to human

 Pharmacological studies
 Pharmacokinetic studies
 Toxicological studies
Relevance of Non-
Non-clinical
Studies in Drug Development
Basic Goals:
Identification of the pharmacological properties:
• Pharmacodynamics (mode of action)
• Safety (pharmacology)
• Pharmacokinetics (ADME)
• Comparative physiology (extrapolation of animal data to
humans)
Understand the toxicological profile:
• Characterize potential adverse drug effects (define organ
toxicity & reversibility)
• Establish a safe initial dose level of the first human exposure
• Identify parameters for clinical monitoring of potential adverse
effects
• Special toxicity (e.g. genotoxicity, carcinogenicity, reproductive
toxicity)
Non--clinical Studies During Discovery Phase
Non
Non--clinical Studies During Discovery Phase
Non
Non--clinical Studies During Development Phase
Non
Who has to comply with Good Clinical
Practice (GLP) regulations?

GLP is needed for: GLP is not needed for:

• Basic research
Non-clinical safety studies of • Studies to develop new analytical
development of drugs: methods
• Toxicity tests • Chemical tests used to drive the
• Toxicokinetics specifications of marketed food
• Local tolerance studies product
• Mutagenicity, Carcinogenicity,
Emebryotoxicity tests, etc
• Agricultural pesticide development
• Development of toxic chemicals
• Food control (food additives)
• Test of substance with regard to
explosive hazards
Non--clinical Study Requirements
Non
Module 4:
4.2.1 Pharmacology
4.2.1.1 Primary pharmacodynamics
4.2.1.2 Secondary pharmacodynamics
4.2.1.3 Safety pharmacology
4.2.1.4 Pharmacodynamic drug interactions

4.2.2 Pharmacokinetics
4.2.2.1 Analytical methods & validation reports
4.2.2.2 Absorption
4.2.2.3 Distribution
4.2.2.4 Excretion
4.2.2.5 Pharmacokinetic drug interactions
4.2.2.6 Other pharmacokinetic drug interactions

4.2.3 Toxicology
4.2.3.1 Single-dose toxicity
4.2.3.2 Repeated-dose toxicity
4.2.3.3 Genotoxicity
4.2.3.4 Carcinogenicity
4.2.3.5 Reproductive & developmental toxicity
4.2.3.6 Local tolerance
4.2.3.7 Other toxicity tests
Pharmacological Studies
Pharmacology
In vitro/in vivo studies related to the
mechanisms of action:
• Primary PD Studies: Proposed mechanism of action
• Secondary PD Studies: Identification of effects not
related to desired therapeutic target
Safety Pharmacology
(ICH S7
S7A, S
S7
7B)

 A series of protocol-driven studies, aimed


specifically at detecting possible undesirable or
dangerous effects of exposure to the drug in
therapeutic doses

 Effects on specific organ systems, e.g., CNS,


CVS, respiratory systems, etc

 Drug-induced long QT syndrome (ICH S7B)


Activation of Other Receptors/Enzymes

 hERG = ‘human ether-a-go-go related


gene’ (KCNH2) that codes for a protein
known as Kv11.1, the alpha subunit of a
potassium ion channel
 Activation causes prolongation of
electrical impulses regulating heart
beat
 Long QT syndrome: QTc > 440 msec.
 Can lead to fatal arrhythmias

‘T’ wave is delayed


R R

P T P T
QS QS
Normal heart beat Activation of hERG
Pharmacokinetic Studies
 Comparison of ADME in species used
for toxicology studies and extrapolation
to humans
 Drug-drug interaction, bio availability, etc.
 May suggest modification in the intended
doses, routes or schedule for the clinical trials
 Assist in:
o Identification of bioaccumulation potential
o Identification of potential differences in gender
In vitro toxicity profile of terfenadine
terfenadine:: an
example of a false positive
Toxicological Studies
GOALS (ICH S6S6):
 To identify an initial safe starting dose for Phase I studies
and subsequent dose escalation schemes in humans
 To identify potential target organs for toxicity and potential
reversibility
 To identify safety parameters for clinical monitoring
 To assess relationship to exposure
 To assess dose dependence
 To assess hazards that cannot be evaluated in clinical trials
(e.g. carcinogenicity and teratogenicity)

TK may be incorporated.
Toxicity & Safety Assessments
IND NDA
GLP

Pre-clinical
Discovery Development Clinical Phase I-III Clinical Phase IV

Exploratory Toxicology
In vitro & In silico screen Single & repeat dose
range finding studies
• Mutagenicity
in 2 spp.
• Cytotoxicity
Regulatory Toxicology
• Immunogenicity
Safety pharmacology 3-12 month chronic 24 month
• Hepatotoxicity toxicity in 2 spp. carcinogenicity
Genotoxicity (in vitro Reproductive toxicity in in 2 spp.
• Embryotoxicity
& 1 spp.
in vivo) - Fertility &
• 28-day repeat dose implantation
toxicity in 2 spp. - Fetal development
- Pre- & post-natal
effects
Predictive Value of Non-
Non-clinical
Safety Evaluation
Safety Pharmacology
TYPE Physiological Tests
system
Core CNS Observations on conscious animals
battery -Motor activity, Behavioral changes
-Coordination, Reflex responses
-Body temperature
Cardiovascular Measurements on anaesthetized
animals
-BP, Heart rate, ECG changes
-Tests for delayed ventricular
repolarization
Respiratory Measurements on anaesthetized or
conscious animals
-Respiratory rate, Tidal volume
-Arterial oxygen saturation
TYPE Physiological Tests
system
Follow-up CNS Tests on learning & memory
tests More complex test for changes in
(examples) behavior & motor function
Tests for visual & auditory function
Cardiovascular Cardiac output
Ventricular contractility
Vascular resistance
Regional blood flow
Respiratory Airways resistance & compliance
Pulmonary arterial pressure
Blood gases
TYPE Physiological system Tests
Supplementary tests Renal function Urine volume, Osmolality, pH
(examples) Proteinuria
Blood urea/Creatinine
Fluid/Electrolyte balance
Urine cytology
Autonomic nervous CVS, GI, Respiratory responses
system to agonists and stimulation of
autonomic nerves
Gastrointestinal Gastric secretion
system Gastric pH
Intestinal motility
Gastrointestinal transit time
Others (Endocrine,
Blood coagulation,
Skeletal muscle
function…)
Non-clinical Requirements To
Non-
Support First-
First-In
In--Man (FIM) –
Phase I
 Basic package of non-clinical studies for Phase I trials
 Reduced non-clinical requirements for Phase 0 trials
 Non-clinical safety testing continues during clinical
development
Core Toxicology Evaluation
Single-dose & repeated dose studies up
to 14 days duration:

 Single dose in 2 mammalian species (ICH M3)


 14-day repeated dose in rodents and non-rodents
(ICH M3)
 TK in rodents & non-rodents

 A control and > 3 drug concentrations


• identification of the NOAEL and MTD
• identify shape of the dose-response curve
MTD

 LOAEL (Low Observed Adverse Effect Level) is the lowest dose at


which there is an observed toxic or adverse effect
 NOAEL (No Observed Adverse Effect Level): the highest dose tested
in animal species that does not produce a significant increase in
adverse effects in comparison to the control group
 MTD (Maximum tolerated dose): the highest dose tested in animal
species that produces the desired effect without unacceptable
toxicity
Other Toxicology Evaluation

• Genotoxicity
• Reproductive toxicity
• Carcinogenicity
• Immunotoxicity
• Local tolerance
M3: The Primary ICH Guideline for
Non--clinical Requirement for
Non
Clinical Trials

ICH M3 (R2)
(Jul 2008)
ICH M3 (R2) Requirements (new)
1. Toxicity studies
2. Safety pharmacology (CV, CNS, RS)
3. PK and TK (ADME)

Maximum duration of Clinical Minimum duration of repeated dose toxicity studies


trial
Rodent Non-rodent

Up to 2 weeks 2 weeks 2 weeks


Between 2 weeks and 6 Same as clinical trial Same as clinical trial
months
> 6 months 6 months 9 months

+ Genotoxicity in vitro (Ames test, Chromosome abberation)


+ Male/ fertility
ICH M3
M3 (R2
(R2) Revision
Exploratory Clinical Trial (Phase 0)
ICH M3 (R2) June 2009)

 Conducted early in Phase I


 Clinical exploratory approach to enable sponsors to more
efficiently identify and develop promising compounds
under an IND, based on a more limited non-clinical data
set than that required for a traditional Phase I study
5 Clinical Approaches supported by more limited
non-clinical testing programs to allow better
informed decision about whether or not to progress
within the compound
• Involved limited exposure
• No therapeutic diagnostic intended
• No intention to estimate MTD
Approach 1 Microdose max 100 µg Microdose
Approach 2 Microdose 5 x 100 µg

Approach 3 Single dose


Anticipated therapeutics
range Therapeutic dose
Approach 4
Dosing up to 14 days
Approach 5
Information Obtained from Exploratory IND

 PK including biodistribution (using imaging


technologies, e.g. PET)
 Selecting the most promising lead product
from a group of candidates
 Understanding the relationship between a
MOA and the treatment of a disease. (e.g.,
binding to target or enzyme inhibition)
Phase I vs.
vs. Exploratory (Phase 0)
Trials
Phase I Exploratory
API (kg) 1-3 kg 0.1-0.3 kg
Non-clinical 9-12 studies 5-6 studies
studies 220 rodents 170 rodents
38 non-rodents 6 non-rodents
12-18 months 6-12 months
Advantages Target organs PK-PD
identified Help for GO/NO-GO
Clinical MTD
Drawbacks API No MTD
More time for Delayed Phase II
GO/NO-GO

API = Active pharmaceutical ingredient


Dose Translation from Animals
to Human
First-In-Human (FIH) Study
The Bridge from Animal to Human

Success Rates from FIH to Registration


The data from the ten biggest drug companies during
1991-2000
Kola I and Landis J. Nat Rev Drug Discov (2004)
Starting Dose in Phase I Trials

 Toxicological Approach
 Similar Dose Approach
 Pharmacologically Guided Dose Extrapolation
(Allometric Scaling)
 PB-PK Modeling
1. Toxicological Approach

Calculation of the Maximum


Recommended Starting Dose (MRSD)
Maximum Recommended Starting Dose (MRSD)
MRSD: The highest dose recommended as the initial dose in
a clinical trial

(1) Determination of NOAEL in relevant species


(2) Conversion in Human Equivalent Dose (HED),
corrected with body surface area conversion factor
(3) Apply safety factor (usually 10)

MRSD = HED
Safety factor
= NOAEL
Safety factor * BSA conversion factor
1. The Dose-By-Factor Approach:
 NOAEL of the drug is scaled by using simple allometry
on the basis of body surface area (BSA) to obtain
human equivalent dose (HED) (USFDA 2005)
Increasing the Safety Factor:
• Steep dose-response curve
• Severe toxicities
• Non-monitorable toxicity
• Toxicities without premonitory signs
• Variable bioavailability
• Irreversible toxicity
• Unexplained mortality
• Large variability in doses or plasma drug levels
eliciting effects
• Non-linear pharmacokinetics
• Inadequate dose-response data
• Novel therapeutic targets
• Animal models with limited utility
(4) Consideration of the pharmacologically
active dose (PAD)??:
PAD: The lowest dose tested in an animal species with the
Intended pharmacological activity

• Examples: toxicity from exaggerated pharmacologic


effects (e.g. anticoagulants, vasodilators)
• PAD derived from appropriate PD models
• If the PAD is obtained from an in vivo study, an HED
can be derived from a PAD estimate using a BSA-CF
(and should be compared with MRSD)
• If the pharmacologic HED < MRSD, decrease the FIH dose
Selection of MRSD
Tragedy of Research: TGN1412 Study
Problems
o Signify precaution in the interpretation of pre-clinical
(primate) studies
o TGN1412 (TeGenero): CD28 superagonist antibody
(activation of regulatory T cells regardless of signal received
by T-cell receptor)
o Pre-clinical in primates:
 Well-tolerated expansion of T cells without any measurable
pro-inflammatory reaction
 Therapeutic potential for auto-immune diseases
o Phase I (2006): immediately withdrawn due to multi-
organ failure (500 times lower dose than the safe dose
in animal studies)
Changes Triggered By TGN 1412

o European guidelines for FIM phase-I clinical


trials of biologics:
 Revision of the pre-clinical studies with immune-
modulating drugs (and other high-risk drugs)
 Revision of the dose selection algorithm for
immunological agents (MABEL instead of NOAEL)
 Design of FIM phase I trials
 Qualification of investigator
 Regulatory changes – more rigorous evaluation of pre-
clinical studies

Ref.EMEA/CHMP/SWP/28367/07
Dose Calculation for the Starting Dose:
New Approach for High Risk Medicinal Products
(CHMP 2007)

 When there are concerns that serious adverse


reactions in FIM clinical trials may occur:
• Uncertainties on the MOA (novelty), nature of the
target, relevance of animal models

 Consider the MABEL (Minimal Anticipated Biological


Effect) approach based on biological effect
Calculation by MABEL Approach

Use all relevant in vitro & in vivo information


from the PD and the PK:
• Receptor binding and receptor occupancy
• Concentration-response curves
• Exposure at PK doses in the
relevant species
Minimal
Effect
Example of an MRSD based on the MABEL for an
agonist mAb
2. Similar Dose Approach

 Can be used when:


Drugs in the same class of index drug with similar
PK and PD
 Assume that the ratio of the starting dose/NOAEL will
be the same for both compounds

 The optimal starting dose of the index drug is defined as


a single dose that produces no PD response including
toxic effects
Example:
• Drug X belongs to the same class as drug A, which is
licensed for used in humans; NOAEL = 3 mg/kg/day
• Drug A:
• Pre-clinical toxicological studies revealed that
the most sensitive species was rats with a NOAEL of
2 mg/kg/day
• The optimal starting dose of drug A is 30 mg/kg/day
• So:
Dose X /NOAEL X = Dose A/NOAEL A
Dose X = Dose A * NOAEL X
NOAEL A
= 30 * 3
2
= 45 mg/kg/day
3. Pharmacologically Guided Dose
Extrapolation (Allometric
(Allometric Scaling)
STEPS:
• The NOAEL and AUC are determined in 6 species
and the species that gives the lowest NOAEL is
used as the index species for scaling
• Clearance (CL) in the index species is scaled
allometrically to obtain the estimated CL in
humans

FIH starting dose = AUC (index species) * Human estimated CL


Safety factor
Assumptions:
• Linear PK
• Only parent drug is active
Example:

• AUC obtained at the NOAEL for drug A = 23.5 µg*hr/ml


in rats
• Predicted CL in humans (from scaling) = 20.0 L/hr.
• Safety factor applied is 10

FIH starting dose = AUC (index species) * Human estimated CL


Safety factor

• Starting dose = 23.5 * 20.0 = 470 mg


4. Physiological-
Physiological-Based Pharmacokinetic
(PBPK) Modeling
 Taken into account a range of physiological variables
(transit time, change in pH during GI transit, blood flow,
plasma & tissue protein binding)
 Highly depend on obtaining multiple parameters from
in vitro & in vivo experiments – CL (predicted human CL)
 A useful tool, especially if non-linear PKs or non-
metabolic routes
Application of PBPK Modeling to Low
Dose/Interspecies Extrapolation

Developing a Human PBPK Model:


o Use the tissue: blood partition coefficients
developed from the animal model, or by physical
chemical extrapolation
o Use values for organ clearance based on either
human experimental data (in vivo or in vitro) OR by
allometric extrapolation developed in at least two
other species
Animal scaling
Resources
Guidances and Guidelines:
o ICH- http://www.fda.gov/cder/guidance/index.htm
 S1 Carcinogenicity
 S2 Genetic toxicity
 S3 Toxicokinetics
 S4 Duration of Chronic Toxicity Testing
 S5 Reproductive toxicity
 S6 Biotechnology
 S7 Safety Pharmacology
 M3 Nonclinical Safety Studies for the conduct of
Human Clinical Trials
Tragedy of Research: TGN1412 Study
Problems
o Signify precaution in the interpretation of pre-clinical
(primate) studies
o TGN1412 (TeGenero): CD28 superagonist antibody
(activation of regulatory T cells regardless of signal received
by T-cell receptor)
o Pre-clinical in primates:
 Well-tolerated expansion of T cells without any measurable
pro-inflammatory reaction
 Therapeutic potential for auto-immune diseases
o Phase I (2006): immediately withdrawn due to multi-
organ failure (500 times lower dose than the safe dose
in animal studies)
Changes Triggered By TGN 1412

o European guidelines for FIM phase-I clinical


trials of biologics:
 Revision of the pre-clinical studies with immune-
modulating drugs (and other high-risk drugs)
 Revision of the dose selection algorithm for
immunological agents (MABEL instead of NOAEL)
 Design of FIM phase I trials
 Qualification of investigator
 Regulatory changes – more rigorous evaluation of pre-
clinical studies

Ref.EMEA/CHMP/SWP/28367/07
Dose Calculation for the Starting Dose:
New Approach for High Risk Medicinal Products
(CHMP 2007)

 When there are concerns that serious adverse


reactions in FIM clinical trials may occur:
• Uncertainties on the MOA (novelty), nature of the
target, relevance of animal models

 Consider the MABEL (Minimal Anticipated Biological


Effect) approach based on biological effect
Calculation by MABEL Approach

 Use all relevant in vitro & in vivo information


from the PD and the PK:
• Receptor binding and receptor occupancy
• Concentration-response curves
• Exposure at PK doses in the
relevant species
Minimal
Effect
Example of an MRSD based on the MABEL for an
agonist mAb
2. Similar Dose Approach

 Can be used when:


• Drugs in the same class of index drug with similar
PK and PD
 Assume that the ratio of the starting dose/NOAEL will
be the same for both compounds

 The optimal starting dose of the index drug is defined as


a single dose that produces no PD response including
toxic effects
Example:
• Drug X belongs to the same class as drug A, which is
licensed for used in humans
• Preclinical toxicological studies revealed that the
most sensitive species was rats with a NOAEL of
2 mg/kg/day
• The optimal starting dose of drug A is 30 mg/kg/day
• So:
Dose X/NOAEL X = Dose A/NOAEL A
Dose X = Dose A * NOAEL X/NOAEL A
= 10 * 2/14
= 1.4 mg/kg
3. Pharmacologically Guided Dose
Extrapolation (Allometric
(Allometric Scaling)
STEPS:
• The NOAEL and AUC are determined in 6 species
and the species that gives the lowest NOAEL is
used as the index species for scaling
• Clearance (CL) in the index species is scaled
allometrically to obtain the estimated CL in
humans

FIH starting dose = AUC (index species) * Human


estimated CL/ Safety factor
Assumptions:
• Linear PK
• Only parent drug is active
Example:

• AUC obtained at the NOAEL for drug A = 23.5 µg*hr/ml


in rats
• Predicted CL in humans (from scaling) = 20.0 L/hr.
• Starting dose = 23.5 * 20.0 = 470 mg
• If the safety factor applied is 10, the starting dose
is 47 mg
4. Physiological-
Physiological-Based Pharmacokinetic
(PBPK) Modeling
 Taken into account a range of physiological variables
(transit time, change in pH during GI transit, blood flow,
plasma & tissue protein binding)
 Highly depend on obtaining multiple parameters from
in vitro & in vivo experiments – CL (predicted human CL)
 A useful tool, especially if non-linear PKs or non-
metabolic routes
Application of PBPK Modeling to Low
Dose/Interspecies Extrapolation

Developing a Human PBPK Model:


o Use the tissue: blood partition coefficients
developed from the animal model, or by physical
chemical extrapolation
o Use values for organ clearance based on either
human experimental data (in vivo or in vitro) OR by
allometric extrapolation developed in at least two
other species
Animal scaling
Annex: Non-
Non-clinical Study
Guidances
Pharmacology
• Non-clinical Safety Studies for the Conduct of Human Clinical Trials and
Marketing Authorization for Pharmaceuticals (ICH M3(R2)) Dec 2009
• Guideline on the Strategies to Identify and Mitigate Risks for First-in
Human Clinical Trials with Investigational Medicinal Products (CHMP/
SWP/28367/07) Sep 2007
• Safety Pharmacology Studies for Human Pharmaceuticals (ICH S7A)
Jun 2001
Pharmacokinetics
• Pharmacokinetics: Guidance for Repeated Dose Tissue Distribution
Studies (ICH S3B) Jun 1995
• Toxicokinetics: The Assessment of Systemic Exposure in Toxicity
Studies (ICH S3A) Jun 1995
• Pharmacokinetics and Toxicokinetic Studies in the Safety Evaluation of
New Medicinal Products in Animals (ICH 3BS11A) Apr 1994
Toxicology
• Single dose
Q & A on the Withdrawal of the “Note for Guidance on Single
Dose Toxicity” EMA/CHMP/SWP/81714/2010 Jun 2010
• Repeated dose
Guideline on Repeated Dose Toxicity CHMP/SWP/1042/99
Rev. 1 Corr. Nov 2010
• Genotoxicity
Draft Guidance on Genotoxicity Testing and Data
Interpretation for Pharmaceuticals Intended for Human Use
(ICH S2 [R1]) – under discussion
• Carciogenicity
Possible ICH Topic in 2012 – Discussion ongoing
Carcinogenic Potential CPMP/SWP/2877/00 Jan 2003
ICH S1A 1996, S1B Mar 1998 and S1C(R2) Oct 20008

• Reproductive & Development Toxicity


Guideline on the Risk Assessment of Medicinal Products on
Human Reproduction and Lactation: From Data to Labelling
(EMEA/CHMP/203927/05) Jan 2009
Guideline on the Need for Non-Clinical Testing in Juvenile
Animals on Human Pharmaceuticals for Paediatric Indications
(CHMP/SWP/169215/05) Aug 2008
Detection of Toxicity to Reproduction for Medicinal Products
Including Toxicity to Male Fertility (ICH S5A(R2) Nov 2000
• Local Tolerance
Non-clinical local tolerance testing of medicinal products
CPMP/SWP/2145/00 Feb 2001 – under revision

• Other Toxicity
Reflection Paper on Non-clinical Evaluation of Drug Induced
Liver Injury (DILI) EMEA/CHMP/SWP/1501152/2006) Jun
2010
Questions & Answers on the Note for Guidance on Photosafety
Testing EMEA/CHMP/SWP/336670/2010 Mar 2011 – New
ICH Topic S10
Immunotoxicity Stuidies for Human Pharmaceuticals (ICH S8)
May 2006
Replacement of Animal Studies by In Vitro Models.
CPMP/SWP/728/95 Feb 1997 – Under revision to include 3Rs
development
• General Guidelines
Pre-clinical Safety Evaluation of Biotechnology-derived
Pharmaceuticals (ICH S6R1) – under revision to include
addendum – Finalized pending publication
Non-clinical Studies for Generic Nanoparticle Iron Medicinal
Product Applications EMA/CHMP/SWP/100094/2011 Apr
2011
Non-clinical Evaluation for Anticancer pharmaceuticals (ICH
S9) May 2010
Pre-clinical Pharmacological and Toxicological Testing of
Vaccines CPMP/SWP/465/95 Jun 1998
General FIM Guidances
 FDA (December 2002, Jul 2005) - Estimating the
Safe Maximum Starting Dose in Initial Clinical Trials for
Therapeutics in Adult Healthy Volunteers
 FDA (November 2000), M3 (R2) (June 2009) –
Non-clinical Safety Studies for the Conduct of
Human Clinical Trials and Marketing Authorization for
Pharmaceuticals (Sep 2007)
 CHMP Guidance on Requirements for FIM Clinical Trials
for Potential High Risk Medicinal Products
 Special guidelines: oncology, microdose, human
equivalent dose, minimum recommended dose
Resources
Guidances and Guidelines:
o ICH- http://www.fda.gov/cder/guidance/index.htm
 S1 Carcinogenicity
 S2 Genetic toxicity
 S3 Toxicokinetics
 S4 Duration of Chronic Toxicity Testing
 S5 Reproductive toxicity
 S6 Biotechnology
 S7 Safety Pharmacology
 M3 Nonclinical Safety Studies for the conduct of
Human Clinical Trials
83
8
4
Approach 1: Microdose (Single Dose Trials)
2 Approaches

Total dose of < 100 µg single dose or divided


to 5 doses

 Primary pharmacology
 In vitro target/receptor profiling
 Extended single dose toxicity study in 1 spp.
with TK by the intended route of administration
or via IV
 No genotoxicity but SAR assessment
Extended Single Dose Toxicity Study:

Rodent 10 animals/sex/group Day 2


5 animals/sex/group Day 14
Non-rodent 3 animals/sex/group Day 2

2 animals/sex/group Day 14

 Hematology
 Clinical chemistry
 Necropsy
 Histopathology
 Further evaluation 2 weeks later
Approach 2:
Microdose (Repeated Dose Trials)
Total accumulative dose < 500 µg of a maximum of 100 µg/
administration + washout (> 6 predicted half-life)

 Primary pharmacology
 In vitro target/receptor profiling
 + 7 day repeated-dose toxicity study in 1 species (rat) by
intended route with TK, haematology, clinical chemistry,
necropsy and histopathology
 No genotoxicity but SAR assessment
Approach 3: Single Dose Trials at Sub
Sub--
therapeutic Doses or into the Anticipated
Therapeutic Range

 In vitro target/receptor profiling


 Primary pharmacology
 Core batter of safety pharmacology
 Extended single dose toxicity in both rodent
&non-rodent
 Ames test (mutagenicity)
Approach 4: Multiple Dose Trials
Dosing up to 14 days into therapeutic range but not
intended to evaluate clinical MTD

 In vitro target/receptor profiling


 Primary pharmacology
 Core batter of safety pharmacology
 Standard 2-week repeated dose toxicity in rodent
and non-rodent
 Ames test and chromosomal damage in a mammalian
system (mutagenicity)
 If toxicity: apply MRSD
 If no toxicity (or in 1 species): starting dose = AUC ~ 1/50th of
the AUC at the NOAEL
Approach 5: Multiple Dose Trials
Dosing up to 14 days and not to exceed duration of
dosing in non-rodent, into the therapeutic range but not
intended to evaluate MTD

Same as Approach 4, but:


 Non-rodent (n=3) at the anticipated NOAEL exposure in rodent
> 3 days or escalating dose in non-rodent
 Ames test + Chromosomal damage in mammalian system
• Starting dose: 1/50th of the NOAEL in the most sensitive
species on mg/m-2 basis
• Maximum dose not higher than AUC at the NOAEL in non-
rodent or not higher than ½ the AUC at the NOAEL in
rodents whichever is lower

You might also like