Lecture 14: Drug Development and Biosimilars Flashcards
What Constitutes a “Drug”?
A drug is a substance that produces change in cellular or physiologic functioning
Drug development influenced by:
High cost of research; complex regulatory environment & legislations;
insurer reimbursements; and patient perspectives
Some of factors that influence drug development include
Development cost (~ $1.3 - $2.8 billion or more) and time (can be as much as or >15 years) Regulatory approval process Changes in laws Patient acceptance Insurance reimbursement
Investing in new drug development is
expensive and risky
FDA Regulates drugs under two legislative acts
- Food, Drug, and Cosmetic Act – for small molecule drugs
- Public Health Service Act – for biologicals
The FDA’s mission is to promote and protect the public health by promoting:
safe and effective products reach the market in a timely safe and effective products reach the market in a timely way
- However: once FDA determines a drug is safe and effective, it has little control over
its use in medical practice, such as for non-approved indications (“off-label” use)
Specific FDA Function/Role in Drug Development
Process and Drugs in the Marketplace
- Approves drugs on the basis of purity, safety, efficacy
- Regulate all labeling of prescription and nonprescription drugs
- Advertising OTCs the responsibility of the Federal Trade Commission (FTC)
- Regulate manufacturing processes of drugs and institution of drug recalls
- Regulate bio-equivalence for generics and bio-similars
- Monitor safety of US blood supply
Conceptualization
performed by independent investigators, small research companies, academic institutions & large pharmaceutical companies
Intramural
NIH investigators perform research
Extramural
independent investigators win research grants to perform research
Drug development has
3 stages prior to approval
STAGE 1
Drug discovery
Discovery ( Laboratory based)
5,000 – 10,000 compounds may be synthesized and tested
- 250 “may” make it to pre-clinical testing
STAGE 2
Pre-clinical Testing
- Tests performed to determine if candidate molecule safe to test in humans
- Candidates for drugs are first tested in animals
- Very few candidate materials for drugs make it through pre-clinical testing
- < 0.1% of potential candidate drugs go from discovery to clinical testing stage
- Varies in number of years it takes
STAGE 3
Clinical testing has 3 DISTINCT PHASES with distinct goals
- Investigational New Drug (IND) application must be submitted to FDA
in order to begin clinical testing in humans
STAGE 4
Post approval and post clinical testing (also called Phase IV)
To begin clinical testing,
Investigational New Drug (IND) Application must be filed
When all clincial testing is complete,
The New Drug Application (NDA) must be filed
PHASE IV
addition research post-marketing by FDA
- Used to identify events not detected during clinical testing
IND
Investigational New Drug
NDA
New Drug Application
3 phases before drug approval : Clinical testing
- Phase 1: Learn how the drug works in (small number) of humans – set dosage
- Phase II: Demonstrate efficacy (proof of concept) and safety (toxicity) in patients
- Phase III: Provide strong evidence of efficacy and safety by studying candidate drug
in a few thousand people with target disease in randomized, controlled trials using
doses and settings more like “real world” practice
Phase I Clinical Testing Goals Include
- Learn how candidate drug works in HEALTHY humans
Studies of <100 healthy individuals
Establish dose range, metabolism, excretion, toxicities
Usually takes one year or less to complete
Phase II Clinical Testing Goals Include
- Study candidate drug in a few hundred people with target disease
- Efficacy (proof of concept) and safety (toxicity) in patients
- Single arm studies (No comparison group)
- Usually takes up to two years to complete
(occurs after Phase I testing is completed)
Phase III Clinical Testing Goals Includ
Study a few thousand people with target disease in randomized, controlled trials
Strong evidence for efficacy and safety
Use doses and settings more like real-world practice
Success is necessary to move to the next stage of approval
Usually takes three to four years to complete
(occurs after Phase I and Phase II testing is completed
Efficacy
Extent to which a specific intervention, procedure, regimen, or service
produces a beneficial result under ideal conditions
“Ideal conditions” are achieved in clinical trials
Effectiveness
- Measure of success of a therapeutic intervention carried out in a typical
environment on typical patients - “real-world” conditions
Post-Clinical testing
NDA submitted to FDA upon successful completion of clinical testing (Stage 3)
NDA specifies the following information about the candidate drug:
Manufacturing
Action in the body
Marketing
Labeling and toxicologic information
Phase IV testing and evaluation
Post-marketing surveillance in larger samples and for longer time frame
evaluation of safety and effectiveness not detected in earlier trials
“real-world” settings, “real-world” patients, and very large numbers of people
MedWatch
- FDA’s safety information adverse event reporting program
Health professionals encouraged to report adverse events in their patients
Manufacturers are required to report adverse events to FDA
Adverse Event Reporting System
Supports the FDA’s post-marketing safety surveillance program
Contains adverse drug event reports FDA received from manufacturers required by regulation
ANDA
Abbreviated New Drug Application
Abbreviated New Drug Application (ANDA). What is ANDA
- submitted to FDA for a drug already “approved” but required when requesting:
Change from prescription to over-the-counter status
Change in ingredients, dosage, manufacturing, formulation, or labeling
Who submits to ANDA
Manufacturers of generics or biosimilars
- FDA examines the bio-equivalence, bioavailability, and
pharmacokinetic/pharmacodynamic properties
Intellectual Property (IP) rights
protect innovators & provide
incentives for drug development
But: allow Pharma Companies to have a monopoly & charge higher prices
Drug Price Competition & Patent Term Restoration Act of 1984
(Hatch Waxman Act) & PPACA
Hatch-Waxman Act Characteristics
- Applies to small molecule drugs
Innovator data exclusivity for 5 years from FDA approval*
Additional 3 years of data exclusivity for new indications
Additional 6 months of exclusivity for pediatric applications
No new clinical trials required for generics (ANDA)
Regulation pertains to small-molecule drugs
Hatch-Waxman Act
established the approval pathway for generic drug products, under which applicants can submit an abbreviated new drug application (ANDA)
Biologic
are a large, new part of the Rx marketplace
- Very important uses in major diseases
Very, VERY expensive medications
Majority of Biologicals Use Living Cells to
Produce a Protein Product
Insert gene encoding the protein of interest
Cells require proper conditions for optimal growth
(temp, pH, oxygen, feeds, etc.)
Culture and fermentation can take weeks
Complex Purification Steps
Safe product with desired potency
Small molecule drugs vs. Biological drugs
Biological drugs are large and have a high MW. They are complex. Produced in living cell structure. Unstable and immunogenic
Small molecule drugs are non-immunogenic, stable, produced by chemical synthesis
BLA
Biologic License Application- which includes
- Manufacturing info to demonstrate company can properly and consistently manufacture the product
The biological product is licensed and can be marketed in the US only if:
FDA determines that the data shows the biological product is safe and effective for it’s proposed use, and
-the biological product can be manufactured in a way that ensures a quality product
Biologics Price Competition and Innovation Act (2009)
Abbreviated approval pathway in place under the 2010 Patient Protection and
Affordable Care Act (PPACA)
Allows for licensing with less than full complement of pre-clinical and clinical data
Focus on the manufacturing process
“highly similar” with no clinically significant differences in safety, purity, potency,
immunogenicity
Tested against a reference product, NOT against placebo!
“The goal is not to independently establish safety & effectiveness of proposed product
Biosimilarity”
A biosimilar product is a biological product that is approved based on a showing that it is
highly similar to an FDA-approved biological product, known as a reference product, and
has no clinically meaningful differences in terms of safety and effectiveness [purity,
potency] from the reference product
Only minor differences in clinically inactive components are allowable in biosimilar
products”
Interchangeable means
An interchangeable biological product may be substituted for the reference product
by a pharmacist without the intervention of the health care provider who prescribed
the reference product
-They have different brand names,
making their identification more confusing