You are on page 1of 7

17/01/2014

OBJECTIVES

After completing this module, you should be able:


• To understand the process of development of
drug
• To recognise problems related to the drug
development and its assessment

Erna Kristin

THE PROCESS OF DRUG Depart. Pharmacology


Faculty of Medicine

DEVELOPMENT GMU

Preclinical
Preclinical development evelopment

Clinical drug development Phase I-III

Regulatory approval-product license Regulatory


approval

Phase IV
Postmarketing surveillance Postmarketing
surrveillance

1
17/01/2014

Phases of Drug Development


Clinical Trials
Preclinical
Phase I Phase II Phase III FDA Phase IV
Testing

15
Years 6.5 1.5 2 3.5 1.5
total
20 to 80 100 to 300 1000 to
Laboratory
Test Fil healthy patient 3000
and animal
Population e volunte volunteer patient
studies File Review
IN ers s volunteers Additional
NDA process
D Evaluate Verify Post
at /
at Determi effectiven effectivene marketing
Assess FDA Approva
FD ne ess ss, monitor testing
safety and A l required
Purpose safety adverse
biological by FDA
and Look for reactions
activity
dosage side from long-
long-
effects term use

5,000 1
Success
compounds 5 enter trials approve
Rate
evaluated d

Total average cost for one new drug approval $359 million to $1.7 billion
Average time from test tube to market: 12-15 years

The changed context of drug Sources of drugs


discovery and development
The 1800s: The 1990s: insulin (pig, cow)
Animal growth hormone (man) (Creutzfeldt-Jakob)
 natural sources;  synthetic source;
 limited possibilities;  unlimited possibilities;
prepared by individuals; prepared by companies; digitalis (digitalis purpurea - foxglove)
  Plant morphine (papaver somniferum)
 small scale;  massive scale;
 not purified,  highly purified,
 standardised or tested;  standardised and tested; arsenic mercury
Inorganic lithium
 limited administration;  world-wide administration;
 no controls;  tight legislative control;
 no idea of mechanisms.  mechanisms partly chemical (propranolol)
understood. Synthetic biological (penicillin)
biotechnology (human insulin)

2
17/01/2014

orphan drug status (drugs intended to treat rare diseases),

accelerated approval (drugs to treat serious or life-threatening illness and


provide meaningful therapeutic benefit over existing treatments)

Subpart E designations (procedures for speeding the availability of drugs


to treat severely debilitating or life-threatening diseases).

Where do you find new drugs?

Preclinical
testing
Effects on body
functions,
mechanism of
action, toxicity

Pre Clinical Pre Clinical


• During preclinical drug development, a sponsor • At the preclinical stage, the FDA will generally ask, at
evaluates the drug's toxic and pharmacologic effects a minimum, that sponsors:
through in vitro and in vivo laboratory animal • (1) develop a pharmacological profile of the drug;
testing. • (2) determine the acute toxicity of the drug in at least
• Genotoxicity, mutagenecity, carcinogenecity, two species of animals, and
teratogenecity screening is performed, as well as • (3) conduct short-term toxicity studies ranging from 2
investigations on drug absorption and metabolism, weeks to 3 months, depending on the proposed
the toxicity of the drug's metabolites, and the speed duration of use of the substance in the proposed
with which the drug and its metabolites are excreted clinical studies
from the body.

3
17/01/2014

Preclinical development

PHARMACOLOGY

•Compounds must be shown to demonstrate specific


the fundamental tenet of testing drugs in pharmacological activity.
animals is that such information helps to •General pharmacological profiles of compounds are
predict the efficacy and toxicity of drugs also required (e.g. autonomic, cardiovascular, central
that are candidates for clinical use. nervous system

4
17/01/2014

ACUTE TOXICITY

1. Estimated lethal dose (LD50)


2. Usually evaluated in rodents

CHRONIC TOXICITY

• based on acute toxicity data,


• 3-4 dose levels are selected and
administered daily for certain periods
• to 2 species , one rodent (usually rat)
and one nonrodent (usually dog)

The doses selected are intended to include


• biochemical,
 a „no effect“ dose level • hematological and
 low dose • urinalysis profiles are performed
 mild dose • as well as physical examination,
 and of a top dose at which fatalities
• blood samples are taken for
will occur
 it is essential to include a placebo measurement of concentrations of the
dose group. drugs
• some animals are killed, and a full
histopathological examination is
performed on all organs.

Duration of chronic studies


What other testing will be provided
during preclinical development?
Doses to be given to human Duration of animal study

Up to 3 doses 14 days

repeat dosing up to animal toxicity testing


10 days for 28 days mutagenicity carcinogenicity reproductive testing
longer administration 6 or 12 months

5
17/01/2014

Clinical drug development Clinical drug development

Phase I Phase I
• initial exposure of humans to investigational • to determine the metabolic and pharmacological
drug (first in humans) actions of the drug in humans, the side effects
• safety evaluation associated with increasing doses, and, if possible,
to gain early evidence on effectiveness.
• assessment of pharmacokinetics and
• to evaluate drug metabolism, structure-activity
pharmacodynamics in healthy subjects. relationships, and the mechanism of action in
• These individuals volunteer to be given the humans.
drug under strictly controlled, hospitalized, • The total number of subjects 20-80.
expert medical supervision

Clinical drug development Clinical drug development

Phase II Phase II
• initial safety evidence • Phase 2 includes the early controlled clinical studies
• initial efficacy evidence conducted to obtain some preliminary data on the
• identification of the dose range for phase III effectiveness of the drug for a particular indication
or indications in patients with the disease or
trial
condition.
• well controlled with narrowly defined patients • This phase of testing also helps determine the
population common short-term side effects and risks associated
• open trial with the drug. Phase 2 studies usually involve
• 100-150 patients in total several hundred people

Clinical drug development Clinical drug development

Phase III Phase III


• efficacy and safety objective • Phase 3 studies are intended to gather the
• expanded and controlled additional information about effectiveness and
• collection of data on a large patient safety that is needed to evaluate the overall benefit-
risk relationship of the drug.
population with indication
• Phase 3 studies also provide an adequate basis for
• provide information on overall benefit/risk extrapolating the results to the general population
• can be placebo or active controls and transmitting that information in the physician
• large multicenter labeling.
• 1000-5000 patients in total • Phase 3 studies usually include several hundred to
• usually double blind several thousand people

6
17/01/2014

Regulatory approval-product license Regulatory approval-product license


• Formal application to the Licensing Authority
is made. • All prescribing doctores are provided
• The size of submission documents may
with a factual data sheet giving
extend to several hundredweights of paper.
• Marketing approval may be general or information on each new medicine.
granted subject to certain limitations which • It must provide all the information
may include restriction to hospital practice necessary to make a proper decision as
only, restriction in indications for use or a to whether the drug is indicated (i.e. it is
requirement to monitor in a specified number not an advertizement) and must also
of patients. detail all known hazards of the drug.

Post marketing surveilance

Phase IV

• studies are prospective trials performed after


marketing approval (the granting of a Product
License).

They may be studies to investigate:


• new formulations
• dosage requirements
• drug interactions
• detection of previously unrecognized unwanted
events

You might also like