You are on page 1of 41

Prekliniske dokumentasjonsstudier

og forventet kvalitet
Katrine W Rustad
LINK Medical Research

Norsk Biotekforum, 13.februar 2012
Outline
Preclinical safety studies
Regulatory guidelines
Good Laboratory Practice (GLP)
Purpose of GLP
OECD principles of GLP

Why, how and when are preclinical safety studies
performed?

Preclinical
research and
development
Clinical research
and development
Short-term
Long-term
Phase 1
Phase 2
Phase 3
MAA/NDA
review
Post-marketing
surveillance
Phase 4
Animal
testing
Adverse
reaction
reporting
File submission Approval
Initial
synthesis
Guidelines
FDA (US Food and Drug Administration)

EMA (The European Agency for the Evaluation of
Medicinal Products)

OECD (Organization for Economic Co-operation and
Development)

ICH (International Conferences of Harmonisation)



Guidelines
cont.
ICH topics divided into 4 major categories:


S=Safety topics, relating to in vitro and in vivo preclinical
studies
Carcinogenicity Studies
Genotoxicity Studies
Toxicokinetics and Pharmacokinetics
Toxicity Testing
Reproductive Toxicology
Biotechnological Products
Pharmacology Studies
Immunotoxicology Studies

Q=Quality topics, relating to chemical and pharmaceutical Quality
Assurance

E=Efficacy topics, relating to clinical studies in human

Guidelines
cont.
ICH topics divided into 4 major categories:

M= Multidisciplinary Topics i.e. cross-cutting topics

M1: Medical terminology
M2: Electronic standards for transmission of regulatory
information

M3: Timing of Pre-clinical Studies in Relation to Clinical
Trial
o M3(R2):
Guidance on Non-Clinical Safety Studies for the Conduct of
Human Clinical Trials and Marketing Authorization for
Pharmaceuticals

M4: The Common Technical Document (CTD)
M5: Data elements and standard for drug dictionaries


Good Laboratory Practice (GLP)
FDA reported that a number of preclinical laboratory
studies which had been performed to investigate the
toxicity of novel compounds were deficient in a variety
of ways

The problem was sufficiently serious to warrant the
introduction of legislation which should prevent future
deficiencies

The first proposals for GLP were made by FDA in 1976
and finally published as law in 1979
Purpose of GLP:
Quality
generate scientific data that comply with regulatory
authorities requirements with respect to quality

Reliability of safety data
data submitted to regulatory authorities are a true
reflection of the results obtained during the study

Human safety
data can be relied upon when making risk/safety
assessments

GLP is not:
GLP can become confused with the standards of
laboratory safety
wearing appropriate gloves, glasses and clothing to
handle materials safely

GLP is not laboratory work in general!


Good Laboratory Practice (GLP)
Planned
Performed
Monitored
Recorded
Reported
Archived

(ref. OECD GLP Principles)

GLP is a quality system concerned with the organizational process
and the conditions under which nonclinical health and
environmental safety studies are:

OECD Principles of GLP include:
Ensure test facility fulfill GLP requirements:
suitable labs
essential equipment and material available
adequate, competent study personnel
Must have knowledge in those parts of GLP applicable to
their involvement in study
Record raw data promptly and accurately in accordance
with GLP
Responsible for quality of raw data


GLP principles:


Quality assurance program:
Assure QA responsibility performed in accordance with
GLP:
Verify and document study plan performed in
accordance with GLP
Conduct inspections to determine if study conducted in
accordance with GLP
Inspect final report to confirm that methods,
procedures, observation are accurate and completely
described
Reported results accurately and completely reflect raw
data of study
Sign a GLP statement in final study report incl. type and
date of inspections were reported to management and
study director


GLP principles:

Standard operating procedure (SOP):
Appropriate and technically validated SOPs should be
available for, but not limited to the following categories:
Test and reference items
Apparatus, material, reagents and solutions
Computerised systems
Record keeping, reporting, storage and retrieval
Test system
QA procedures

GLP principles:

Facilities for Test and Reference Items:
Should be separate rooms/areas for receipt and storage of
test item and reference items
To prevent contamination or mix-ups
Mixing of the test item with vehicle

Test and Reference Items
Receipt, handling, sampling and storage
Labelling, expiry date, storage conditions, stability
Characterisation:
Identity (batch number), purity, composition,
concentration


GLP principles:

Performance of the study:
Appoint Study Director (SD)
Documented approval of study plan issued by SD

Study Director Responsibilities:
single point of study control
responsible for overall conduct of the study
issue protocol, QA involvement, ensure computerised
systems are validated, documentation of raw data,
final report and archiving

Principle Investigator Responsibilities
Responsible for delegated phase of the GLP study in
multi-site studies


GLP principles:

Test System Facilities:
Suitable rooms/areas available

Biological:
Newly arrived animals should be isolated until health
status evaluated
Maintain record of source, date and conditions at arrival
Should be acclimatised to the environment for an adequate
period before first adminstration of test item
All info needed to properly identify test system should
appear on their housing during the conduct of the study
During use, housing for test systems should be cleaned
and sanitised at appropriate intervals
Bedding for animals should be changed as required by
sound husbandry practice
GLP principles:

Storage/archiving of records and reports:
secure storage and retrieval of study plan, raw data,
final reports, sample of test item and specimens


GLP inspections:
All labs having GLP accreditation will be inspected
GLP inspection include a general review of the quality system
in the lab and an audit of selected GLP studies
Report will be sent to the lab and to the authority
Common findings/deviations:
Report not in accordance with raw data
Unauthorized corrections in raw data
Implementation of protocol deviations without amendment
Procedures not done in accordance with SOP/no SOP
established
Incomplete traceability of sample (receipt, storage, time,
temp)
Archiving routine
Why, How and When are
preclinical toxicology and
pharmacology studies performed?

Why perform toxicology studies?
To be as sure as possible (within reasonable limits)
that the products we develop are not harmful to man
at clinically relevant doses

To fulfill the regulatory requirements for registration of
new products

To identify possible risk parameters that need to be
followed-up in clinical studies



How to perform toxicology studies?
Single dose toxicity (rat and dog)
Repeat dose toxicity (rat and dog)
Local tolerance (rabbit)
Genetic toxicity, in vitro + in vivo (rat)
Reproductive toxicology
Carcinogenicity

All studies are conducted according to GLP

Single dose toxicity studies


Investigate the toxic effect of a single high dose of the
product
Single dose via intended clinical route
Minimum 3 doses + control
2 species: rodent + non-rodent
14 Days observation (clinical signs)
Body weight
Necropsy
Target organ weight
Target organ histology

Repeat dose toxicity studies


The drug may not produce immediate toxic effects
Delayed effects due to accumulation of the drug in
animals
2 species: rodent + non-rodent
Toxicokinetics required to document exposure and aid
to interpretation
Recovery period to investigate reversibility of findings

Repeat dose toxicity studies
cont.

Clinical signs
Food consumption
Water consumption
Ophthalmology
Body weight
Antibody determination

Clinical pathology
-clinical chemistry
-haematology
-urinalysis

Anatomic pathology
-organ weight
-macroscopic
-microscopic

Observations, analysis and measurements:
Irritation & Local tolerance studies




Local tolerance
-Intravenous
-Intraarterial
-Paravenous
-Subcutaneous
-Intramuscular
Irritation
-Skin
-Eye

Ascertain that medicinal products are tolerated at sites
in the body which may come in contact with the product
as a result of its administration in clinical use

Genotoxicity

In vivo and in vitro tests for detection of compounds
that induce genetic damage directly or indirectly

Compounds positive in these tests may have the
potential of inducing cancer and/or heritable effects

A battery of tests is necessary to assess the genotoxic
potential of a compound

Genotoxicity
cont.

The recommended standard test battery:

A test for gene mutation in bacteria (Ames test)

An in vitro test with cytogenetic evaluation of
chromosomal damage with mammalian cells or an
in vitro mouse lymphoma tk assay (MLA test)

An in vivo test for chromosomal damage using
rodent hematopoietic cells (micronucleus test)

Reproductive toxicology
Study for effects on fertility and early embryonic
development to implantation

Study for effects on pre- and postnatal
development, including maternal function

Carcinogenicity
The need for carcinogenicity studies:
- Expected clinical use is continuous > 6 months

The rodent bioassays are amongst the most costly
elements in preclinical testing, in terms of time and
resources
Chronic studies (usually 2 years)
Over 1000 animals used per drug


Pharmacology studies
Pharmacology studies can be divided into three
categories
Primary pharmacodynamic:
studies on the mode of action and/or effects of a
substance in relation to its desired therapeutic
target

Secondary pharmacodynamic:
studies on the mode of action and/or effects of a
substance not related to its desired therapeutic
target

Safety pharmacology studies:
Performed according to GLP
Safety Pharmacology Core Battery
Purpose to investigate the effects of the test substance on vital
organs or systems (acutely critical for life):

Cardiovascular system:
Blood pressure, Heart rate, electrocardiogram (ECG)
Telemetry study (dog)

Respiratory system:
Respiratory rate and other measures of respiratory
function (rat)

Central nervous system:
Motor activity, Behavioral changes, Coordination,
Sensory/motor reflex responses, Body temperature
Modified Irwin screen test (rat)
QT prolongation
Delayed ventricular repolarization
Drug induced cardiac arrhythmia recognized as a major hurdle
in development of new drugs

Most common problem is:
acquired long QT syndrome caused by drugs that block the
myocardial Herg potassium channel
delay cardiac repolarization
increase the risk of torsades de pointes arhythmia

Testing of compounds for interactions with the hERG channel
allows the identification of potential risk of QT prolongation in
humans
hERG assay, in vitro electrophysiology study
whole-cell patch-clamp technique



Safety margins
Need a margin of safety for each drug product

Safety margin:
Multiples of the clinical dose dose to be used in
human studies that have been tested in preclinical
safety studies

No observed effect level (NOEL):
The highest dose tested in an animal species with
no detected effects

FDA recommendation:
the no-observed-adverse-effect level (NOAEL) in
toxicity studies in suitable animal species to be at
least one hundred times (100x) greater than the
maximal mass dose to be used in human studies


Clinical trials
Phase I - Human Pharmacology Studies
- to evaluate PK and tolerance in healthy volunteers


Phase II - Therapeutic Exploratory Studies
-to explore efficacy and safety in patients


Phase III - Therapeutic Confirmatory Studies

- to confirm efficacy and safety in patients


Phase IV Therapeutic use

-to study the safety after drug has been released on the market
When to perform preclinical studies?
Timing of studies in relation to
clinical trials
ICH M3:
timing of preclinical studies in relation to clinical trials

recommended preclinical safety studies needed to
support clinical trials

duration of preclinical safety studies





When to perform preclinical studies?
Timing of studies in relation
to clinical trials
Before Phase I:
Single dose toxicity
Repeat dose toxicity
Local tolerance
Genotoxicity
in vitro mutagenicity & clastogenicity
Safety pharmacology
PK & TK
appropriate information by the time Phase I is
completed
When to perform preclinical studies?
Timing of studies in relation
to clinical trials
Before Phase II:
Repeat dose toxicity (dependent on duration)
Genotoxicity
complete standard battery

Before Phase III:
Repeat dose toxicity (dependent on duration)

Before New Drug Application (NDA):
Carcinogenicity studies (unless cause for concern)
not required for all pharmaceuticals
may be conducted post-approval for
pharmaceuticals treating serious diseases

Summary and conclusion:
Preclinical toxicological studies:
Single dose toxicity (rat and dog)
Repeat dose toxicity (rat and dog)
Local tolerance (rabbit)
Genotoxicity (in vitro and in vivo)

Safety pharmacology core battery:
Combined respiratory and cardiovascular study in
conscious (telemetered) dogs
Irwin screen test (rat)
hERG assay (in vitro)


Summary and conclusion:
Preclinical safety studies have to be performed prior
to clinical trials in order to fulfil regulatory
requirements

All safety studies are conducted according to GLP

You might also like