Você está na página 1de 31

Forensic Chemistry (PHR 476)

Lecture of week 12

Regulatory Aspects of Safety in Process


of drug Discovery & Development
By:
Maged Wasfy
Lecturer of Pharmacology
Faculty of Pharmacy and Drug Manufacturing
Pharos University in Alexandria

Regulatory Aspects of Safety in Process of drug


Discovery & Development

Regulatory Aspects of Safety in Process of drug


Discovery & Development
Drug discovery and development is a long and complex process.
Only one of every 5,000-10,000 molecules screened in the
laboratory ever makes it to market as a new drug.
FDA & Research-based pharmaceutical companies take
extraordinary measures to ensure the safety and efficacy of all
approved prescription medicines.
To ensure patient safety, FDA and the pharmaceutical company
follow careful scientific procedures in four distinct stages:
1. Preclinical safety assessment

2. Pre-approval safety assessment in humans


3. Safety assessment during FDA regulatory review
4. Post-marketing safety surveillance.

Regulatory Aspects of Safety in Process of drug


Discovery & Development
1. Preclinical Safety Assessment
The relative safety of newly synthesized compounds is initially
evaluated in both in vitro and in vivo tests.
If a compound appears to have important biological &
pharmacological activity special tests are conducted to
evaluate its safety in the major organ systems (for example, the liver;
the central nervous, cardiovascular, and respiratory systems).
Other organ systems are evaluated when potential problems appear.
The goal of preclinical animal studies are:

A. To ensure that a drug is safe enough to be tested in humans.


B. Characterize any relationship between increased doses of a drug
and toxic effects in the animals.

Regulatory Aspects of Safety in Process of drug


Discovery & Development
2. Pre-Approval Safety Assessment in Humans (clinical trials)
A drug sponsor can begin clinical trials in humans once FDA is
satisfied that the preclinical animal data do not show an
unacceptable safety risk to humans.
New Drug Application (NDA) seeking FDA regulatory review to
market a new product.

Each clinical trial evaluates:


A. Safety & adverse effects regardless of the stated objective of
the trial.
B. Effect of new drug on the quality-of-life
C. Pharmacoeconomic of the new drug.
All harmful reactions are reported to FDA and, when appropriate, the
information is incorporated in a drugs package labeling.

Regulatory Aspects of Safety in Process of drug


Discovery & Development
2. Pre-Approval Safety Assessment in Humans (clinical trials)
The average NDA for a novel prescription medicine is based on
approximately 70 clinical trials involving over 4,000 patients.
Clinical trials are conducted in three stages:
A.Phase I:
Drugs are evaluated for safety in healthy volunteers using single
dose &multiple dose
B.Phase II:
To evaluate the safety and efficacy of a drug using patients instead of
healthy volunteers.
C.Phase III:
These large trials evaluate safety and efficacy in groups of patients
(with the disease to be treated) from different populations (elderly,
patients with multiple diseases, those who take other drugs, and
patients whose organs are impaired).

Regulatory Aspects of Safety in Process of drug


Discovery & Development
3. Safety Assessment During FDA Regulatory Review
A sponsor submits an NDA to FDA for approval to manufacture,
distribute, and market a drug based on the safety and efficacy data
obtained during the clinical trials.
FDA usually completes its review of a standard drug in 10 to 12
months. 120 days prior to a drugs anticipated approval, a sponsor
must provide the agency with a summary of all safety information in
the NDA, along with any additional safety information obtained
during the review period.
The addition of 600 new reviewers made possible by the user fees
paid by pharmaceutical companies has enabled FDA to approve new
drugs while maintaining high safety standards.

Regulatory Aspects of Safety in Process of drug


Discovery & Development
4. Post-Marketing Safety Surveillance
Monitoring and evaluating a drugs safety become more complex
after it is approved and marketed as drug will be taken by many
more patients and physicians are free to use it in different doses,
different dosing regimens and different patient populations.
Adverse reactions that occur in fewer than 1 in 3,000 to 5,000
patients are unlikely to be detected in pre-approval investigational
clinical trials, and may be unknown at the time a drug is approved.
These rare adverse reactions are more likely to be detected when
large numbers of patients use a drug after it has been approved.

If the drug shows serious adverse effects, FDA has its authority to
RECALL the drug from the market.

Preclinical Safety Assessment


(Toxicity Testing )

Preclinical Safety Assessment


(Toxicity Testing )
Toxicity testing is the structural unit of two branches of
toxicology:
A.Descriptive toxicology:
It is concerned with toxicity testing in experimental animals and
used to evaluate the risks posed to humans by exposure to specific
chemicals.
B.Regulatory toxicology:
It is concerned with deciding, on the basis of the data provided by
mechanistic and descriptive toxicologists, whether a drug or
another chemical poses a sufficiently low risk to be marketed for a
stated purpose.

Classification of Toxicity Studies


1. Acute Toxicity studies (Limit Tests, LD50, LC50, ED50, MTD,
Irritation)
2. Subchronic Toxicity studies
3. Chronic Toxicity studies
4. Special toxicity studies:
A. Tests for Developmental toxicity ( teratogenicity).
B. Tests for Reproductive Toxicity
C. Tests for Mutagenicity
D. Tests for Carcinogenicity

E. Tests for Neurotoxicity & Behavioral toxicity.


F. Tests for Immunotoxicity

1- Acute Toxicity Studies


Definition:
It is studying of the adverse effects occurring within a short
time of administration of a single dose or multiple doses
given within 24 hours.
Objectives of Acute toxicity studies are:
1.Identify the symptoms & possible mode of action of relative
toxicity of the compound.
2.To determine the existence of species differences.
3.Give information for further toxicity testing (i.e define more
precisely the doses to be used in subchronic toxicity tests)

1- Acute Toxicity Studies


Parameters obtained from Acute toxicity testing:

1. LD50 (Median lethal dose)


The calculated dose of a chemical that causes death in 50% of
the tested animals after acute, single exposure.
For inhaled chemicals, median lethal dose is expressed as LC50
(median lethal concentration) which is median concentration to
which animals are exposed for a fixed time that will kill 50% of the
animals.
2. NOAEL ( Non Observed Adverse Effect Level).
The highest dose in a toxicity study at which no toxic or adverse
effects are observed.

3. MTD (Maximum tolerated dose)


The highest dose of a chemical that when administered to a
group of test animals does not increase the death rate during a
long-term study.

1- Acute Toxicity Studies


Experimental Design of Acute toxicity testing:

1. Animal species:
At least 2 species are required
Rats and mice are usually used, but sometimes dogs and rabbits are
also included.

2. Animal number & gender:


At least (8-12) of each species, (4-6) of each sex.

3. Route of administration:
The chemical should be administered by the same route with which
human would be administered. (e,g: Oral gavage, Ocular, Dermal,
Intraperitoneal, lnhalation, intravenous, Intramuscular, Intranasal,
Subcutaneous)
Sometimes, we use more than two route of administration to assess
potential hazardous effects of chemical handling by human.
e.g: if the route of human exposure would be oral, acute dermal, eye,
and inhalation studies may be indicated to assess the hazard to
personnel's handling the compound.

1- Acute Toxicity Studies


Experimental Design of Acute toxicity testing:

4. Dosing :
single dose or multiple doses

5. Animal Monitoring:
The animals are observed for 14 days after chemical administration.
Observations include:

A. Recording of any serious symptomatic adverse effects.


B. Recording the number of animals dying during observation period, to
calculate LD50.
C. Any animals that haven't already died are then sacrificed and
dissected.
Postmortem examination
In general, all animals dying during the observation period and all surviving
animals should be autopsied by a qualified pathologist. The autopsy should
include gross and histopathological examination of all organs.

1- Acute Toxicity Studies


Examples of Acute toxicity testing

A. Inhalation test
It is used for assessment of the toxic hazard of agents whose
principal route of exposure is via inhalation and to Study of the
comparative toxicity of agents administered by different routes.
LC50 (median lethal concentration) is the parameter of choice of
this test. (where C = concentration of inhaled chemicals for a
fixed time T
Comparative LC50 data are often obtained for 4-hrs exposures
and a 14-day post-exposure observation period.
The most species used for inhalation studies include rats, dogs,
and monkeys.

1- Acute Toxicity Studies


Examples of Acute toxicity testing

B. Draize test
It is an acute toxicity tests that initially used for testing of topical
preparations.
It includes:
1. Ocular test
In this test, conscious rabbits are immobilized while chemicals are put
directly into their eyes. They are often left for as long as a week.
Injuries to the eyes after the exposure are determined by an irritancy
value (score). After the test, the rabbits are usually killed and internal
effects on the rabbits are examined.
2. Dermal irritation test
Skin corrosion and dermal irritation tests are typically conducted by
placing a chemical or chemical mixture on the skins of animals, usually
rabbits.

1- Acute Toxicity Studies


Limitation of Acute toxicity testing:
1. Parameters obtained from acute toxicity testing as LD50 is
subjected to interspecies difference (differences between toxic
effects of chemicals on human and other mammals)
e.g: LD50 of caffeine = 192 mg/kg in human & 620 mg/kg in mouse
The causes of interspecies difference include:

a. Differences in pharmacokinetics of the chemicals in different


species.
b. Existence of different physiology, organ or tissue types.
2. Can not predict the long-term organ toxicity.
Thus, several modifications have been done on Acute toxicity testing

1- Acute Toxicity Studies


Modified Acute toxicity testing:
1. In vitro cytotoxicity test
It is done using human cell culture taken from specific human tissue
that was expected to be affected, for example the skin, or the liver.
Advantages:
a. Obtained results are more reliable and more predictable than LD50
data as they avoid species differences.
Parameters used in In vitro cytotoxicty assays include:
1.Cell viability.
2.Cell proliferation.
3.Cellular protein content.
4.Cellular metabolism.

1- Acute Toxicity Studies


Modified Acute toxicity testing:

2. Repeated high-dose studies (Subacute studies)


The design of these studies based on repeated daily administration
of a series of doses to groups of animals for 5-21 days.
Advantages:
1. Obtained results are able to predict long-term toxicity, rather than
LD50 .
2. They define more precisely the doses to be used in subchronic
tests.

2- Subchronic Toxicity Studies


Definition:
It is studying of the adverse effects occurring as a result of daily or
frequent exposure of the compound over a period up to about 90
days (10% of animal life time).
Objectives of Subchronic toxicity studies are:
1. Identify the major toxic effects & target organ toxicity of the test
compound.
2. To determine the potential development of delayed toxicity.
3. To determine the potential reversibility of developed toxic effects.
4. Give information for further toxicity testing (i.e define more
precisely the doses to be used in chronic toxicity tests).

2- Subchronic Toxicity Studies


Experimental Design of subchronic toxicity testing:

1. Animal species:
At least 2 species are required, one rodent and one nonrodent.
Rats and dog are usually used (because of their availability &
complete physiological background of these species).
These tests should be done during the period of the most rapid
growth of the animal. (e.g: just after weaning of rats)

2. Animal number & gender:


At least 10 animals of each sex of each species.

3. Route of administration:
The chemical should be administered by the same route with
which human would be administered.

2- Subchronic Toxicity Studies


Experimental Design of subchronic toxicity testing:

4. Dosing : (4 dose levels)


Selection of doses for subchronic studies obtained from the results
of acute and repeated high-dose studies.
Doses include
1. The highest dose that provide a distinct toxic effect
2. The lowest dose that should not produce any detectable toxic
reaction.
3. At least two intermediate doses should be included.

2- Subchronic Toxicity Studies


Experimental Design of subchronic toxicity testing:

5. Animal Monitoring :
The animals are observed at 3-week intervals during exposure and
last up to 3 months following termination of exposure.
Parameters to be tested include:
A. Animal weights
B. Daily Food and water consumption
C. Biochemical organ function tests
D. Metabolic studies
E. Hematologic studies
F. Postmortem examination
These parameters are statistically compared to those obtained
from control groups of animal.
Control groups include Negative control and vehicle received
control.

2- Subchronic Toxicity Studies


Biochemical organ function tests
Using dog instead of a rodent species offer larger samples of
blood for biochemical organ function tests.
Organ function studies should be done prior to initiation of the
test, 3 and 10 days after the start of dosing, at 30-day intervals
throughout the test, and terminally.

Metabolic studies
The toxic nature of the metabolites and the rate of metabolic
transformation of the test compound are of essential parameters t
be evaluated in this toxicity studies.
Urine and faeces can be collected and examined for the presence
of metabolites.

2- Subchronic Toxicity Studies


Hematologic studies
Hematological studies should be started prior to initiation of the
test, at 30-day intervals, and on all animals terminally.
Bone marrow should be examined terminally.

Postmortem examination
All animals should be subjected to autopsy that include gross and
histopathological examination of all organs.

3- Chronic Toxicity Studies


Definition:
It is studying of the adverse effects occurring as a result of
exposing the animals to the chemical for the greater part of their
life span (i.e. exposure occurs from early life).
Objectives of Chronic toxicity studies are:
1. To determine precisely the major toxic effects of the test
compound and its mode of action.
2. To determine the potential carcinogenic action of the test
compound.

3- Chronic Toxicity Studies


Experimental Design of chronic toxicity testing:

1. Animal species:
A wide variety of animal species should be used in this type of work.
At least 2 animal species are required, these include:
A.Rodents are the animals of choice, since large numbers can be
used to aid in the statistical interpretation of the results.
B.Dogs and monkey (Larger animals) should also be used for such
species have the advantage that larger samples of blood can be
obtained on a routine basis.
N.B: for studying of the carcinogenic potential of a compound, the
rat, mouse, or hamster is usually chosen because of its:
Shorter lifetime.
Ability of using large numbers (to increase the sensitivity of the
test).

3- Chronic Toxicity Studies


Experimental Design of chronic toxicity testing:

2. Animal number & gender:


Large number should be used.

3. Route of administration:
The chemical should be administered by the same route with
which human would be administered.
N.B: For food additives, pesticides, and other chemicals likely to
come into contact with food or water, it is recommended to
incorporate the test chemical into the diet or drinking water.

3- Chronic Toxicity Studies


Experimental Design of chronic toxicity testing:

4. Dosing :
Selection of doses for chronic studies obtained from the results
of subchronic toxicity studies.
At least 2 dose levels required.

5. Animal Monitoring :
The same as subchronic toxicity studies.

Assignment
Assignment

For students with ID: from 108291 End of


students IDs including transfer students
(106. & 107..).

Discuss the following topic:


1. Example of the recently FDA recalled drugs
Presentation: 1 2 page word document, font type: arial,
font size 14, line spacing 1.5

Thank you

Você também pode gostar