drug discovery : from lead optimization to clinical ... discovery : from lead optimization to...

65
Drug Discovery : From Lead Optimization to Clinical Candidate and beyond Giovanni Gaviraghi Chief Executive Officer Siena Biotech [email protected]

Upload: lamnhu

Post on 25-May-2018

217 views

Category:

Documents


1 download

TRANSCRIPT

Drug Discovery : From Lead

Optimization to Clinical Candidate

and beyond

Giovanni Gaviraghi

Chief Executive Officer

Siena Biotech

[email protected]

Drug Discovery Technologies & Competencies

Where are they concentrated?

Launch, Marketing

etc.Phase IIIPhase II

FTIHTo

PoC

Candidateto ClinicalCandidate

Lead to Candidate

Hit toLead

Screen to Hit

Target to Validated

Target

Gene to Function

Skill and Competencies available at University, Hospital and basic research Centres

Skills and Competencies available at Big Pharma & partially in the small biotech Drug Discovery

Skills and Competencies available in Fully Integrated Pharma and Biotech Companies

Basic Research Pharmaceutical Research Clinical Research

Studio della

malattia

Il Modello di Siena Biotech : ponte fra ricerca accademica e

clinica

Nuove cure piu’ efficaci e

sicure

Università

Grandi Aziende

Farmaceutiche

Multinazionali

Ricerca applicata per

nuovi farmaci

Ricerca di baseRicerca applicata

Clinica Mercato

Attività e competenze presenti in: Università, Ospedali, CNR ecc.

Attività e competenze presenti nelle grandi aziende farmaceutiche multinazionali

Selecting and Progressing the Right

Compounds

Launch,

Marketing

etc.

Manufac-

turing

Clinical

Phase III

Clinical

Phase II

Clinical

Phase I

Candidate

to Clinical

Candidate

Lead to

Candidate

Hit to

Lead

Screen

to Hit

Target to

Validated

Target

Gene to

Function

Discover molecules to modulate a target

Assess whether the molecules could be optimised

Optimise the series for multiple drugability parameters

Assess whether the compound can enter clinical trials

€ €€ €€€€

€€€€

Non-Linear Project Progress Path

Commit to

bioassay design

and screening

Commit to lead

optimization

Commit to target

validation

Commit to lead

identification

Lead to

Candidate

PhenotypeScreening

Commit to

exploratory

biology

Target ID

ForwardChemicalGenetics

Drug DesignChemistry

Gene to Function

Hit to Lead

Target Validation

Screen to Hit

Selecting and Progressing the Right

Compounds

Launch,

Marketing

etc.

Manufac-

turing

Clinical

Phase III

Clinical

Phase II

Clinical

Phase I

Candidate

to Clinical

Candidate

Lead to

Candidate

Hit to

Lead

Screen

to Hit

Target to

Validated

Target

Gene to

Function

Discover molecules to modulate a target

Assess whether the molecules could be optimised

Optimise the series for multiple drugability parameters

Assess whether the compound can enter clinical trials

€ €€ €€€€

€€€€

How to start the discovery process

TPP TCPLead

Criteria

Hit

Criteria

Target

Criteria

Project Progression

Market Need

Backwards Planning

Decision Gates

(“Will this product be competitive ?”)

Design Criteria

(“What properties should the drug have ?”)

Target

Product Profile

Target

Candidate Profile

The Current Development Paradigm

Patho-

physiologyCIM Confidence in Mechanism

CIS Confidence in Safety

– Iterative, parallel

– Data-driven

– Bi-directional

Evaluating the Project Opportunity; Financial returns.

Timing and Probability of Technical Success

0 2 4 6 8 10 12

Assay

development

Hit to lead

Lead optimization

D

I

S

C

O

V

E

R

Y

HTS screening

Target ID

Years

Preclinical

Phase I

Phase IIA

Phase IIB

Phase III

D

E

V

E

L

O

P

M

E

N

T

Registration

37

28

25

19

9

5

3

2

1.5

1.1

1

Cost*

0.4

1.2

9.0

0.5

~0

5.3

9.3

12.5

32.1

98.0

11.8

0.5

1.5

<0.25

1.25

1

Time**

0.25

0.25

1

1.5

2

1.5

37

POS***

75%

75%

45%

90%

100%

60%

67%

60%

75%

75%

88%

* $, millions

** Years

*** Probability of success

2.7%

Industry averages

~$1 B and 10-12 years

What determines the financial value of R&D?

Sales of the

resulting

product(s)

Cost

Time

Risk

Operating and

strategic options

The financial value of

an R&D project

Many Sources of Uncertainity

Technical

Timing

Market

Commercial

Efficacy

SafetyTolerability / Convenience

Manufacturing ( eg. CoG )

Annual cash flow

Order of market entry

Epidemiology / Segmentation

Healthcare

reimbursement/access

DiagnosisCompliance

Competition

Price

Resources

Development Phase is reached: Re-assessing Product Profile,

Project Potential and Market Opportunity

In other words, construction of the

final Target Product Profile starts at this point.

Target Product Profile is developed on the basis of

a combination/compromise of many customers’

needs and expectations

Key Indicator Document

Key Indicator Document

– Detailed analysis of disease/condition from a clinical, marketing

and regulatory perspective

– Current and future situation; epidemiology

– Careful analysis of competitor situation

– Market dynamics and Healthcare Provider aspects

– Produced by Business and Clinical Research as starting point for

setting corporate strategy

The Logic of a Profile-Driven Process

Disease Description

Commercial Opportunity

Competitive Landscape

Market Forecast

Key Commercial Potential

Market Dynamics

Unmet Medical Need

Epidemiology

Current Gold Standard

Future Gold Standard

Target

Product

Profile

Identifying what

characteristics the

market product

should have to

be competitive

Target

Candidate

Profile

Specifies criteria

predicting the TPP

Indication

Population

Efficacy

Risk/Benefit

Dosage and

Administration

IP

Differentiators

Risk Management

– Cost of Discovery Process (to Candidate) M€ 5-10 in 3-5 years

– Cost of Development Process (to Market) M€ 500-1000 in 5-10 years

therefore

– Milestone decisions in Discovery mainly represent scientific risk-taking,

aiming to reach the TCP.

– Milestone decisions in Development represent economic risk-taking,

aiming to reach the TPP.

The Lead to Candidate Phase

Integrated R&D Phases

Launch,

Marketing

etc.

Manufac-

turing

Clinical

Phase III

Clinical

Phase II

Clinical

Phase I

Candidate

to Clinical

Candidate

Lead to

Candidate

Hit to

Lead

Screen

to Hit

Target to

Validated

Target

Gene to

Function

Discover molecules to modulate a target

Assess whether the molecules could be optimised

Optimise the series for multiple drugability parameters

Assess whether the compound can enter clinical trials

€ €€ €€€€

€€€€

Primary Screen

(IC50, EC50)

Secondary Screens(selectivity, functional end-points)

Med ChemTier ISolubility

Chemical stability

Protein Binding

Permeability

Metabolic Stability

Electrophilicity

PK/metabolism in

non-rodent species

Human target plasma level,

preliminary safety assessment

Global/local in silico

property prediction,

QSPR, PRISM,

target organ/species

prediction

In vitro PK/PD

PB/PK

PK/PD, biomarker

modelling

Allometry, PB/PK

Population approaches

Clinical trial

simulation

Tier IIIntrinsic clearance

Metabolite profile

MDCK-I

hERG

Ames test

Micronucleus test

Cytotoxicity

Preformulation

Interaction potential

Phenotyping

P-gp ATPase

C-AM Uptake

CYP induction

...

PK screening, IV/PO

Primary in vivo

pharmacology modelExploratory in vivo

toxicity studies

Profiling

Parameters

In Silico ADMET

& PK/PD

Screening Cascade

A Continuous Process

Drug Design

Tier I

Tier II

Lead optimisation is not just about potency!

Other important parameters for a successful new medicine

Desired Lead Profile

Confirmed structures and structural classes

SAR indicating potential for optimisation

What are the areas of the molecule responsible for affinity?

Which areas of the molecule can be changed without affecting affinity?

Potential for patent protection (looks promising with general searches,

identified areas to avoid)

Potent in vitro activity has been achieved for the series

e.g. <100 nM or <5 M in a phenotypic assay

Some selectivity activity versus closely related targets

Tractable chemistry - no obvious synthetic problems with class

Promising Tier 1 & 2 ADME data with an understanding of potential problems

with the lead class

An indication of activity in vivo.

Desired Preclinical Candidate Profile

The molecule(s) should have\be

Potent and selective in vitro activity

Efficacy demonstrated in appropriate in vivo models

Acceptable selectivity profile in “Cerep” and “cardiac” panels

Acceptable/manageable toxicity profile in preliminary in vivo settings

Patented/patentable

Scaleable chemistry – “fit for purpose” route available to make 50-500g

“Fit for purpose” formulation for tox and additional efficacy studies

Physicochemical properties aligned for a candidate molecule (more later)

Backup molecule(s)

Build understanding of mechanism

At Preclinical Candidate Selection

Alea iacta est

This is the end of “structural optimisation”

We are now selecting the molecule (or in some companies 2-3

molecules) to go into preclinical development.

It will succeed or fail based on its developability

The only changes to the compound that can be made to improve

chances of success are:

Drug delivery – formulation, dose regime etc

Cost of goods – improved synthesis on large scale

Typical good hits/initial leads are/have:

Potent against the cellular target (low nanomolar IC50)

Defined Structure Activity Relationship (SAR)

Synthetically accessible

Good to moderate patent position

Some indication/promise of efficacy in vivo

Moderate solubility

Selectivity vs. other enzymes/receptors not ideal

Needing improvement of in vivo potency

Unoptimised oral bioavailability or other PK parameters

Possible dose limiting toxicity

No advanced backup class should problems occur

So what are we trying to optimise?

Secondary

properties

of the

molecules

Where to modulate secondary properties of the molecule?

Mapping key pharmacophore features during H2L is

important in deciding where changes can be made

O

N

NHO

R3

O

R2

R1H

H

H

HH

R3 = small groupEssential

R1 = wide rangeof substituents

unsubstituted

R2 = H, F

R1 = wide range of

substituents

Unsubstituted

R3 = small groupEssential

R2 = H, F

Introduction of solubilising groups

N

N NH

N

NH

O

N

N

Key for interaction with targetHydrophilic solubilising group

Glivec (Novartis)kinase inhibitor

Leukaemia

NN

O

OH

O

NH

F

NH

O

O O

O

ONH

2

Cl

Amlodipine (Pfizer)Ca2+ channel blocker

Hypertension

Indinavir (Merck)Protease inhibitor

HIV

N

N

N

NH

O

OH

O

NH

OH

Ciprofloxacin (Bayer)Gyrase A inhibitor

Antibacterial

Introduction of solubilising groups

N

N NH

N

NH

O

N

N

Glivec (Novartis)kinase inhibitor

Leukaemia

Why do drugs still fail in development?

...and how optimising drug-like properties can help

Changing reasons for failure during development

10%

30%

0%

40%

4%11%

5%

12%

26%

4%

9%

4%

20%6%

15%

18%

3%

6%

4%

35% 7%

19912000

2008

What are drug-like properties?

The compound has to dissolve, survive in and be absorbed from the

gastrointestinal tract before reaching the site of action

Poor GI absorption (oral bioavailability) will reduce the dose reaching the

target organ.

Higher initial doses required (compliance)

GI side-effects increase (e.g. antibacterials)

A goal of drug discovery is to maximise the oral bioavailability of

compounds as early in optimisation as possible.

A simple but radical approach

“`the rule of 5' predicts that poor [oral] absorption or permeation is more

likely when there are....

>5 H-bond donors,

>10 H-bond acceptors (≈ N+O atoms),

the molecular weight is greater than 500

calculated Log P (CLogP) is greater than 5”

1.C. A. Lipinski et al. Advanced Drug Delivery Reviews, 1997, 23, 3-25

Breaking two of these rules is very likely to cause a major permeability problem

OH

N

O

H

OMe

N

O

H

Changing reasons for failure during development

10%

30%

0%

40%

4%11%

5%

15%

18%

3%

6%

4%

35% 7%

1991

2008

Significant reduction in failure due to PK/Bioavailability

Current challenges

toxicity/safety (side effects)

lack of efficacy

How can we improve the situation in Lead Optimisation?

Development failure due to lack of efficacy

If related to poor links between the biological target, in vitro assays

or in vivo models to the human disease there may be little that can

be done in this phase, especially for “first in class” projects

Evidence that some alleged development failures due to lack of

efficacy are actually due to safety problems

Toxicity/Safety - a complex subject with many potential causes

Modulating the biological target may cause unwanted/ unpredicted

side effects

• Potential for improved safety if cause is tissue selectivity

• For example increased/decreased brain penetration with a new

molecule

Lack of selectivity is a significant cause of toxicity/side effects

• Selectivity improvements through increased potency for target

• Selectivity through changes to physicochemical properties.

Summary

Lead optimisation is our last chance to find the best compound in

an active series

It’s not about in vitro potency

We must reduce dose required for efficacy

Improvements to ADME/PK through physicochemical property

improvements

We must increase concentration required for toxicity

Reduce toxicity liabilities through physicochemical property

improvements

We must deliver the molecule (plus backups) for rapid progression

to the clinic

No perceived major issues for large-scale synthesis,

pharmaceutical development etc.

Therapeutic

index or

Therapeutic

window

Design Criteria for candidate to clinical candidate

From candidate to clinical candidate – Overview

Objectives

• enable Phase I Clinical Trials through the filing of an IND/CTA

Goals and activities

• Pharmacology:

– confirm efficacy in relevant animal models

– define the Minimum Active Biological Expected Level (MABEL)

• Preclinical Development:

– define safety profile

– API process development and manufacturing

• Regulatory/Clinical:

– Regulatory documents preparation and submission (IB/IMPD)

– Clinical site selection and clinical protocol definition

Project

Milestones

API/

Pharm. Dev.

Toxicology

Clinical/

Regulatory

2007 2008 2009 2010

ADME/PK

Pharmacology

H2H1

FTIMCommit

to GLPLOIIND/CTA

Submitted

2011

H2H1 H2H1 H2H1 H2H1

R6/2

EnablingR6/2

Main StudyBiomarker Development and Validation

Other HD Models

1° Non-

GMP Lot

Process

ResearchPreformulation

1° GMP Lot 2nd GMP Lot

Clinical Supply

Regulatory Stability

DRFs28-Day

GLP

Safety

Pharma-

cology

Gene

Tox

Mini-

Ames

hERG

MTD

R6/2

Enabling

2° Species

Selection

Formulation DevelopmentAnalytical Package

Formulation Development

Clinical

Supply

Bioanalytical

Method Toxicokinetics

Orphan Designation

IMPD/CTA

Phase I

Subchronic Toxicology

Segment II + III

Phase IIa

Discipline interaction

From candidate to clinical candidate: Preclinical

Development

Safety profile definition:

• Preliminary (Non GLP) studies

• Regulatory (GLP) studies

Process development and pharmaceutics need to ensure GLP/GMP

quality standard for API used for preclinical/clinical studies

Regulatory guidelines very clearly defined

Design activities to support the proposed clinical program, delaying

non-critical studies until after cPoC

Current Industry benchmark: first GLP dose to FTIM in 7 months

Safety profile definition continues in parallel with Clinical

Development, timed to coincide with requirements of clinical program

up to the NDA/MAA filing

From candidate to clinical candidate: Interaction among

disciplines

Toxicology

Single and repeated dose,

Genotox, Reprotox, Local

tolerance, etc.

Safety Pharmacology

core battery studies (CNS, CV,

Resp), special studies.

ADME

Bioanalysis, (PK/TK),

protein binding,

biotransformation

Process Development &

Pharmaceutics

Formulation supply and

conc. checksQuality Assurance

Facility and Study Audits

From candidate to clinical candidate: Critical Path

definition

From candidate to clinical candidate: Safety assessment (1)

Preliminary safety evaluation (non-GLP pakage)

ADME• Single dose PK (rodents and non rodents)

• Met profile invivo

Toxicology • Single/repeared dose DRF: in parallel (rat) or escalating (dog or monkey)

• Repeated MTD tox + TK (2wk), rat and dog or monkey

Safety Pharmacology• CV: cardiac panel, purkinjie fibers, isolated heart, single dose ECG, telemetry

• CNS: Irwin test/FOB – rat

Genotox studies• In vitro mutagenicity in bacteria – Miniames

• In vitro chorm abs in mammalian cells - COMET or µnucleous

09/02/2011

Pharmacokinetics

Single dose

Multiple dose

Linking pharmacology, toxicology and pharmaceutical development:

● What dose ?

● How often ?

● How to deliver ?

PK data in support of:

● Compound selection

● Dose finding

● Interspecies scaling

● Human predictions

● Understanding pharmacology and

toxicology data

Pharmacokinetics

Dedrick Scaling

RatDog

Predicted Human

Allometry

PK/PD Models

Population Approaches

Metabolism

Identification of active and reactive metabolites from in vitro and in vivo samples across species by LC-MS/MS (exact mass):

● Potential toxicity issues relating to reactive intermediates

● Toxicology species selection

● Monitoring of metabolites in toxicology and clinical studies

● IP Opportunities

From candidate to clinical candidate: Safety assessment (2)

Regulatory safety evaluation (GLP pakage)

Toxicology – repeated dose studies

• Repeated dose + TK in rodents (mouse, rat)

• Repeated dose + TK in non rodents (dog, monkey, minipig)

Safety Pharmacology – single dose studies

• CV: telemetry (dog, monkey, ferret, guineapig)

• CNS: Irwin test /FOB (rat, mouse)

• Respiratory: Plethismographic system (rat, dog, monkey)

• Other studies based on specific issues (e.g. GI, kidney, etc.)

Genotox studies

• In vitro mutagenicity in bacteria – Ames

• In vitro chorm abs in mammalian cells – HPLA

• In vivo: rat µnucleous

Study Type RouteSpecies/

StrainDose Levelsa

Repeat-Dose Toxicity Studies

2-Week toxicity (Non GLP) Oral Rat/Sprague Dawley 0, 30, 100, 200 and 300 mg/kg/day

4-Week toxicity with 4-week recovery (GLP) Oral Rat/Sprague Dawley 0, 20, 60 and 150 mg/kg/day c

5-Day escalating dose (non-GLP) Oral Dog/ Beagle 10, 30, 60, 100, 150 mg/kg/day

2-week toxicity (non-GLP) Oral Dog/ Beagle 50 and 100 mg/kg/day

4-week toxicity with 4-week recovery (GLP) Oral Dog/ Beagle 0, 12, 30 and 75 mg/kg/day

Genotoxicity

Ames Assay (GLP) In vitro

Bacterial/

S.typhimurium and

E.coli

1.6 to 1000µg per plate with and without S9

Chromosomal aberrations (GLP) In vitroHuman peripheral

lymphocytes3h at 5 to 130 µg/ml with and without S9

Mammalian erythrocyte micronucleus test

(GLP) Oral Rat/Sprague Dawley 0, 75, 150, or 300 mg/kg

Study Type RouteSpecies/

StrainDose Levelsa

Safety Pharmacology Studies in vivo

Neurobehavioral study – Irwin test (GLP) Oral Rat/Sprague Dawley 0, 30, 100, and 300 mg/kg/day

Respiratory function – Plethysmographic

test (GLP) Oral Rat/Sprague Dawley 0, 30, 100, and 300 mg/kg/day c

Cardiovascular function - Telemetry (GLP) Oral Dog/ Beagle 0, 25, 50, and 100 mg/kg/day

Safety Pharmacology Studies in vitro

Ikr channel – Manual pach clamp (GLP)] In vitroHEK 293 trasfected

with hERG

0, 3, 10, 30, 100, and 300 µM

Overview of a preclinical safety package

From candidate to clinical candidate: to do list (1)

Before GLP Tox pakage starting

explore toxicity in a broad dose range in vivo

define MTD and possible target organs

define PK profile at different dose levels and schedule (single and repeated)

identify relevant metabolites in vitro and in vivo in different species

define study design for Regulatory studies

• animal species

• dose level and schedule

• formulation schedule.

prepare GLP/GMP API batch for the following regulatory studies

set up and validate analitical and Bioanalitical methods

verify stability of API and dosing solutions

define API specs and release GLP/GMP batch(es)

From candidate to clinical candidate: to do list (2)

During GLP Tox pakage

investigate in deep the toxicity in a well defined (non lethal) dose range

• accomodation to toxic effects and/or recovery

• gender differences

• target organs and the reversibility of lesions

• systemic exposure

compare toxicity findings across species

define NOAEL /NOEL for each in vivo study

Identify potential risk for humans and possible monitoring strategies

confirm metabolite profile in vivo

prepare regulatory documents to support IND/CTA

select clinical site for Ph1 study and book study slot

prepare formulation for clinical use based on escalation rules

From candidate to clinical candidate: Decision to be

taken

At the end of preliminary Tox pakage

Evaluate the overall potential for harm on acute and repeated administration

Evaluate the relevance of lesions for humans

Identify unacceptable risks

Evaluate preliminary safety margins

Understand mechanism of toxicity and the possibility to monitoring tox effect in man

Decide whether or not progressing in the Regulatory phase

At the end of GLP Tox pakage

Confirm the overall safety profile of the compound

Confirm that changes are reversible and monitorable

Calculate the starting dose for clinical studies

Commit the beginning of clinical study and regulatory submission (IB/IMPD)

Active Pharmaceutical Ingredient (API)

• Structure, names, physico-chemical properties

• Manufacturing Process and In-Process Controls

• Physical form, solubility, solid state properties, hygroscopicity, polymorphism, dissolution rate,

impurities and degradation

• Stability (solid, solution, pH, humidity, light)

• Analytical Specifications

Finished Dosage Form (FDF)

• Pharmaceutical Formulation

• Excipient properties, compatibility and stability

• FDF solubility, solid state properties, hygroscopicity, polymorphism, dissolution rate, impurities

and degradation

• Stability (solid, solution, pH, humidity, light)

• Analytical Specifications

From candidate to clinical candidate: Physical, Chemical

and Pharmaceutical Properties

Drug Product Specification SENX

Test Method Limits

Appearance Visual Size 0 Swedish Orange capsule with no signs of

splitting or breakages, containing white to off-

white powder.

Identity by HPLC AM2665 Identity is confirmed if the main peak in the

sample corresponds to the main peak in the

standard

Assay by HPLC AM2665 95.0 – 105.0% of Label Strength

Impurities by HPLC AM2665 Any Unspecified Impurity NMT 0.5%

Total Impurities NMT 2.0%

Content uniformity by

HPLC

AM2665 Complies with Ph.Eur. 2.9.0

Dissolution AM2664 Q 75% at 45mins

Microbial Limit Test1 Ph Eur 2.6.12

& 2.6.13

Total Aerobic Count: NMT 1000cfu/g

Total Microbial Count: NMT 100cfu/g

Absence of E.Coli: Confirmed

From candidate to clinical candidate: Clinical

Formulation Development and Manufacturing

Clinical and Regulatory Development

The Current Development Paradigm

Full DevelopmentLife

Cycle

Management

Initial Approval

DiscoveryPreclinical

Development “Proof-of-Concept”

Confirm

Full Approval

Line Extension

Learn

–Proof of Pharmacokinetics

–Proof of Safety

–Proof of Target Modulation

–Proof of Mechanism

–Proof of Efficacy

–Proof of Commercial Viability

– Iterative, parallel

– Data-driven

– Bi-directional

Clinical Development

– Clinical Development strives to achieve registration according to the TPP

– The Clinical Development process is strongly dependent on Health Authority

regulations

– Guidelines relating to clinical studies define minimal criteria for Manufacturing,

Pharmaceutical form, Preclinical Safety and Quality. Such activities are

conducted according to GxP:

o Good Clinical Practice (GCP)

o Good Manufacturing Practice (GMP)

o Good Laboratory Practice (GLP)

– Clinical studies for use in regulatory submissions need to have a defined study

design – typically, registrational studies are conducted with a double-blind,

placebo-controlled design

“Do only what is required to get to the next decision point”

– Process-driven rather than science-driven

– Clear definition of “need-to-have” vs. “nice-to-have” and decision trees

– Delay non-critical activities as far as possible

– Continuous focus on the critical path

– Guidelines aim to ascertain safety and ethics of clinical trials

– Guidelines establish minimal criteria for risk/benefit assessments

– Usually, competitor activities force Sponsors to conduct studies not required by

guidelines

– Guidelines enforced by regular Inspections, especially as regards manufacturing and

documentation

“Development

Mind-Set”

Regulatory Affairs and the Development Process

What is an Indication ?

– A drug can only be prescribed for the indication(s) for which it has been approved

– A drug is approved only for a specific indication(s), for which safety and efficacy data have

been collected

– “Indication” is a precise regulatory term, with a precise meaning and is NOT the same as

“disease”

Disease Indication

Small Cell Lung Cancer Second-line treatment of patients with locally advanced or metastatic non-small cell lung cancer after failure of both platinum-based and docetaxel chemotherapy.

Glioblastoma Multiformae First line treatment of adult patients with newly diagnosed glioblastoma multiforme concomitantly with radiotherapy and then as main-tenance treatment. Second-line treatment of adult patients with refractory anaplastic astro-cytoma, i.e. patients who have experienced disease progression on a drug regimen containing nitrosourea and procarbazine.

Depression Clinically manifest bipolar disorder as adjunctive to lithium or valproate in pediatric patients aged 10 to 17 years

What is a Label Claim ?

– The Label Claim comprises the Safety and Efficacy chapters of the TPP and forms the

basis for NDA/MAA approval

– A Label Claim is often written in the form of Clinical and Commercial Objectives, e.g.

“Compound X is indicated for the first-line treatment of Y, with efficacy not inferior to Gold

Standard, and associated with a frequency of adverse events lower than Gold Standard.”

– Approvable label claims are defined in dialogue with regulatory agencies and must be

reached to achieve registration

– Selection of label claims depends on data from exploratory clinical trials and positioning

vs. competitor products

– Each label claim has to be associated with a unique study

Clinical Development

Prior to Starting a Clinical Trial

1. Approved IND (US, Japan); CTA/IMPD (EU)

2. Local Ethics Committee Approval (IEC/IRB)

3. Signed Clinical Study Protocol

4. Clinical Site Defined (Academic ? CRO ? Investigators ?)

5. Import/Export Licenses

6. Clinical Supply and related documentation

IND: Investigational New Drug application

CTA: Clinical Trial Application

IMPD: Investigational Medical Product Dossier

IEC: Independent Ethical Committee

IRB: Institutional Review Board

CRO: Contract Research Organisation

Phase I

Clinical Phase I Studies

Study Population

Male and female healthy volunteers, 6+2 per cohort

Study Design

Randomized, double-blind single ascending dose (SAD) and

a multiple ascending dose (MAD) element with placebo control

Primary Objective

To determine the safety and tolerability

of single oral escalating doses of SEN0012345

Secondary Objectives

To determine the pharmacokinetic profile of single oral escalating doses of SEN0012345. To determine

expression of XXX in blood/plasma/urine/CSF/tumour/bone marrow

Primary Endpoints

– Safety (laboratory safety test results, telemetry and 12-lead ECG including QTc intervals, vital signs

and physical examination findings.

– Tolerability (type and frequency of adverse events)

Secondary Endpoints

Genotyping for expression of YYY, phenotyping of functional expression of YYY, food effects,

pharmacokinetic interactions etc

0 12 24 36 48

Time (h)

0.01

0.1

1

PH

A-6

83

42

5 C

on

ce

ntr

ati

on

(n

g/m

L)

P H A -683425 P las m a C onc en t ra t ion A f t e r 0 .5 m g P H A -690509 in f us ion (1 h )

1

23456

789101112

Not all “Phase I studies” are conducted during “Phase I”

– Food Interactions

Single dose level w/ and w/o standard meal to assess impact on absorption. Patients or volunteers.

– Thorough QT Study

Required in new ICH S7 guidelines if compounds demonstrates potential for QT prolongation, even if

no in-life findings in pre-clinical studies.

– Drug-drug Interactions

Required if compound demonstrates potential for DDIs at the levels of CYPs or transporters

– Impaired Renal Function

Required if drug is predominantly cleared via renal excretion.

– Impaired Hepatic Function

Required if drug is predominantly cleared via hepatic clearance.

– Bioequivalence Studies

Required if formulation has been changed and Sponsor wants to make use of “old” data. Can be

onerous and expensive.

Other Phase I Studies

“Phase II”

Proof-of-Concept

What is meant by cPoC ?

– May not be a single study, but the accumulated and integrated evidence from Discovery and early clinical

studies

Not an official term, no guidelines; represents what the Sponsor considers sufficient evidence to progress

to Full Development. Based on Pharmacokinetics, Safety, target modulation, MoA.......

– Phase IIa vs. IIb

Phase IIa often small, open-label studies aimed at establishing response in a relevant population. Read-

out based on a small set of clinical end-points and/or biomarkers. Phase IIb confirmatory studies with

double-blind design, often using the intended clinical end-point.

– Learn Process !

Both positive and negative data fed back to Discovery for back-up and/or second-generation compounds.

CONCLUSION

Preclinical drug development is a complex, long and risky

process

To achieve the success you may need:

Top science

A clear objective (product profile)

Accurate planning of activities

Integrated development competences

Financial resources

Customer focus

Capability to predict the market value of the drug

Luck