Sie sind auf Seite 1von 40

DRUG DEVELOPMENT

A view on the process from the idea to the


registered pharmaceutical

Dr. Matthias Kreuter


Head of Alpinia Laudanum Institute of Phytopharmaceutical Sciences
AG Walenstadt, Switzerland
Organisation of the presentation

I. DISCOVERY

Identification of target and resource


Organisation of the presentation

II. HIT GENERATION

Perspectives: A) Research and Development


B) Quality Control and Production
C) Marketing Authorisation
Organisation of the presentation

III. LEAD GENERATION

Perspectives: A) Research and Development


B) Quality Control and Production
C) Marketing Authorisation
Organisation of the presentation

IV. CLINICAL DEVELOPMENT

Perspectives: A) Research and Development


B) Quality Control and Production
C) Marketing Authorisation
Organisation of the presentation

V. POST REGISTRATION

Perspectives: A) Research and Development


B) Quality Control and Production
C) Marketing Authorisation
I. DISCOVERY

Identification of target and resource


I. DISCOVERY

Target identification

- Area of interest in terms of drug indication ?

- Relevant cellular or molecular targets ?

- Appropriate assays – established or to be developed ?

- Available relevant literature ?

- Patent situation in the target area ?


I. DISCOVERY

Resource identification

Potential resources for novel drugs:

- Natural organisms (plants, fungi, bacteria, animals)


- Combinatorial chemistry
- Structure-based drug design

Methods for drug discovery:


- High throughput screening of random samples (HTS):
Including screen development, primary and secondary
screening

- Ethnobiological approach:
Traditional use of natural organisms for medicines
I. DISCOVERY

Resource identification - Alpinia Institute

Natural organisms, in particular plants

Medicinal plants continue to play a significant role


as a resource for the discovery of novel drugs (1)

1) Balunas and Koinghorn, Life Sci


2005.
I. DISCOVERY

Resource identification - Alpinia Institute

Natural organisms, in particular plants

Medicinal plants continue to play a significant role


as a resource for the discovery of novel drugs (1)

- 52% of the drugs approved in the U.S. from 1981-2002


were natural products or derived from them (2).
- 26 plant based drugs were approved during 2000-2006,
including novel-molecular based drugs (3).
- In the future multicomponent botanical therapeutics will
experience an increasing interest in biomedicine (4).

2) Newman, J Nat Pr 2002. 3) Saklani & Kutty, Drug Disc Today 2008. 4) Schmidt et al., Nature Chem
Biol 2007.
I. DISCOVERY

Method of drug discovery - Alpinia Institute

Ethnobotanical approach

Systematic screening of:


- Published literature on traditional medicinal plant use
(e.g. documented traditional healers‘ experience)
- Historical texts
(e.g. ancient botanico-medicinal manuscripts)

Advantages: - Preselection of potentially active resources


- Promising safety profile (age-long
experience)
- Cost-efficient and comparatively fast
II. HIT GENERATION

Perspectives: A) Research and Development


B) Quality Control and Production
C) Marketing Authorisation
II. HIT GENERATION
A) RESEARCH AND DEVELOPMENT

Process development – in phytopharmacy

Herbal raw Extraction solvent


material
Extractio
n
Miscella (Liquid raw
extract)

Dry extract Liquid extract, Encapsulatable


tincture mass
Tablets, Liquids, Soft capsules
hard capsules drops,
ointments
II. HIT GENERATION
A) RESEARCH AND DEVELOPMENT

Development of the test substance

Define: - Active substance (in phytopharmacy: native extract)


- Dosage form

Establish: - Physico-chemical profile (active compounds, marker)

Investigate: - Pharmacology
- Mode of action

Prepare: - Patent draft


II. HIT GENERATION
B) QUALITY CONTROL AND PRODUCTION

Raw material supply

Availability of raw materials, excipients, consumables

Herbal raw material


- Established market product ?
- Contract cultivation ?
- Wild harvesting ?

Pay attention to: - Continuous availability


- Quality variations
- Sustainable cultivation / harvesting
- Biodiversity regulations
- Existing patent and intellectual property rights
II. HIT GENERATION
B) QUALITY CONTROL AND PRODUCTION

Identity test, controls

Monographs in pharmacopoeias for:


- Chemical substances
- Herbal raw materials

Organisation of a monograph
Definition: chemical characterisation
Characters: appearance, solubility
Identification: microscopy, physico-chemical tests
Tests: qualitative analysis
Assay: quantitative analysis
Impurities: chemical or microbiological impurities
II. HIT GENERATION
B) QUALITY CONTROL AND PRODUCTION

In house controls

Two standard analytical methods in phytopharmacy:

- TLC = Thin layer chromatography

- HPLC = High performance liquid chromatography


D AD -C H 1 218 n m
C N 002.E12. C 01 con tro l
Retenti on T i m e
300 Area 300

14.800 3943972
250 250

200 200
U

U
150 150
A

A
m

m
100 100

50 50

0 0

0 2 4 6 8 10 12 14 16 18 20 22 24 26 28
Minutes
II. HIT GENERATION
C) MARKETING AUTHORISATION PROCESS

CTD documentation

Common Technical Document:


Harmonised format for applications Structure of the CTD
for preparing marketing authorisation Module 1:
in the three ICH* regions (Europe, Information
Japan, USA)
Module 2:
Summaries
*ICH: International
conference for harmonisation
of technical requirements for
registration of Module 3: Module 4: Module 5:
pharmaceuticals for human Quality Non clinical Clinical
use.
study study
reports reports
II. HIT GENERATION
C) MARKETING AUTHORISATION PROCESS

CTD documentation

Prepare Module 3: Quality

- Monograph
- Specification Module 1:
Information
- Development report (on going)

Module 2:
Summaries

Module 3: Module 4: Module 5:


Quality Non clinical Clinical
study study
reports reports
II. HIT GENERATION
A) RESEARCH AND DEVELOPMENT

Preclinical development

In vitro profiling:
- Biochemical assays (e.g. enzyme activity assays)
- Cell culture assays (e.g. cancer cell lines)
- Isolated tissue assays (e.g. mucosa model)

In vitro toxicology:
Investigate potential toxic effects
in bacteria- or cell cultures
II. HIT GENERATION
A) RESEARCH AND DEVELOPMENT

Working with cell cultures

Cells are kept in liquid Medium and culture flasks for Medium for cell cultures is
nitrogen. cell cultures. pipetted into a culture flask.

Changes of the cultivated


cells are evaluated under the
micro-scope after the addition
of a test substance.

Cultivation of cell cultures in


petri-dishes or cell plates
with the addition of test
substances.
III. LEAD GENERATION

Perspectives: A) Research and Development


B) Quality Control and Production
C) Marketing Authorisation
III. LEAD GENERATION
A) RESEARCH AND DEVELOPMENT

Preclinical development

In vivo testing Animal model (mouse or rat)

Drug action: - Behaviour and reaction


- Physiology
- Histopathology

Toxicology: - Acute toxicity


- Subchronic toxicity
- Tissue specific toxicity
- Tolerability

Consider ethical aspects (e.g. number and kind of animals


used)
III. LEAD GENERATION
A) RESEARCH AND DEVELOPMENT

Preclinical development (continued)

Pharmacokinetic studies What does the body to the


drug ?
Investigate: - Liberation
- Absorption
- Distribution
- Metabolism
- Excretion

Pharmacodynamic studies What does the drug to the body ?


Investigate: - Physiological effects
- Drug action
- Relationship between drug concentration and effect
III. LEAD GENERATION
A) RESEARCH AND DEVELOPMENT

Preclinical development (continued)

Patent policy

Explore the related patent environment:


Database of the European Patent Office
(espacencet)
Develop a patent strategy:
- Rationale
- Possibilities
- Desired strength
- Costs
III. LEAD GENERATION
B) QUALITY CONTROL AND PRODUCTION

Scaling up

Scaling up from laboratory to production size


GMP and GLP environments

Validation

Conduct a process validation including various batch sizes

Stability testing

Conduct a stability test under different conditions of temperature,


humidity and exposure time
III. LEAD GENERATION
C) MARKETING AUTHORISATION PROCESS

CTD documentation

Continue Module 3: Quality

- Validation report
- Stability report Module 1:
- Manufacturing protocol Information
- Development report (on going)
Module 2:
Summaries
Prepare Module 4: Non clinical
study reports
Module 3: Module 4: Module 5:
Quality Non clinical Clinical
study study
reports reports
IV. CLINICAL DEVELOPMENT

Perspectives: A) Research and Development


B) Quality Control and Production
C) Marketing Authorisation
IV. CLINICAL DEVELOPMENT
A) RESEARCH AND DEVELOPMENT

Clinical development – “Linking bench to bedside”

Clinical drug studies – Research in humans

Subject to ethical concern:


- Qualify to increase existing knowledge
- Respect freedom of decision of volunteers
- Involve a substantiated risk-benefit assessment

The realisation of a clinical drug study has to be approved by an


Independent Ethics Commitee (IEC).
IV. CLINICAL DEVELOPMENT
A) RESEARCH AND DEVELOPMENT

Clinical development – “Linking bench to bedside”

Phase I studies 20 to 30 healthy volunteers

Investigate: - Safety and tolerability


- Pharmacokinetics
- Pharmacodynamics
Example:
Dose titration - first application in humans

toxic
Dosage

therapeutic
(mg)

subtherapeuti
c

Treatment
groups
IV. CLINICAL DEVELOPMENT
A) RESEARCH AND DEVELOPMENT

Clinical development – “Linking bench to bedside” (continued)

Phase II studies 100 to 500 patient volunteers

Investigate: - Safety and tolerability


- Pharmacokinetics
- Pharmacodynamics
- Efficiency
- Dosage to effect relationship

Study design: - Dosage comparison

Antitumor drugs: Combination of Phase I and II at an early stage of


drug development is possible.
IV. CLINICAL DEVELOPMENT
A) RESEARCH AND DEVELOPMENT

Overall aim of Phase III: Risk-benefit


evaluation
Phase III studies are “pivotal studies” = outcome is crucial for the
decision taking of the regulatory authorities.
IV. CLINICAL DEVELOPMENT
B) QUALITY CONTROL AND PRODUCTION

Clinical samples

Production - Provide appropriate sample quantities (Phase I, II,


III)
- Define sample shipment logistics

Quality control - Prepare complete batch release documentation


- Define short and long term storage of samples

GMP and GLP environments


IV. CLINICAL DEVELOPMENT
C) MARKETING AUTHORISATION PROCESS

CTD documentation

- Prepare Modules: Module 1:


1: Administrative information Information
2: CTD summaries
5: Clinical study reports Module 2:
Summaries

- Compile the whole CTD


Module 3: Module 4: Module 5:
Quality Non clinical Clinical
study study
Regulatory Authorities reports reports

- Submit the completed CTD


- File a New Drug Application with EMEA (Europe) or FDA (USA)
V. POST REGISTRATION

Perspectives: A) Research and Development


B) Quality Control and Production
C) Marketing Authorisation
V. POST REGISTRATION
A) RESEARCH AND DEVELOPMENT

Clinical development after marketing

Phase IV studies Post marketing testing

Investigate specific questions within the frame of the approved


indication:
- Expanded benefit-risk-profile
- Combination with other drugs
- Optimization (e.g. dosage, application)

E.g.: The worldwide use of the approved drug might lead to the
occurrence of very rare side effects.
Reason for expanded epidemiologic studies
V. POST REGISTRATION
B) PRODUCTION & QC / C) MARKETING AUTHORISATION

Production and quality control

Manufacture - Manufacturing of the product


- Controls acc. to the established batch release
process

GMP and GLP environments

Marketing authorisation process

Approval - Drug is approved for marketing by the Authorities


Summary
I. DISCOVERY
Identify target and resource
II. HIT GENERATION
Develop process and test substance
Conduct in vitro testing
III. LEAD GENERATION
Conduct in vivo testing
Pharmacokinetic and pharmacodynamic studies
IV. CLINICAL DEVELOPMENT
Human trials – Phase I, II, III
V. POST REGISTRATION
Human trials – Phase IV
Thank you for your attention !

Das könnte Ihnen auch gefallen