toxicity testing for oncology drugs theresa reynolds ... › upload › content › talk ›...

17
Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni 1 The screen versions of these slides have full details of copyright and acknowledgements Toxicity Testing for Oncology Drugs 1 Theresa Reynolds BA, DABT Director, Toxicology Genentech, Inc. Krishna Allamneni DVM, PhD, DABT Scientist, Toxicology Genentech, Inc. This presentation represents the opinions of the presenters and is not intended to reflect the policy or position of Genentech, Inc. 2 Presentation objectives 1. Convey: a) Special considerations for the development of oncology drugs and b) How they impact the toxicology program 3 2. Describe the value of a PD marker in oncology and cite examples 3. Describe the key similarities & differences in toxicity evaluation for small and large molecules 4. Present current regulatory guidance and impact on drug development for oncology

Upload: others

Post on 04-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

1The screen versions of these slides have full details of copyright and acknowledgements

Toxicity Testing for Oncology Drugs

1

Theresa ReynoldsBA, DABT

Director, ToxicologyGenentech, Inc.

Krishna AllamneniDVM, PhD, DABT

Scientist, ToxicologyGenentech, Inc.

This presentation represents the opinions of the presenters and is not intended

to reflect the policy or position of Genentech, Inc.

2

Presentation objectives

1. Convey:

a) Special considerations for the development of oncology drugs and

b) How they impact the toxicology program

3

2. Describe the value of a PD marker in oncology and cite examples

3. Describe the key similarities & differences in toxicity evaluation for small and large molecules

4. Present current regulatory guidance and impact on drug development for oncology

Page 2: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

2The screen versions of these slides have full details of copyright and acknowledgements

Despite new therapies, little change in overall cancer incidence

4SEER 9: National Cancer Institute, April, 2008http://seer.cancer.gov/index.html(accessed on 1/5/09)

Estimated number of cancer survivors in the United States from 1971 to 2005

5

Data source: Ries LAG, Melbert D, Krapcho M, Stinchcomb DG, Howlader N, Horner MJ, Mariotto A, Miller BA, Feuer EJ, Altekruse SF, Lewis DR, Clegg L, Eisner MP, Reichman M, Edwards BK (eds). SEER Cancer Statistics Review, 1975-2005, National Cancer Institute. Bethesda, MD, http://seer.cancer.gov/csr/1975_2005/, based on November 2007 SEER data submission, posted to the SEER web site, 2008 (accessed 2/5/09).

5-year relative US survival rates from all cancers have improved from 35% (1950-1954) to 67.5% (1996-2004)SEER Cancer Statistics Review 1975-2005, US National Cancer Institute SEER.cancer.gov (accessed 2/5/09)

a) Why do oncology drugs have special considerations for drug development?

1.

6

considerations for drug development?

b) What are they, & how do they impact the toxicology program?

Page 3: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

3The screen versions of these slides have full details of copyright and acknowledgements

Special considerations for oncology drugs - why?

• High unmet medical need

• Life-threatening & refractory nature of the disease

• Failure of standard of care; Limited therapeutic options

7

p p

• Patients eligible for phase 1 & 2 trials have not responded to established therapy

Regulatory guidance for oncology drug development

Recent publication of special regulatory guidance reflects the significance of this patient population

8These will be discussed in detail later in the presentation

Special considerationsf l d

9

for oncology drugs

Page 4: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

4The screen versions of these slides have full details of copyright and acknowledgements

Special considerations for oncology drugs

• Phase 1 in patients

• Treatment extension for patients that benefit

S ll th ti i d

10

• Small therapeutic index

• Timing of toxicity studies

Special considerations for oncology drugs

• Phase 1 conducted in patients

– Have not responded to standard of care (SOC)

– Disease progression despite SOC

11

p g p

– Early enrollment of patients → possibility of benefit

• Phase 1a typically single-dose escalation to maximum tolerable dose (MTD)

• Phase 1b typically multiple-doses for up to 3 months to assess benefit

Special considerations for oncology drugs

Treatment can be extended early in drug development for patients that benefit

– Criteria can be set as early as phase 1 to show evidence of benefit to allow treatment beyond timeframe

12

evidence of benefit to allow treatment beyond timeframe supported by toxicology studies

Page 5: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

5The screen versions of these slides have full details of copyright and acknowledgements

Small therapeutic index

• Desired pharmacologic activity is often interference with life-sustaining pathways

Special considerations for oncology drugs

13

• Accepted that toxicities may occur, risk of potential toxicity weighed against poor disease prognosis

Timing & duration of toxicity studies

• Reproductive toxicity studies can be conducted concurrent with phase 3 or not conducted (cytotoxics)

Special considerations for oncology drugs

14

with phase 3, or not conducted (cytotoxics)

• Duration of chronic toxicity studies

– 6 months for biotherapeutics & small molecule cytotoxic

• Carcinogenicity studies not required for registration

Impact on the toxicology program

15

Page 6: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

6The screen versions of these slides have full details of copyright and acknowledgements

Objectives of toxicity testing

• Identification of initial safe starting dose and dose escalation schemes for clinical trials

• Identification of potential target organs of toxicity and reversibility of effect

16

• Identification of safety parameters for clinical monitoring

Impact on toxicology program• Phase 1 conducted in patients

– Disease state can make patients more sensitive to toxicities

– Impact of toxicology data on clinical risk

Identify findings

C f i h j li i l i i

17

Confer with project clinical scientist

Impact of findings on phase 1 monitoring

Inclusion/exclusion criteria

Toxicology data identify hazards to be monitored and considered in risk/benefit decision making

Impact on toxicology programTreatment can be extended early in drug development for patients that benefit

– Longer phase 1-enabling toxicology studies may be needed to support early clinical trials that intend to show benefit

18

Duration of clinical testing must be supported by

Toxicology studies of equivalent duration

12 weeks

Clinical assumptions, e.g.: projected therapeutic dose (10 mg/kg), route & frequency of administration (e.g. IV, weekly), treatment duration (e.g. 12 weeks)

Page 7: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

7The screen versions of these slides have full details of copyright and acknowledgements

Early extension of treatment for patients who benefit

Phase 1bToxicology

Favorable risk/benefit profilecan enable continued treatment beyond the duration supported

by toxicology

19

Extension phase

Protocol-specified benefit criteria met?

Yes

NoStop

treatment

Impact on toxicology program

Small therapeutic index

• Because toxicities are expected to manifest, characterization of the toxicity profile is balanced with animal welfare

• Mortality is not a desired endpoint; Doses tested in toxicology studies should:

20

studies should:

– Define the severely toxic dose in 10% of rodents (STD10) and

– The highest non-severely toxic dose (HNSTD) in non-rodents*

*JJ De George, CH Ahn, PA Andrews, ME Brower, Cancer Chemotherapy and Pharmacology, 1997

Value of a PD marker in oncology

2.

21

Value of a PD marker in oncology

Page 8: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

8The screen versions of these slides have full details of copyright and acknowledgements

Activity, efficacy & toxicity, value of a PD marker

• Shows drug is active

• Can dose to effect in toxicology studies and clinical trials

• Can separate toxicity from activity

22

Can separate toxicity from activity

• PD is a marker of activity not efficacy

– Must be borne out by clinical efficacy data

Value of a PD markerexample 1: Rituxan®

Rituxan® (monoclonal antibody to B-cell CD20 receptor)

PD marker: depletion of B-cells in blood; Monitorable in toxicology studies & patients

23

– Demonstrates relevance of toxicology studies

– Allows separation of active vs. toxic doses

– Supports activity-based first in human (FIH) dose

– Reduces dose-escalations to attainment of active dose

Clinically validated via reduction in tumor volume

Value of a PD markerexample 2: Avastin®

Avastin® (monoclonal antibody to VEGF)

Inhibits growth of new blood vessels

No PD marker

24

– Active dose extrapolated from animal tumor models

– No prospective way to separate active dose from toxic dose

– First in human (FIH) dose a fraction of safe dose determined toxicology studies

VEGF: vascular endothelial growth factor

Page 9: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

9The screen versions of these slides have full details of copyright and acknowledgements

Key similarities & differences

3.

25

in nonclinical safety evaluation for small and large molecules

Key similarities & differences in toxicity evaluation for small and large molecules

26

• Biologics have complex structural and biological activity compared to small molecules

• Drug development plans for small molecules are different from large molecules

Graphic Courtesy Rolf G. Werner (Boehringer Ingelheim) Apr 2008 Presentation

How do small molecules differ from biologics?

Small molecules BiologicsMolecular weight Low, e.g. <500 daltons High e.g. 10-1000 kilodaltonsDose route Oral Parenteral

Species specificity - Species-independent - Species-specificO t t t i it

27

- Off-target toxicities- Species selection based

on metabolism

- On-target toxicity- Pharmacologically drivenspecies selection

Metabolism Yes Internalized and degraded

Typical toxicities Mechanism/structure-based Exaggerated pharmacology

Immunogenicity Not relevant Yes

Page 10: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

10The screen versions of these slides have full details of copyright and acknowledgements

Small molecules Biologics

Development plans Conventional Nonconventional

Regulatory environment Specific guidelines Case-by-case assessment

Drug development plans for small and large molecules are different

28

Regulatory environment Specific guidelines Case by case assessment

Relevant species Toxicology program in 2 species One relevant species

Highest dose tested Maximally tolerated Multiple approaches

More conservative MABEL, NOEL, NOAEL

Basis for clinical starting dose

Less conservative MTD, NOAEL, HNSTD

MTD: maximum tolerated dose, NOEL: no observed effect level, NOAEL: no observed adverse effect level, MABEL: minimal anticipated biological effect level, HNSTD: highest non-severely toxic dose

Preclinical drivers for small molecules• Early stage discovery toxicology

– In silico, in vitro receptor binding, hERG, cell based/in vivo screens to enable lead selection

• Predictivity of models

29

– In vitro/in vivo correlations

– Cross-species comparisons

• Cross functional interactions

– Lead and backup (safety, activity, PK)

• Established impurity qualification

• Appropriate formulations

Lead optimization schemeTox testing

Potency selectivity In silico genotoxicity Ames;In silico hERG

Tier 1 (solubility/chemical stability)Tier 2 (stability)

Cell assays for efficacy

5-in-1 IV PO rat PK

hERG K+In vitro hepatocytes:

Off-target receptors (Iterative)

Activity, Pharm. Sci, DMPK

30Candidate selection

PK (rodent) CYP IC50

hERG IC50; Bacterial mutagenicityhit profile screen; Hepatocyte TC50Rodent target organ Tox (Iterative)

Metabolite activity in vitroIV/PO PK non-rodent

Exploratory rodent single and repeat doseGenetox panel

In vitro safety pharmAcute toxicity/TK

Page 11: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

11The screen versions of these slides have full details of copyright and acknowledgements

Challenges specific to small molecules• Therapeutic index

• Higher attrition rates– Low translatability of nonclinical risks to clinic

– Substantial off-target activity

– Complex ADME issues

31

• Identify “predictable” liabilities earlier– Therapeutic index

– Nature of toxicities (predictable, reversible, monitorable)

– Biomarkers

– Mode of toxicity

– Human relevance

Cytotoxic vs. non-cytotoxic oncology drugs• Classical anti-neoplastic agents (cytotoxic)

– Relatively non-specific MOA

– Narrow therapeutic index

– Regulatory path relatively straightforward

Genotoxicity, carcinogenicity, and reproductive toxicity i d f i i

32

not required for registration

• Non-cytotoxics (cytostatics, immune modulators)– Relatively specific MOA

– Specific targeted inhibition

Relatively non-toxic

Wider therapeutic margin (??)

– Regulatory path novel

More on targeted inhibitors

• Target specific signaling alterations, e.g. kinases

– Only in presence of activating mutations in cancer cells

• Generally less toxic than cytotoxics

• Complex pathway interactions

33

– E.g. feedback, crosstalk

• Emerging field

– Long term toxicities unknown

– Starting clinical dose considerations

– Translation of preclinical safety to clinic?

Page 12: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

12The screen versions of these slides have full details of copyright and acknowledgements

Toxicology plan for small molecules

Target evaluation

Lead ID

Lead optimization

Candidate selection Phase I Phase II Phase III

Post approval

IND Filing NDA Filing

In cerebro

Genotoxicity

Short-term toxicity (up to 4 wks) l d b k i

Mechanistic

In silicogenotoxicity, cardiotoxicity

34

lead or backup screeningLong-term toxicity (up to 2 yrs)

In vivo SP

Reproductive toxicityLocal tolerance

Parenteral drugs only

SP – Safety Pharmacology

Impurity qualification

toxicology

minimal Indication/MOA-specific

Allergy

In vitro SPCardiotoxicity,

off-target receptor binding

Special Tox, immuno-toxicity, phototoxicity

Oncology drug failures

ntag

e of

suc

cess

35Identifying predictable preclinical safety liabilities earlier

can lead to design/selection of better drug candidates

Kola and Landis, Nature Reviews Drug Discovery 3, 711-716 (August 2004)

Per

cen

Current regulatory guidance and impact

4.

36

Current regulatory guidance and impact on drug development for oncology

Page 13: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

13The screen versions of these slides have full details of copyright and acknowledgements

Regulatory guidance for oncology drug development

Recent publication of special regulatory guidance reflects the significance of this patient population

37

Specific regulatory guidancefor oncology drugs

• US FDA: Regulatory Considerations for Preclinical Development of Anticancer Drugs; JJ De George, CH Ahn, PA Andrews, ME Brower, Cancer Chemotherapy and Pharmacology, 1997

• ICH M3(R2): Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals

38

• EMEA (1998); Note for Guidance on the Pre-Clinical Evaluation of Anticancer Medicinal Products. The European Agency for the Evaluation of Medicinal Products, Committee for Proprietary Medicinal Products, January 1998; (CPMP/SWP/997/96)

• MHLW is developing nonclinical guidance to address various mechanisms of anti cancer therapy excluding biologics

• ICH (2008). Preclinical Guideline on Oncology Therapeutic Development; ICH Topic S9 Step 3 has been released for comments in December 2008; ICH Topic S9 Step 3; EMEA/CHMP/ICH/646107/2008 http://www.emea.europa.eu/pdfs/human/ich/64610708en.pdf

IND-enabling toxicology program for oncology drugs

No standalone studies required

Core battery only in case of concern

Safety pharmacology

One species2 speciesRepeat dose studya

YesYesDose-ranging

yesEncouragedToxicokinetics

BiologicsSmall molecules

39

Starting dose basis NOAEL Rodent STD10/non-rodent HNSTD

Yes (monoclonals) No Tissue cross-reactivity

No MaybeGenotoxicityb

a Duration to match clinical regimen b Only if testing normal volunteers or disease-free patientsPrograms should be developed case-by-case based on scientific rationale; the table above is simplified for comparison purposes only

Target evaluation Lead ID

Lead optimization

Candidate selection Phase I Phase II Phase III

Post approval

IND Filing NDA Filing

Page 14: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

14The screen versions of these slides have full details of copyright and acknowledgements

Selecting phase I starting dose:example

• Drug A resulted in 10% (2/20) mortalities in rats at 30 mg/kg in the 28 day study

– Rat STD10 = 30 mg/kg

– Human equivalent dose is 30 x 6 = 180 mg/m2

• Dogs tolerated 1/10th (10 fold safety

40

• Dogs tolerated 1/10th (10-fold safety margin) of this dose = 18 mg/m2

• Phase 1 starting dose = 18 mg/m2

– mg/m2 to mg/kg in humans,

– Use 37 as the conversion factor

– 18 mg/m2 or 18/37 = 0.5 mg/kg

– =~30 mg (60 kg patient)

Source: http://www.fda.gov/cder/cancer/docs/doseflow.pdf

NoneMaybebCarcinogenicity

NoneYesbGenotoxicity

Chronic toxicity

NDA-enabling toxicology program for oncology drugs

One relevant speciesRodent and non-rodentSpecies

No longer than 6 monthsNo longer than 6 months

None13 wks for non-cytotoxicsSub chronic toxicity

BiologicsSmall molecules

41

Reproductive toxicitya,b,c

yg y

NoneNoneSafety pharmacology

aICH stage C-D, or Segment II reproductive toxicity studies evaluate the period from implantation to birth (period of organogenesis); bMay be unnecessary depending on intended patient population (late stage patients); cEmbryofetal toxicity studies needed concurrent with Phase 3; Fertility studies and peri-postnatal studies needed prior to registration

Programs should be developed case-by-case based on scientific rationale; the table above is simplified for comparison purposes only

If cause for concernUseful if targetedSpecial toxicity

YesYes

Target evaluation Lead ID

Lead optimization

Candidate selection Phase I Phase II Phase III

Post approval

IND Filing NDA Filing

Non-clinical testing for drug combinations

• May not be necessary for testing in advanced cancer patients

• May exclude if:

No PK PD or metabolic interactions anticipated

42

– No PK, PD, or metabolic interactions anticipated

– Drugs are not packaged as a combination

– All components well characterized individually

EMEA: guideline on the non-clinical development of fixed combinations of medicinal products (Oct 2005)

FDA: nonclinical safety evaluation of drug or biologic combinations (March 2006)

Page 15: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

15The screen versions of these slides have full details of copyright and acknowledgements

Regulatory guidance impurities

• Impurity thresholds not appropriate for oncology drugs intended to treat advanced stage patients

• Relevant for cytostatics, or drugs likely to be administered to early stage cancer patients with longer life expectancy

• Guidance documents– ICH Q3A(R) impurities in new drug substances, 2002

43

ICH Q3A(R) impurities in new drug substances, 2002

– ICH Q3B(R) impurities in new drug products, 2003

– ICH Q3C impurities: guideline for residual solvents, 1997

– EMEA, guideline on the limits of genotoxic impurities, 2006

– EMEA, questions and answers on the CHMP guideline on the limits of genotoxic impurities, 2008

– Establishment of allowable concentrations of genotoxic impurities in drug substance and product, 2005, PhRMA position paper.

– FDA 2008: Genotoxic and carcinogenic impurities in drug substances and products: recommended approaches and acceptable limits

ICH S9The following is an overview of current expectations and shouldn’t

be used for designing a drug development program; original source documents and guidance should be consulted

• The current guidance addresses three key topics

– Starting dose

44

– Duration of chronic studies

– Reproductive toxicity studies

• Clear guidance for “late stage or advanced cancer”

– Importance of scientific rationale

Source: ICH topic S9 step 3 released for comments in December 2008;ICH topic S9 step 3; EMEA/CHMP/ICH/646107/2008 http://www.emea.europa.eu/pdfs/human/ich/64610708en.pdf

• Approaches to setting the first in human start dose

– Pharmacologically active dose but reasonably safe

– Based on all available PK/PD, toxicity data

– Allometric scaling by body surface area

ICH S9 major topics (1 of 3) start dose

45

g y yfor small molecules; Body weight basis for biologics

– Biologics with agonistic properties, consider minimally anticipated biologic effect level (MABEL)

Page 16: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

16The screen versions of these slides have full details of copyright and acknowledgements

ICH S9 major topics (2 of 3)duration of chronic toxicity studies

• Current approach – 6 month duration

• ICH S9 proposed 3 month Tox studies t b b itt d i t h 3

46

to be submitted prior to phase 3

• Adequate for biologics?

ICH S9 major topics (3 of 3) reproductive toxicology

• Conducted concurrent with phase 3 and submitted for registration, or not conducted (cytotoxics) altogether

– Mechanism of action of the drug on fertility and fetal development

47

• Provision to include fertility endpoints in general tox studies

Thank you for your attention

• The presenters wish to acknowledge Tonita Rios Sapinoso of Genentech for her generous technical support

• We would like to thank Kate Schofield of Henry Stewart Talks for her patience and support

48

• Thank you to Jeanine Bussiere for her encouragement and the invitation to participate

Page 17: Toxicity Testing for Oncology Drugs Theresa Reynolds ... › upload › content › talk › handouts › 1355.pdf · Toxicology data identify hazards to be monitored and considered

Toxicity Testing for Oncology Drugs Theresa Reynolds, Krishna Allamneni

17The screen versions of these slides have full details of copyright and acknowledgements

49