46
Clinical Pharmacology in Orphan Drug Development KoNECT 3rd Regulatory Symposium CAPT E. Dennis Bashaw, PharmD. Director, Division of Clinical Pharmacology-3 Office of Clinical Pharmacology Office of Translational Sciences CDER/FDA

Clinical Pharmacology in Orphan Drug Development

Embed Size (px)

Citation preview

Page 1: Clinical Pharmacology in Orphan Drug Development

Clinical Pharmacology in Orphan Drug Development

KoNECT 3rd Regulatory Symposium

CAPT E. Dennis Bashaw, PharmD.Director, Division of Clinical Pharmacology-3

Office of Clinical PharmacologyOffice of Translational Sciences

CDER/FDA

Page 2: Clinical Pharmacology in Orphan Drug Development

2

Disclaimer: The presentation today

should not be considered, in whole or

in part as being statements of policy or

recommendation by the US Food and

Drug Administration.

Page 3: Clinical Pharmacology in Orphan Drug Development

3

Outline

• Drug Development and Orphan Drugs/Rare Diseases in the United States: An Overview

• 1983 Orphan Drugs Act-PROBLEM SOLVED?

• Challenges And the Role of the Regulator:Science, Regulation, and Policy

• Bringing Clinical Pharmacology Tools to Bear

• Concluding Thoughts

Page 4: Clinical Pharmacology in Orphan Drug Development

4

DRUG DEVELOPMENT AND ORPHAN DRUGS/RARE DISEASES IN THE UNITED STATES: AN OVERVIEW

Page 5: Clinical Pharmacology in Orphan Drug Development

5

Pre Orphan Drug Act: 1982

• 1973-1982: 10 new drugs for rare diseases

– Little economic incentive for large pharmaceutical companies to pursue rare disease indications

• ≈7,000 rare diseases; 25 million people

– In comparison: 67 million American adults (31%) have high blood pressure—that’s 1 in every 3 people in this room (http://www.cdc.gov/bloodpressure/facts.htm)

• ~85% of orphan diseases have a genetic basis

• Increasing by ~100 diseases/year

Page 6: Clinical Pharmacology in Orphan Drug Development

6

Chronology of Drug Developmentfor Classical and Orphan Drug

Nature Reviews and Drug Discovery, 2003, Volume 2, Page 71

Page 7: Clinical Pharmacology in Orphan Drug Development

7

Trends in Drug Discovery

Scannell, JW, et al “Diagnosing the decline in pharmaceutical R&D efficiency”Nature Reviews Drug Discovery, 11:191-200 (March 2012)

An unsustainable

trend for an Orphan

Drug

Page 8: Clinical Pharmacology in Orphan Drug Development

8

Updated Drug Development Cost Figures

J Health Econ. 2016 May;47:20-33. doi: 10.1016/j.jhealeco.2016.01.012

Page 9: Clinical Pharmacology in Orphan Drug Development

9

Fast Track Designation

• Drug intended to treat a serious condition

• Nonclinical or clinical data needed to

demonstrate the potential to meet an

unmet medical need.

Breakthrough

• Drug intended to treat a serious condition

• Must be preliminary clinical evidence to

indicate the drug may substantially improve a

clinically significant endpoint compared to

available therapies

Priority Review

• Drug must treat a serious condition and, if

approved, offer a significant improvement in

safety or effectiveness

• Designation assigned only at the time of the

original NDA or efficacy filing

Accelerated Approval

• Drug must treat a serious condition and

generally provide a meaningful advantage over

available therapies

• Must demonstrate an effect on a surrogate

endpoint that is likely to predict a clinical

benefit or on a clinical endpoint

FDA’s “Accelerated” Pathways –Not Exclusive to Orphan Drugs

Entering Drug

Development cycleTo Market

https://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm358301.pdf

Page 10: Clinical Pharmacology in Orphan Drug Development

10

Accelerated Pathways Under PDUFA-V

https://www.fda.gov/downloads/forindustry/userfees/prescriptiondruguserfee/ucm552923.pdf

Page 11: Clinical Pharmacology in Orphan Drug Development

11

1983 ORPHAN DRUG ACT –PROBLEM SOLVED?

Page 12: Clinical Pharmacology in Orphan Drug Development

12

The Orphan Drug Act21 CFR314.105

Page 13: Clinical Pharmacology in Orphan Drug Development

13

How Well Are We Doing?

• In past few years

– ~1/3 of all NME approvals are Orphan products

– 2/3 of therapeutic biological product approvals

• While there has been progress in the general science and approval of Orphan Drugs….just like an iceberg much more lies below the surface to be done.

– 7,000 plus indications

• Since 1983

– 4071* drugs with an orphan designation

– 602* drugs approved……..

*as of 4/23/2017

Page 14: Clinical Pharmacology in Orphan Drug Development

14

NME/NDA Approvals: Total vs Orphan

15

24

33

25

30

33

22

14

3

7

11

810

12

9

3

0

5

10

15

20

25

30

35

2010 2011 2012 2013 2014 2015 2016 2017

NME

Orphan NME

*

Page 15: Clinical Pharmacology in Orphan Drug Development

15

NME-BLA Approvals: Total vs Orphan

6 6 6

4

1112

10

6

34

21

7 7

5

3

0

2

4

6

8

10

12

14

2010 2011 2012 2013 2014 2015 2016 2017

BLA

Orphan BLA

*

* As of April 25th, 2017

Page 16: Clinical Pharmacology in Orphan Drug Development

16

CHALLENGES AND THE ROLE OF THE REGULATOR:SCIENCE, REGULATION, AND POLICY

Page 17: Clinical Pharmacology in Orphan Drug Development

17

Regulatory Challenge

• Orphan drugs held to same evidentiary standard as non-Orphan drugs

• To be approved in US, Orphan drugs must:

– Demonstrate substantial evidence of effectiveness/clinical benefit (21CFR 314.50)

– Substantial evidence of benefit requires:

• Adequate and well-controlled clinical study(ies)

– designed well enough so as to be able “to distinguish the effect of a drug from other influences, such as spontaneous change…, placebo effect, or biased observation” (§314.126)

Page 18: Clinical Pharmacology in Orphan Drug Development

18

The REGULATORS “DUAL” Role

National Authority

Regulations Science

Conservative Innovation

ClinicalPharmacology

Regulations provide room for flexibility in the review of treatments for rare diseasesand the application of regulatory standards….i.e., good scientific judgment

Page 19: Clinical Pharmacology in Orphan Drug Development

19

Rare Disease Guidance-Common Issues

Page 20: Clinical Pharmacology in Orphan Drug Development

20

Challenges in Orphan Disease/Rare Drug Development

• Large heterogeneity in disease pathophysiology

• Poorly understood natural histories and progression

• Few patients are available conducting clinical trials

• Uncertain appropriate duration of treatment

• Lack appropriate endpoints that predict outcomes

• Large heterogeneity in treatment effects

• Require compromise, innovation and trade-offs

• Make difficult decisions in absence of ideal information

Proper deployment of Clinical Pharmacology inorphan drug development can extract the mostamount of knowledge from least amount ofinformation

Page 21: Clinical Pharmacology in Orphan Drug Development

21

INNOVATIVE ANALYSES•Improved Computing Resources •Quantitative drug-disease-trial models• Exposure-response models

INNOVATIVE TRIAL DESIGNS• Clinical trial simulations• Enrichment, adaptive, dose-response• In Silico

KNOWLEDGE MANAGEMENT• Leverage prior data• Locate and observe trends• Understand the need

Bringing Clinical Pharmacology Tools to Bear

Page 22: Clinical Pharmacology in Orphan Drug Development

22

Trends in Orphan Drug Approvals and Development

Clin Pharmacol Ther. 2012 May;91(5):932-6.

doi: 10.1038/clpt.2012.23.

Page 23: Clinical Pharmacology in Orphan Drug Development

23

Orphan Drug Approvals (NMEs)by Therapeutic Area

Page 24: Clinical Pharmacology in Orphan Drug Development

24

A Graphical Perspective on Trends inOrphan Drug Development for NMEs

Page 25: Clinical Pharmacology in Orphan Drug Development

25

Orphan/Rare Disease NME Approvalsby Cohort

0

5

10

15

20

25

30

2011-2015 2006-2010

Page 26: Clinical Pharmacology in Orphan Drug Development

26

Orphan Designations vs TOTAL Orphan Approvals*

195 203190

231

291

355333

125

15 26 26 3349 48 39

12

0

50

100

150

200

250

300

350

400

2010 2011 2012 2013 2014 2015 2016 2017

Orphan Designations

Approvals

470 Requests(75% Acceptance)

*Includes Orphan Indications for Approved Drugs (re-purposing)

** As of April 25th, 2017

**

Page 27: Clinical Pharmacology in Orphan Drug Development

27

INNOVATIVE ANALYSES•Improved Computing Resources •Quantitative drug-disease-trial models• Exposure-response models

INNOVATIVE TRIAL DESIGNS• Clinical trial simulations• Enrichment, adaptive, dose-response• In Silico

KNOWLEDGE MANAGEMENT• Leverage prior data• Locate and observe trends• Understand the need

Bringing Clinical Pharmacology Tools to Bear

Page 28: Clinical Pharmacology in Orphan Drug Development

28

Clinical Pharmacology Studies in an NDA

• Bioavailability (Absolute or Relative)

– Radiolabel• Dose and/or Dosage Form Proportionality

• Single Dose Pharmacokinetics

• Multiple Dose Pharmacokinetics

• Metabolism Studies

– P-450 Isoenzymes• Special Populations

– Elderly

– Renal Failure/ Hepatic Failure

– Pediatrics• Misc.

– Formulation Studies

– Bioequivalency (Clinical vs. To-be-marketed)

– Drug Interaction

– Protein Binding

– Mechanism of Action

– Dissolution

Exposure Response

Dose Response

Dose Titration

Surrogate Endpoint

Pharmacometrics

Pharmacogenomics

Page 29: Clinical Pharmacology in Orphan Drug Development

29

Orphan Drug Review at the FDAEliglustat for Gaucher’s Disease

• Gaucher disease is a rare, autosomal recessive lysosomal storage disorder caused by a deficiency in the lysosomal enzyme glucocerebrosidase (or acid-β glucosidase), which catalyzes the hydrolysis of glucosylceramide (or GL-1) to glucose and ceramide. This enzyme deficiency results in the accumulation of GL-1, especially in the liver, spleen, and bone marrow.

http://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/205494Orig1s000ClinPharmR.pdf

Page 30: Clinical Pharmacology in Orphan Drug Development

30

Eliglustat (Cerdelga®)

• Eliglustat is a selective glucosylceramide synthase inhibitor for substrate reduction therapy (SRT) to reduce the synthesis and hence the accumulation of GL-1.

• Received both a Priority Review and Orphan Drug Designation

Page 31: Clinical Pharmacology in Orphan Drug Development

31

Metabolic Information Available on Eliglustat Prior to PBPK Modeling

• Metabolized by CYP2D6 (~80%) and CYP3A (~20%)

• High clearance, nonlinear PK: time-dependent CYP2D6 inhibitor

• Clinical drug interaction studies

– With strong CYP2D6 inhibitor paroxetine: AUC increased by ~8-fold

– With strong CYP3A inhibitor ketoconazole: AUC increased by ~4-fold

• Pharmacogenetic effects: PM/EM ~ 8-fold

Page 32: Clinical Pharmacology in Orphan Drug Development

32

Combining the Workstreams

PharmacogenomicsUtilize in vitro systems

to identify relevant CPY

polymorphism

PBPK ModelingBuild models based on

observed knowledge with a

“learn and confirm” strategy.

Classical PK/PDSynthesize the

available PK/PD data

on Drug Metabolism

Develop

Actionable

InformationInformed labeling for the

prescriber

Page 33: Clinical Pharmacology in Orphan Drug Development

33

Impact of PBPK on Labeling

http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/205494Orig1s000lbl.pdf

Page 34: Clinical Pharmacology in Orphan Drug Development

34

Impact of PBPK on Labeling

http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/205494Orig1s000lbl.pdf

Page 35: Clinical Pharmacology in Orphan Drug Development

35

CONCLUDING THOUGHTS

Page 36: Clinical Pharmacology in Orphan Drug Development

36

FDA-NIH Collaboration

Page 37: Clinical Pharmacology in Orphan Drug Development

37

FDA-TRND Collaboration

Page 38: Clinical Pharmacology in Orphan Drug Development

In October 2011, FDA awarded $2 million to launch Centers ofExcellence in Regulatory Science and Innovation at theUniversity of Maryland and Georgetown University. Theinvestment is part of FDA’s effort, outlined in the Agency's

strategic plan, to foster a robust, collaborative, regulatoryscience culture that enables FDA to address the scientific

challenges presented by revolutions in medical productdevelopment and to improve food safety and quality. In 2014two new centers were established in collaboration with the FDA.

FDA-Academic Collaboration

Page 39: Clinical Pharmacology in Orphan Drug Development
Page 40: Clinical Pharmacology in Orphan Drug Development

40

An International Concern

Page 41: Clinical Pharmacology in Orphan Drug Development

41

Policies to Incentivize Orphan Drug Development - APAC & Major Regions*

POLICY elements USA EU Australia Japan S. Korea Singapore Taiwan

Legal framework (1983)

(2000)

(1997)

(1993)

(1998)

(1991)

(2000)

Prevalence (per 10,000)

7.5 5 1 4 4 36 1

Other criteria

Life-threatening &

chronically debilitating

Incurable disease with no

alternative treatment

No alternative treatment

Marketing exclusivity 7 years 10 years - 10 years 6 years§ - 10 years

Tax credit/Grants for R&D - - - - -

Accelerated MA procedure ** ** **

Fee reduction - - -

Protocol assistance - - -

*Slide modified from David Tsui (Shire Pharmaceuticals** For life saving drugs where there is no therapeutic alternative§ proposal to increase to a maximum of 10 years

Page 42: Clinical Pharmacology in Orphan Drug Development

42

International Update (beyond US and APAC)

Page 43: Clinical Pharmacology in Orphan Drug Development

43

Development of Safe and Effective Drugs For ALL Requires a Team Effort

Academia

IndustryInternational

Collaboration

Patient

Advocacy

FDA Science

& Policy

Benefits

To All

Page 44: Clinical Pharmacology in Orphan Drug Development
Page 45: Clinical Pharmacology in Orphan Drug Development

45

Contact Information

CAPT Edward D. Bashaw, PharmD.Director, Div. of Clinical Pharmacology-3US FDA10903 New Hampshire AveBuilding 51, Rm [email protected]

Page 46: Clinical Pharmacology in Orphan Drug Development

46

Acknowledgements• The Staff of the Division of Clinical Pharmacology-3

• The Office of Clinical Pharmacology

• The Office of Translational Sciences