SlideShare uma empresa Scribd logo
1 de 55
New Drug Development
DR G A WAGHMARE
21/8/11 1
21/8/11 2
Contents
1. Introduction
2. Definition of NEW DRUG.
3. Regulatory Guidelines.
4. Steps in the drug development
5. Timelines of Patent
21/8/11 3
1.Introduction
• Development of new drug is very arduous, time
consuming & very expensive process.
• During last 50 years, hundreds of new drugs have
been introduced, & many older drugs have been
deleted (withdrawn).
• < 1% of compounds that go into test eventually
become licensed medicines.
21/8/11 4
• To bring a new drug to market requires a good
understanding of drug development process &
integral role preclinical testing plays in that process.
• 5000– 10,000 compounds yield 1 new drug to
market.
• Overall time required for successful results is 10-
12 years.
21/8/11 5
A Pyramid of Uncertainty
• Time:- It takes 12 years on average for an
experimental drug to travel from lab to medicine
chest.
• Success:- Only five in 5,000 compounds that
enter preclinical testing make it to human
testing.
• One of these five tested in humans is approved.
5
21/8/11 6
Steps Number of compounds
• Discovery 5,000-10,000
• Preclinical testing 250
• Phase I 5
• Phase II 1
• Phase III 1
• FDA review & approval 1
Attrition rate of overall drug development
&
Average cost of drug development
Chemical
Labs
Decision
Filter
10,000
compounds
Pharmacology
Toxicology
Decision
Filter
1000
compounds
Clinical
Trials
Decision
Filter
10
compoundsNDA submission
Decision
Filter
1
compound
NDA Approval
> $ 250million
> $ 250
million
IND submission
21/8/11 7
21/8/11 8
2.New drug
• New drug: definition under Schedule Y
– New substance, which except during local clinical
trials, has not been used ever before in the
country.
– Drug already approved for certain claims but which
is now proposed to be marketed with modified or
new claims (indications, dosage, dosage form,
route of administration).
– FDC of two or more drugs, proposed to be
combined for first time in a fixed ratio.
21/8/11 9
3.Regulatory Guidelines.
• Authority to grant permission for New Drug to test &
market new drug in India rests with “Drugs Controller
General of India (DCGI)”, and is given as per Drugs &
Cosmetic rules.
• Schedule Y of Drugs & Cosmetic Rules, regarding
investigation of new drug (NDA) gives details of
preclinical (animal) data required as also issues
guidelines on clinical trials which are required to be
carried out for import & /or manufacture of new drug
in India.
21/8/11 10
Regulatory Authorities.
US – FDA
Europe – EMEA
UK – MHRA
Japan – MHLW
21/8/11 11
4.Steps in new drug development
A. Idea or Basic Research
B. New drug discovery
C. Screening
D. Preclinical studies
E. Formulation development
F. IND application
G. Clinical studies
H. Official license / Regulations/Marketing
21/8/11 12
A. Basic research
1. Start by studying normal & abnormal body
functions.
2. Investigation of each component of the
disease (pathophysiology).
3. Look up information obtained in previous
research and publication.
4. Find out at which stage we can stop
disease progression or development (OUR
TARGET!).
5. Search for targeted drug.
6. Isolate the index compound.
7. Perform animal testing to obtain safety
data.
8. Approval to test in humans.
21/8/11 13
Investigate Disease
Investigate each
component of a disease:-
• What is/are the
symptom(s) ?
• What is the cause?
• Which is the target
organ?
• What is/are Biochemical
pathway(s)?
13
21/8/11 14
B. New Drug discovery
1. Target Identification
2. Target validation
3. Lead Identification
4. Lead Optimization
21/8/11 15
Target Identification and Validation :
– Often begins with target identification - choosing a biochemical
mechanism involved in a disease condition.
– Drugs usually act on either cellular or genetic chemicals within
our body, known as targets believed to be associated with the
disease.
– Drug candidates are tested for their interaction with drug target.
– Up to 5000-10000 molecules for each potential drug candidate
are subject to rigorous screening process.
– Once scientists confirm interaction with drug target, they
typically validate that target by checking activity against the
disease condition for which the drug is being developed.
21/8/11 16
Lead identification :
– Lead compound or substance is one that is believed to
have potential to treat disease. Laboratory scientists
compare known substances with new compounds to
determine their likelihood of success.
– Leads are sometimes developed as collections, or
libraries, of individual molecules that possess properties
needed in new drug. Testing is then done on each of
these molecules to confirm its effect on drug target.
21/8/11 17
Lead optimization:
– Compares properties of various lead compounds and
provides information to help pharmaceutical and
biotechnology companies select compound/s with
greatest potential to be developed into safe and effective
medicines.
– Often during this same stage of development, lead
prioritization studies are conducted in living organisms
(in vivo) and in cells in a test tube (in vitro) to compare
various lead compounds, their metabolism, etc.
21/8/11 18
Characteristics of Ideal Drug
Candidate• High Potency
• High Selectivity
• Good oral
Bioavailability
• Low or no
interaction with
CYP450
• Less or minimal
adverse effects
• Good therapeutic
index
21/8/11 19
C. Screening
– NCEs are subject to battery of screening tests
designed to determine different types of biological
activity.
– Such tests include studies on animal behavior,
isolated tissues, intact animals, animal models of the
disease.
– 1 in every 4000-5000 NCEs screened is marketed.
21/8/11 20
Pharmacological screening of candidate
molecules
• Pharmacological observations are made,
depending on expected pharmacological
properties. e.g.
– fall in BP, fall in blood glucose, etc.
21/8/11 21
Pharmacological screening of
candidate molecules
• At the cellular level, it is possible to understand the
mechanism of action of a drug, whether is acts as:
– Receptor agonist / antagonist; if so, its
affinity and selectivity.
– Inhibitor of key enzyme, etc.
Through this process one can rapidly identify active
compounds, antibodies or genes which modulate
a particular biomolecular pathway.
The results of these experiments provide starting
points for drug design and for understanding the
interaction or role of a particular biochemical
process in biology.
21/8/11 22
21/8/11 23
Pharmacological screening of candidate molecules is
done in isolated organ system and in whole animal
experiment (in vivo), effects of potential drug candidate
on different systems (CVS, CNS, RS etc.) can be studied.
D. Pre-clinical Studies
21/8/11 24
• Pharmacologic studies :
– Carried out in experimental animals using in vivo or in vitro
techniques.
– These studies bring out specific biological activities,
mechanism of action, PK, effective dose range of the test
compounds.
• Toxicity studies :
– Detailed toxicity studies are conducted on compounds
tested positive pharmacologically.
21/8/11 25
Preclinical safety and toxicity testing
• Evaluated for toxicity-potential risk.
Goals:
• Identifying safe drug
• Identifying potential human toxicity
• Predicting specific and relevant toxicities to be
monitored in clinical trials
E. Formulation development
• DRUG +
Additive: filler, lubricant, coating, stabiliser, colour,
binder, disintegrator
Dosage form: capsule, tablet, injection, other?
Manipulate duration/profile: e.g. sustained release
Bioequivalence
Bioavailability
Ease of use
2621/8/11
21/8/11 27
F. IND application
• IND application is a result of successful preclinical
development program.
• IND is a vehicle through which the sponsor advances to the
next stage of drug development - clinical trials.
• IND not an application for marketing approval.
• Contents :
– Animal pharmacological & toxicology studies.
– Manufacturing information.
– Clinical protocols and investigator information.
Sponsor/FDA Meetings ( Pre-IND)
• Prior to clinical studies, the sponsor needs
evidence that the compound is biologically
active, and both sponsor and the FDA needs
data showing that the drug is reasonably safe
for initial administration to humans.
• Meeting at such an early stage in the process
are useful opportunities for open discussion
about testing phases, data, requirements, and
any scientific issues that may need to be
resolved prior to IND submission
21/8/11 28
21/8/11 2929
IND
Review
Process
21/8/11 30
G. Clinical studies
A new drug, which is expected to have some
advantage over existing ones, after pre-clinical
testing in vitro, short-term and long-term animal
testing for safety, efficacy and toxicity, after due
regulatory and ethics committee permissions, is
subjected to studies in humans as per the
regulatory requirements.
21/8/11 31
• Clinical pharmacology (human studies)
– Phase I
– Phase II
– Phase III
– Phase IV
21/8/11 32
Phase I trials
– Human / Clinical Pharmacology Trials
– This is a open study conducted in healthy human
volunteers (20-80).
– In special populations (e.g. for anti-cancer drugs).
21/8/11 33
Objectives
• Safety & tolerability
Maximum tolerated dose
• Pharmacokinetics
• Pharmacodynamics
• Measurement of drug activity
21/8/11 34
Players In Phase I Trials
Participants (20-80)
• Healthy volunteer subjects
• Patients
Place
• Special testing facilities
• Monitored closely
Physician
• Trained investigator
21/8/11 35
Phase II trials
• Therapeutic exploratory trials
• First trial in patients with the disease to be
treated.
• 50-300 patients are used for this study.
21/8/11 36
Objectives
• Effectiveness of the drug
• Common short term side effects and risks
with I.N.D.
• Determine doses and regimens for phase III
trials
• PK, PD, safety
Additional objectives
• Therapeutic regimens
• Target populations for further studies in phase III
21/8/11 37
Players In Phase II Trials
Participants (50-300)
• Patients
Place
• Specialized hospital units
• Closely monitored
Physician
• Trained investigators
Sponsor/FDA Meeting (End of Phase
2)
• One month prior to the “end of the Phase 2”, the sponsor
should submit the background information and protocols
for phase 3 studies.
• This information should include data supporting the claim
of the new drug product, chemistry data, animal data and
proposed additional animal data, results of Phase 1 and 2
studies, statistical methods being used, specific protocols
for phase 3 studies, as well as a copy of the proposed
labeling for a drug, if available.
• This summary provides the review team with information
needed to prepare for a productive meeting.
21/8/11 38
21/8/11 39
Phase III trials
• They are initiated when the data generated
shows evidence of efficacy.
• Patients (300-3,000)
• Phase III includes multiple sites , hence most
expensive and most time consuming
• These studies should be intended to provide
an adequate basis for marketing approval.
21/8/11 40
Objectives of Phase –III Trials
• Efficacy and safety profile on a larger number of
population (1000+)
• Comparison with current Gold Standard
treatment
• Identification of the disease sub types for which
drug is effective
• To provide data for the product package insert.
21/8/11 41
Players in Phase III Trials
• Patients-250-1000+
• SITE
Multi-speciality hospital with adequate
patient attendance and laboratory facilities
• Principal investigator assisted by a clinical
research co-ordinator .
21/8/11 42
Clinical Trial Design
• PROSPECTIVE RANDOMISED MULTICENTRIC
• CONTROLLED OR UNCONTROLLED TRIALS
IIIa Trials carried out on a large number of
patients – Regulatory requirement for NDA
IIIb Extended trials of IIIa after applying for
approval but before launch
21/8/11 43
H .Official license /Marketing
NDA /MAA
• New drug application (NDA) is a vehicle through which
sponsors formally propose that FDA approve it - new
pharmaceutical - for sale in United States.
• These are examples of applications to market a new drug.
• Such applications document safety and efficacy of the
investigational drug and contain all the information collected
during the DDP.
• Obtaining approval to market a new drug frequently takes
between six months and two years.
21/8/11 4444
NDA Review Process
21/8/11 45
Phase IV
• This constitutes a vigilant post-marketing
surveillance to monitor safety of new drug.
• PMS is a systemic method for surveillance of
adverse reactions, patterns of drug utilization,
additional indication discovered, and is carried
out in patients given marketed drug for therapy.
21/8/11 46
Objectives
• Conform the efficacy and safety profile in large
populations during practice
• Detect the unknown adverse drug reaction/s
• Evaluation of over-dosage and concomitant treatments
• Identifications of new indications
• Pharmacoeconomics
21/8/11 47
Conduct of Phase IV trial
• Post authorization studies, post marketing
studies, post licensing studies.
• Site Clinics and hospitals
• Players
Principal investigators-General practitioners and
Specialists
Participants patients-2000-10000+
21/8/11 48
From Laboratory to Clinics
A Phased Journey
Preclinical IND SUBMISSION
Phase I
Phase II a,b
Phase III b
III a
Phase IV, PMS
Observational
studies
N
D
A
S
U
B
M
I
S
S
I
P
R
O
D
U
C
T
L
A
U
N
C
H
21/8/11 49
5. Timelines of Patent
• Lifetime of patent-20 yrs
• Pharma company has exclusive rights for
limited time
• After expiration of patent- any company may
produce and market the drug as a generic
product
21/8/11 50
50
The New Drug Development Process:
Steps from Test Tube to New Drug Application Review
21/8/11 51
• Drug discovery:
Ideas Drug candidates & biochemical tools
• Drug development:
Drug candidates Drugs
Discovery=find new active structure
Development=convert it to a useful drug
52
Advances in drug development.
21/8/11
53
Phase 0 microdosing
A new approach to obtain human pharmacokinetic information
before the usual expensive phase I safety program is conducted is
the phase 0 microdosing.
It is hypothesized that microdosing will help to reduce or replace the
extensive animal testing of compounds for kinetics, which may
later be rejected in human studies.
Thus, microdose studies use minute quantities of drug and are not
intended to produce any pharmacologic effect, when administered
to humans, and, therefore, may not cause any adverse events also,
but may produce useful pharmacokinetic information and help in
further development of the compound.21/8/11
54
• 1/100th of dose anticipated to produce a pharmacological
effect
• A small dose of your own medicine…..
• Provides sufficiently useful PK information to decide on
confirmatory development (human & animal toxicology)
• Helps in early de-selection: Cost saving related to
manufacturing, scaling up & CTs
21/8/11
55
Microdosi
ng
21/8/11

Mais conteúdo relacionado

Mais procurados

Investigational New drug application [INDA]
Investigational New drug application [INDA]Investigational New drug application [INDA]
Investigational New drug application [INDA]Sagar Savale
 
Stages of drug discovery
Stages of drug discoveryStages of drug discovery
Stages of drug discoveryPawanDhamala1
 
Investigator's Brochure
Investigator's BrochureInvestigator's Brochure
Investigator's BrochureRiyaz Gohil
 
Regulatory affairs in Pharmaceutical Industry
Regulatory affairs in Pharmaceutical IndustryRegulatory affairs in Pharmaceutical Industry
Regulatory affairs in Pharmaceutical IndustryRama Shukla
 
Drug regulatory authority
Drug regulatory authority Drug regulatory authority
Drug regulatory authority Rahul Gawande
 
Drug discovery and development
Drug discovery and developmentDrug discovery and development
Drug discovery and developmentrahul_pharma
 
Abbreviated New Drug Application [ANDA]
Abbreviated New Drug Application [ANDA]Abbreviated New Drug Application [ANDA]
Abbreviated New Drug Application [ANDA]Sagar Savale
 
Preclinical studies
Preclinical studiesPreclinical studies
Preclinical studiesAzeemsales
 
Cdsco Roles and Responsibilities
Cdsco Roles and ResponsibilitiesCdsco Roles and Responsibilities
Cdsco Roles and ResponsibilitiesSandip Mavchi
 
Preclinical studies, clinical trails and pharmacovigilance
Preclinical studies, clinical trails and pharmacovigilancePreclinical studies, clinical trails and pharmacovigilance
Preclinical studies, clinical trails and pharmacovigilancekamrudeen samani
 
INVESTIGATOR’S BROCHURE (IB)
 INVESTIGATOR’S BROCHURE (IB) INVESTIGATOR’S BROCHURE (IB)
INVESTIGATOR’S BROCHURE (IB)SachinFartade
 
REGULATORY AFFAIRS ( IP-2 / UNIT 3 )
REGULATORY AFFAIRS ( IP-2 / UNIT 3 )REGULATORY AFFAIRS ( IP-2 / UNIT 3 )
REGULATORY AFFAIRS ( IP-2 / UNIT 3 )JAYACHANDRA AKUTHOTA
 
Bioequivalence studies
Bioequivalence studiesBioequivalence studies
Bioequivalence studiesSujit Patel
 

Mais procurados (20)

Investigational New drug application [INDA]
Investigational New drug application [INDA]Investigational New drug application [INDA]
Investigational New drug application [INDA]
 
Stages of drug discovery
Stages of drug discoveryStages of drug discovery
Stages of drug discovery
 
Schedule y
Schedule ySchedule y
Schedule y
 
Investigator's Brochure
Investigator's BrochureInvestigator's Brochure
Investigator's Brochure
 
Regulatory affairs in Pharmaceutical Industry
Regulatory affairs in Pharmaceutical IndustryRegulatory affairs in Pharmaceutical Industry
Regulatory affairs in Pharmaceutical Industry
 
Cdsco ppt
Cdsco pptCdsco ppt
Cdsco ppt
 
Schedule y
Schedule ySchedule y
Schedule y
 
Drug regulatory authority
Drug regulatory authority Drug regulatory authority
Drug regulatory authority
 
Drug discovery and development
Drug discovery and developmentDrug discovery and development
Drug discovery and development
 
ICH AND ICH GUIDELINES
ICH AND ICH GUIDELINESICH AND ICH GUIDELINES
ICH AND ICH GUIDELINES
 
Abbreviated New Drug Application [ANDA]
Abbreviated New Drug Application [ANDA]Abbreviated New Drug Application [ANDA]
Abbreviated New Drug Application [ANDA]
 
Preclinical studies
Preclinical studiesPreclinical studies
Preclinical studies
 
Ich guidelines
Ich guidelinesIch guidelines
Ich guidelines
 
Regulatory affairs
Regulatory affairsRegulatory affairs
Regulatory affairs
 
Cdsco Roles and Responsibilities
Cdsco Roles and ResponsibilitiesCdsco Roles and Responsibilities
Cdsco Roles and Responsibilities
 
Preclinical studies, clinical trails and pharmacovigilance
Preclinical studies, clinical trails and pharmacovigilancePreclinical studies, clinical trails and pharmacovigilance
Preclinical studies, clinical trails and pharmacovigilance
 
INVESTIGATOR’S BROCHURE (IB)
 INVESTIGATOR’S BROCHURE (IB) INVESTIGATOR’S BROCHURE (IB)
INVESTIGATOR’S BROCHURE (IB)
 
REGULATORY AFFAIRS ( IP-2 / UNIT 3 )
REGULATORY AFFAIRS ( IP-2 / UNIT 3 )REGULATORY AFFAIRS ( IP-2 / UNIT 3 )
REGULATORY AFFAIRS ( IP-2 / UNIT 3 )
 
final year project
final year projectfinal year project
final year project
 
Bioequivalence studies
Bioequivalence studiesBioequivalence studies
Bioequivalence studies
 

Destaque

Key Considerations in Stability Studies for Biopharmaceuticals
Key Considerations in Stability Studies for BiopharmaceuticalsKey Considerations in Stability Studies for Biopharmaceuticals
Key Considerations in Stability Studies for BiopharmaceuticalsSGS
 
Regulations for drug approval in USA, E.U & India
Regulations for drug approval in USA, E.U & IndiaRegulations for drug approval in USA, E.U & India
Regulations for drug approval in USA, E.U & IndiaDr. Pankaj Bablani
 
Drug development and clinical trial phases
Drug development and clinical trial phasesDrug development and clinical trial phases
Drug development and clinical trial phasesSunil Boreddy Rx
 
US and EU Submission – Comparative
US and EU Submission – ComparativeUS and EU Submission – Comparative
US and EU Submission – ComparativeGirish Swami
 
As process development and manufacturing (cmc) for biologics development-an o...
As process development and manufacturing (cmc) for biologics development-an o...As process development and manufacturing (cmc) for biologics development-an o...
As process development and manufacturing (cmc) for biologics development-an o...Steven S. Lee
 

Destaque (7)

Key Considerations in Stability Studies for Biopharmaceuticals
Key Considerations in Stability Studies for BiopharmaceuticalsKey Considerations in Stability Studies for Biopharmaceuticals
Key Considerations in Stability Studies for Biopharmaceuticals
 
New Drug Development Process
New Drug Development ProcessNew Drug Development Process
New Drug Development Process
 
Regulations for drug approval in USA, E.U & India
Regulations for drug approval in USA, E.U & IndiaRegulations for drug approval in USA, E.U & India
Regulations for drug approval in USA, E.U & India
 
Clinical Trial Phases
Clinical Trial PhasesClinical Trial Phases
Clinical Trial Phases
 
Drug development and clinical trial phases
Drug development and clinical trial phasesDrug development and clinical trial phases
Drug development and clinical trial phases
 
US and EU Submission – Comparative
US and EU Submission – ComparativeUS and EU Submission – Comparative
US and EU Submission – Comparative
 
As process development and manufacturing (cmc) for biologics development-an o...
As process development and manufacturing (cmc) for biologics development-an o...As process development and manufacturing (cmc) for biologics development-an o...
As process development and manufacturing (cmc) for biologics development-an o...
 

Semelhante a New drug development

Introduction to drug discovery and development.pptx
Introduction to drug discovery and development.pptxIntroduction to drug discovery and development.pptx
Introduction to drug discovery and development.pptxMingmaLhamuBhutia
 
Applications of bio-pharmaceutics in new drug delivery
Applications of bio-pharmaceutics in new drug  deliveryApplications of bio-pharmaceutics in new drug  delivery
Applications of bio-pharmaceutics in new drug deliveryAkshata shettar
 
Drug discovery & clinical evaluation of new drugs
Drug discovery & clinical evaluation of new drugsDrug discovery & clinical evaluation of new drugs
Drug discovery & clinical evaluation of new drugsHeena Parveen
 
safety data generation.pptx
safety data generation.pptxsafety data generation.pptx
safety data generation.pptxGayatriBahatkar1
 
Drug development process
Drug development process Drug development process
Drug development process Zobayer Hossain
 
Lecture # 03 New Drug Approval and Development Process,
Lecture # 03 New Drug Approval and Development Process,Lecture # 03 New Drug Approval and Development Process,
Lecture # 03 New Drug Approval and Development Process,PakistanPharmaCareerDoor
 
Clinical pharmacology and drug development
Clinical pharmacology and drug developmentClinical pharmacology and drug development
Clinical pharmacology and drug developmentJavedAkhtar170
 
regulatory requirements for drug approval ( IP-2 / UNIT -3 )
regulatory requirements for drug approval ( IP-2  / UNIT -3 )regulatory requirements for drug approval ( IP-2  / UNIT -3 )
regulatory requirements for drug approval ( IP-2 / UNIT -3 )JAYACHANDRA AKUTHOTA
 
Drug Development Process
Drug Development ProcessDrug Development Process
Drug Development ProcessTusharJ7
 
Discovery of Drug and Introduction to Clinical Trial__Katalyst HLS
Discovery of Drug and Introduction to Clinical Trial__Katalyst HLSDiscovery of Drug and Introduction to Clinical Trial__Katalyst HLS
Discovery of Drug and Introduction to Clinical Trial__Katalyst HLSKatalyst HLS
 
Drug Development Process
Drug Development ProcessDrug Development Process
Drug Development ProcessINDERGOHRI
 
Stages of drug development by Dr Joseph Oyepata Simeon (Ph.D)
Stages of drug development by Dr Joseph Oyepata Simeon (Ph.D)Stages of drug development by Dr Joseph Oyepata Simeon (Ph.D)
Stages of drug development by Dr Joseph Oyepata Simeon (Ph.D)oyepata
 
PreClinical Development_Final_clinical.pdf
PreClinical Development_Final_clinical.pdfPreClinical Development_Final_clinical.pdf
PreClinical Development_Final_clinical.pdfsayedjannatfatema72
 
Drug discovery & development
Drug discovery & developmentDrug discovery & development
Drug discovery & developmentShubham Patil
 
Clinical Research, A Basic Understanding...........
Clinical Research, A Basic Understanding...........Clinical Research, A Basic Understanding...........
Clinical Research, A Basic Understanding...........Jobin Kunjumon Vilapurathuu
 
Regulatory requirements for drug approval
Regulatory requirements for drug approvalRegulatory requirements for drug approval
Regulatory requirements for drug approvalAudumbar Mali
 

Semelhante a New drug development (20)

Introduction to drug discovery and development.pptx
Introduction to drug discovery and development.pptxIntroduction to drug discovery and development.pptx
Introduction to drug discovery and development.pptx
 
Applications of bio-pharmaceutics in new drug delivery
Applications of bio-pharmaceutics in new drug  deliveryApplications of bio-pharmaceutics in new drug  delivery
Applications of bio-pharmaceutics in new drug delivery
 
Drug discovery & clinical evaluation of new drugs
Drug discovery & clinical evaluation of new drugsDrug discovery & clinical evaluation of new drugs
Drug discovery & clinical evaluation of new drugs
 
safety data generation.pptx
safety data generation.pptxsafety data generation.pptx
safety data generation.pptx
 
CLINICAL TRIALS.pptx
CLINICAL TRIALS.pptxCLINICAL TRIALS.pptx
CLINICAL TRIALS.pptx
 
Drug development process
Drug development process Drug development process
Drug development process
 
Lecture # 03 New Drug Approval and Development Process,
Lecture # 03 New Drug Approval and Development Process,Lecture # 03 New Drug Approval and Development Process,
Lecture # 03 New Drug Approval and Development Process,
 
Clinical pharmacology and drug development
Clinical pharmacology and drug developmentClinical pharmacology and drug development
Clinical pharmacology and drug development
 
regulatory requirements for drug approval ( IP-2 / UNIT -3 )
regulatory requirements for drug approval ( IP-2  / UNIT -3 )regulatory requirements for drug approval ( IP-2  / UNIT -3 )
regulatory requirements for drug approval ( IP-2 / UNIT -3 )
 
Drug Development Process
Drug Development ProcessDrug Development Process
Drug Development Process
 
Discovery of Drug and Introduction to Clinical Trial__Katalyst HLS
Discovery of Drug and Introduction to Clinical Trial__Katalyst HLSDiscovery of Drug and Introduction to Clinical Trial__Katalyst HLS
Discovery of Drug and Introduction to Clinical Trial__Katalyst HLS
 
Drug Development Process
Drug Development ProcessDrug Development Process
Drug Development Process
 
Stages of drug development by Dr Joseph Oyepata Simeon (Ph.D)
Stages of drug development by Dr Joseph Oyepata Simeon (Ph.D)Stages of drug development by Dr Joseph Oyepata Simeon (Ph.D)
Stages of drug development by Dr Joseph Oyepata Simeon (Ph.D)
 
PreClinical Development_Final_clinical.pdf
PreClinical Development_Final_clinical.pdfPreClinical Development_Final_clinical.pdf
PreClinical Development_Final_clinical.pdf
 
Drug discovery & development
Drug discovery & developmentDrug discovery & development
Drug discovery & development
 
NEW DRUG
NEW DRUGNEW DRUG
NEW DRUG
 
drug discovery.pptx
drug discovery.pptxdrug discovery.pptx
drug discovery.pptx
 
Clinical Research, A Basic Understanding...........
Clinical Research, A Basic Understanding...........Clinical Research, A Basic Understanding...........
Clinical Research, A Basic Understanding...........
 
Regulatory requirements for drug approval
Regulatory requirements for drug approvalRegulatory requirements for drug approval
Regulatory requirements for drug approval
 
Drug development process
Drug development processDrug development process
Drug development process
 

Mais de Dr Gangaprasad Waghmare (16)

Nasal polyp
Nasal polypNasal polyp
Nasal polyp
 
Diseases of external ear
Diseases of external earDiseases of external ear
Diseases of external ear
 
Anatomy of pharynx
Anatomy of pharynxAnatomy of pharynx
Anatomy of pharynx
 
GJO.MS.ID.555594
GJO.MS.ID.555594GJO.MS.ID.555594
GJO.MS.ID.555594
 
ANATOMY OF NOSE
ANATOMY OF NOSEANATOMY OF NOSE
ANATOMY OF NOSE
 
6890
68906890
6890
 
14-ORAL CAVITY
14-ORAL CAVITY14-ORAL CAVITY
14-ORAL CAVITY
 
Chronic Suppurative Otitis Media
Chronic Suppurative Otitis MediaChronic Suppurative Otitis Media
Chronic Suppurative Otitis Media
 
MYRINGOTOMY,
MYRINGOTOMY,MYRINGOTOMY,
MYRINGOTOMY,
 
OTITIS MEDIA
OTITIS MEDIAOTITIS MEDIA
OTITIS MEDIA
 
tongue
tonguetongue
tongue
 
RHINITIS
RHINITISRHINITIS
RHINITIS
 
Anatomy of tooth
Anatomy of toothAnatomy of tooth
Anatomy of tooth
 
Management_of_Epistaxis_Goal_is_Control
Management_of_Epistaxis_Goal_is_ControlManagement_of_Epistaxis_Goal_is_Control
Management_of_Epistaxis_Goal_is_Control
 
Epistaxis
EpistaxisEpistaxis
Epistaxis
 
Clinical Research
Clinical ResearchClinical Research
Clinical Research
 

New drug development

  • 1. New Drug Development DR G A WAGHMARE 21/8/11 1
  • 2. 21/8/11 2 Contents 1. Introduction 2. Definition of NEW DRUG. 3. Regulatory Guidelines. 4. Steps in the drug development 5. Timelines of Patent
  • 3. 21/8/11 3 1.Introduction • Development of new drug is very arduous, time consuming & very expensive process. • During last 50 years, hundreds of new drugs have been introduced, & many older drugs have been deleted (withdrawn). • < 1% of compounds that go into test eventually become licensed medicines.
  • 4. 21/8/11 4 • To bring a new drug to market requires a good understanding of drug development process & integral role preclinical testing plays in that process. • 5000– 10,000 compounds yield 1 new drug to market. • Overall time required for successful results is 10- 12 years.
  • 5. 21/8/11 5 A Pyramid of Uncertainty • Time:- It takes 12 years on average for an experimental drug to travel from lab to medicine chest. • Success:- Only five in 5,000 compounds that enter preclinical testing make it to human testing. • One of these five tested in humans is approved. 5
  • 6. 21/8/11 6 Steps Number of compounds • Discovery 5,000-10,000 • Preclinical testing 250 • Phase I 5 • Phase II 1 • Phase III 1 • FDA review & approval 1
  • 7. Attrition rate of overall drug development & Average cost of drug development Chemical Labs Decision Filter 10,000 compounds Pharmacology Toxicology Decision Filter 1000 compounds Clinical Trials Decision Filter 10 compoundsNDA submission Decision Filter 1 compound NDA Approval > $ 250million > $ 250 million IND submission 21/8/11 7
  • 8. 21/8/11 8 2.New drug • New drug: definition under Schedule Y – New substance, which except during local clinical trials, has not been used ever before in the country. – Drug already approved for certain claims but which is now proposed to be marketed with modified or new claims (indications, dosage, dosage form, route of administration). – FDC of two or more drugs, proposed to be combined for first time in a fixed ratio.
  • 9. 21/8/11 9 3.Regulatory Guidelines. • Authority to grant permission for New Drug to test & market new drug in India rests with “Drugs Controller General of India (DCGI)”, and is given as per Drugs & Cosmetic rules. • Schedule Y of Drugs & Cosmetic Rules, regarding investigation of new drug (NDA) gives details of preclinical (animal) data required as also issues guidelines on clinical trials which are required to be carried out for import & /or manufacture of new drug in India.
  • 10. 21/8/11 10 Regulatory Authorities. US – FDA Europe – EMEA UK – MHRA Japan – MHLW
  • 11. 21/8/11 11 4.Steps in new drug development A. Idea or Basic Research B. New drug discovery C. Screening D. Preclinical studies E. Formulation development F. IND application G. Clinical studies H. Official license / Regulations/Marketing
  • 12. 21/8/11 12 A. Basic research 1. Start by studying normal & abnormal body functions. 2. Investigation of each component of the disease (pathophysiology). 3. Look up information obtained in previous research and publication. 4. Find out at which stage we can stop disease progression or development (OUR TARGET!). 5. Search for targeted drug. 6. Isolate the index compound. 7. Perform animal testing to obtain safety data. 8. Approval to test in humans.
  • 13. 21/8/11 13 Investigate Disease Investigate each component of a disease:- • What is/are the symptom(s) ? • What is the cause? • Which is the target organ? • What is/are Biochemical pathway(s)? 13
  • 14. 21/8/11 14 B. New Drug discovery 1. Target Identification 2. Target validation 3. Lead Identification 4. Lead Optimization
  • 15. 21/8/11 15 Target Identification and Validation : – Often begins with target identification - choosing a biochemical mechanism involved in a disease condition. – Drugs usually act on either cellular or genetic chemicals within our body, known as targets believed to be associated with the disease. – Drug candidates are tested for their interaction with drug target. – Up to 5000-10000 molecules for each potential drug candidate are subject to rigorous screening process. – Once scientists confirm interaction with drug target, they typically validate that target by checking activity against the disease condition for which the drug is being developed.
  • 16. 21/8/11 16 Lead identification : – Lead compound or substance is one that is believed to have potential to treat disease. Laboratory scientists compare known substances with new compounds to determine their likelihood of success. – Leads are sometimes developed as collections, or libraries, of individual molecules that possess properties needed in new drug. Testing is then done on each of these molecules to confirm its effect on drug target.
  • 17. 21/8/11 17 Lead optimization: – Compares properties of various lead compounds and provides information to help pharmaceutical and biotechnology companies select compound/s with greatest potential to be developed into safe and effective medicines. – Often during this same stage of development, lead prioritization studies are conducted in living organisms (in vivo) and in cells in a test tube (in vitro) to compare various lead compounds, their metabolism, etc.
  • 18. 21/8/11 18 Characteristics of Ideal Drug Candidate• High Potency • High Selectivity • Good oral Bioavailability • Low or no interaction with CYP450 • Less or minimal adverse effects • Good therapeutic index
  • 19. 21/8/11 19 C. Screening – NCEs are subject to battery of screening tests designed to determine different types of biological activity. – Such tests include studies on animal behavior, isolated tissues, intact animals, animal models of the disease. – 1 in every 4000-5000 NCEs screened is marketed.
  • 20. 21/8/11 20 Pharmacological screening of candidate molecules • Pharmacological observations are made, depending on expected pharmacological properties. e.g. – fall in BP, fall in blood glucose, etc.
  • 21. 21/8/11 21 Pharmacological screening of candidate molecules • At the cellular level, it is possible to understand the mechanism of action of a drug, whether is acts as: – Receptor agonist / antagonist; if so, its affinity and selectivity. – Inhibitor of key enzyme, etc.
  • 22. Through this process one can rapidly identify active compounds, antibodies or genes which modulate a particular biomolecular pathway. The results of these experiments provide starting points for drug design and for understanding the interaction or role of a particular biochemical process in biology. 21/8/11 22
  • 23. 21/8/11 23 Pharmacological screening of candidate molecules is done in isolated organ system and in whole animal experiment (in vivo), effects of potential drug candidate on different systems (CVS, CNS, RS etc.) can be studied. D. Pre-clinical Studies
  • 24. 21/8/11 24 • Pharmacologic studies : – Carried out in experimental animals using in vivo or in vitro techniques. – These studies bring out specific biological activities, mechanism of action, PK, effective dose range of the test compounds. • Toxicity studies : – Detailed toxicity studies are conducted on compounds tested positive pharmacologically.
  • 25. 21/8/11 25 Preclinical safety and toxicity testing • Evaluated for toxicity-potential risk. Goals: • Identifying safe drug • Identifying potential human toxicity • Predicting specific and relevant toxicities to be monitored in clinical trials
  • 26. E. Formulation development • DRUG + Additive: filler, lubricant, coating, stabiliser, colour, binder, disintegrator Dosage form: capsule, tablet, injection, other? Manipulate duration/profile: e.g. sustained release Bioequivalence Bioavailability Ease of use 2621/8/11
  • 27. 21/8/11 27 F. IND application • IND application is a result of successful preclinical development program. • IND is a vehicle through which the sponsor advances to the next stage of drug development - clinical trials. • IND not an application for marketing approval. • Contents : – Animal pharmacological & toxicology studies. – Manufacturing information. – Clinical protocols and investigator information.
  • 28. Sponsor/FDA Meetings ( Pre-IND) • Prior to clinical studies, the sponsor needs evidence that the compound is biologically active, and both sponsor and the FDA needs data showing that the drug is reasonably safe for initial administration to humans. • Meeting at such an early stage in the process are useful opportunities for open discussion about testing phases, data, requirements, and any scientific issues that may need to be resolved prior to IND submission 21/8/11 28
  • 30. 21/8/11 30 G. Clinical studies A new drug, which is expected to have some advantage over existing ones, after pre-clinical testing in vitro, short-term and long-term animal testing for safety, efficacy and toxicity, after due regulatory and ethics committee permissions, is subjected to studies in humans as per the regulatory requirements.
  • 31. 21/8/11 31 • Clinical pharmacology (human studies) – Phase I – Phase II – Phase III – Phase IV
  • 32. 21/8/11 32 Phase I trials – Human / Clinical Pharmacology Trials – This is a open study conducted in healthy human volunteers (20-80). – In special populations (e.g. for anti-cancer drugs).
  • 33. 21/8/11 33 Objectives • Safety & tolerability Maximum tolerated dose • Pharmacokinetics • Pharmacodynamics • Measurement of drug activity
  • 34. 21/8/11 34 Players In Phase I Trials Participants (20-80) • Healthy volunteer subjects • Patients Place • Special testing facilities • Monitored closely Physician • Trained investigator
  • 35. 21/8/11 35 Phase II trials • Therapeutic exploratory trials • First trial in patients with the disease to be treated. • 50-300 patients are used for this study.
  • 36. 21/8/11 36 Objectives • Effectiveness of the drug • Common short term side effects and risks with I.N.D. • Determine doses and regimens for phase III trials • PK, PD, safety Additional objectives • Therapeutic regimens • Target populations for further studies in phase III
  • 37. 21/8/11 37 Players In Phase II Trials Participants (50-300) • Patients Place • Specialized hospital units • Closely monitored Physician • Trained investigators
  • 38. Sponsor/FDA Meeting (End of Phase 2) • One month prior to the “end of the Phase 2”, the sponsor should submit the background information and protocols for phase 3 studies. • This information should include data supporting the claim of the new drug product, chemistry data, animal data and proposed additional animal data, results of Phase 1 and 2 studies, statistical methods being used, specific protocols for phase 3 studies, as well as a copy of the proposed labeling for a drug, if available. • This summary provides the review team with information needed to prepare for a productive meeting. 21/8/11 38
  • 39. 21/8/11 39 Phase III trials • They are initiated when the data generated shows evidence of efficacy. • Patients (300-3,000) • Phase III includes multiple sites , hence most expensive and most time consuming • These studies should be intended to provide an adequate basis for marketing approval.
  • 40. 21/8/11 40 Objectives of Phase –III Trials • Efficacy and safety profile on a larger number of population (1000+) • Comparison with current Gold Standard treatment • Identification of the disease sub types for which drug is effective • To provide data for the product package insert.
  • 41. 21/8/11 41 Players in Phase III Trials • Patients-250-1000+ • SITE Multi-speciality hospital with adequate patient attendance and laboratory facilities • Principal investigator assisted by a clinical research co-ordinator .
  • 42. 21/8/11 42 Clinical Trial Design • PROSPECTIVE RANDOMISED MULTICENTRIC • CONTROLLED OR UNCONTROLLED TRIALS IIIa Trials carried out on a large number of patients – Regulatory requirement for NDA IIIb Extended trials of IIIa after applying for approval but before launch
  • 43. 21/8/11 43 H .Official license /Marketing NDA /MAA • New drug application (NDA) is a vehicle through which sponsors formally propose that FDA approve it - new pharmaceutical - for sale in United States. • These are examples of applications to market a new drug. • Such applications document safety and efficacy of the investigational drug and contain all the information collected during the DDP. • Obtaining approval to market a new drug frequently takes between six months and two years.
  • 45. 21/8/11 45 Phase IV • This constitutes a vigilant post-marketing surveillance to monitor safety of new drug. • PMS is a systemic method for surveillance of adverse reactions, patterns of drug utilization, additional indication discovered, and is carried out in patients given marketed drug for therapy.
  • 46. 21/8/11 46 Objectives • Conform the efficacy and safety profile in large populations during practice • Detect the unknown adverse drug reaction/s • Evaluation of over-dosage and concomitant treatments • Identifications of new indications • Pharmacoeconomics
  • 47. 21/8/11 47 Conduct of Phase IV trial • Post authorization studies, post marketing studies, post licensing studies. • Site Clinics and hospitals • Players Principal investigators-General practitioners and Specialists Participants patients-2000-10000+
  • 48. 21/8/11 48 From Laboratory to Clinics A Phased Journey Preclinical IND SUBMISSION Phase I Phase II a,b Phase III b III a Phase IV, PMS Observational studies N D A S U B M I S S I P R O D U C T L A U N C H
  • 49. 21/8/11 49 5. Timelines of Patent • Lifetime of patent-20 yrs • Pharma company has exclusive rights for limited time • After expiration of patent- any company may produce and market the drug as a generic product
  • 50. 21/8/11 50 50 The New Drug Development Process: Steps from Test Tube to New Drug Application Review
  • 51. 21/8/11 51 • Drug discovery: Ideas Drug candidates & biochemical tools • Drug development: Drug candidates Drugs Discovery=find new active structure Development=convert it to a useful drug
  • 52. 52 Advances in drug development. 21/8/11
  • 53. 53 Phase 0 microdosing A new approach to obtain human pharmacokinetic information before the usual expensive phase I safety program is conducted is the phase 0 microdosing. It is hypothesized that microdosing will help to reduce or replace the extensive animal testing of compounds for kinetics, which may later be rejected in human studies. Thus, microdose studies use minute quantities of drug and are not intended to produce any pharmacologic effect, when administered to humans, and, therefore, may not cause any adverse events also, but may produce useful pharmacokinetic information and help in further development of the compound.21/8/11
  • 54. 54 • 1/100th of dose anticipated to produce a pharmacological effect • A small dose of your own medicine….. • Provides sufficiently useful PK information to decide on confirmatory development (human & animal toxicology) • Helps in early de-selection: Cost saving related to manufacturing, scaling up & CTs 21/8/11