all patients with flt3 mutant aml should receive ... · aml should receive midostaurin-based...

36
All patients with FLT3 mutant AML should receive midostaurin- based induction therapy: of course ( if they are fit for standard induction) Richard M Stone, MD Director, Adult Leukemia Program Dana-Farber Cancer Institute Professor of Medicine Harvard Medical School

Upload: dinhphuc

Post on 08-Apr-2018

218 views

Category:

Documents


5 download

TRANSCRIPT

All patients with FLT3 mutant

AML should receive midostaurin-

based induction therapy: of

course ( if they are fit for

standard induction)

Richard M Stone, MD

Director, Adult Leukemia Program

Dana-Farber Cancer Institute

Professor of Medicine

Harvard Medical School

Disclosures- Richard M. Stone, MD

• Consulting relationships:

– AbbVie; Agios. Amgen, Argenix ( DSMB),

Arog, Astellas, Celgene (includes DSMB and

steering committee), Fujifilm, Janssen; Jazz,

Juno, Karyopharm, Merck, Novartis,

Ono,Orsenix, Sumitomo, Pfizer, Roche

• Securities, employment, promotional

activities, intellectual property, gifts, grants

– None

2

Dana-Farber Cancer Institute in Boston

Dana/Mayer Building

(Office)Brigham and Women’s

Hospital

Yawkey Center

(Clinic)

Litzow MR. Blood. 2005;106:3331-3332.

FLT3 Structure and Activating

Mutations

Over-

expression is

common

25-30%

5-10%

Both mutations cause spont dimerization,

ligand independent growth, and MPD in murine model

Mutant FLT3 AML is a bad disease

Kotarridis PD, et al. Blood. 2001;98:1752-1759.

Background

• Midostaurin (PKC412; N-benzoylstaurosporine) is a potent FLT3 (both ITD and TKD) inhibitor (IC50 <10 nM) (also inhibits VEGFR, PKC, KIT, and PDGFR)1, 2

• Midostaurin specifically inhibits growth of leukemic cell lines made factor independent by transfection of activating FLT3 mutation (ITD or D835Y)2

• Midostaurin increased survival in a murine BMT model ofFLT3 ITD myeloproliferative disorder 3

1. Propper DJ et al. J Clin Oncol. 2001; 19:1485-1492.

2. Weisberg E, et al. Cancer Cell. 2002;1:433-443.

3. Kelly LM, et al. Blood. 2002;99:310-318. 6

Phase II Trial of PKC412:

Clinical Activity (75 mg po TID)

• >50% reduction in BM blasts: 5/20 (25%)

– 2 patients with <5% blasts; 1 on D 28, 1 on D 60*

• >50% reduction in PB blasts: 14/20 (70%)

• 7 (35%) with clinical benefit:

Baseline PB blasts

110K 65K 21K 5K 16K 71K 46K

Best response

0, D 29

.06K, D 42

0, D 50

0.1K, D 22

0, D 15

0, D 57

0, D 51

• Comparable results with imatinib with CML-blast crisis

– 31% HEME RESPONSE (8% CR, 18% RTC, 4% NEL))

* Met CR in BM/PB

except for hypocellular BM. Stone RM, et al. Blood, 2005.Stone et al, Blood, 2005

Study 2104: Single Agent Midostaurin Induces

Blast Reduction But Not CR

Response

75 mg TID FLT3mut

n=20

50 or 100 mg BID FLT3mut

n=35

50 or 100 mg BID

FLT3wt

n=57

Complete response

0/20 0/35 0/57

Partial response 1/201/35

[in 100 mg BID cohort]0/57

50% PB blast or BM reduction

14/20 (70%)

25/35 (71%)

[67% for 50 mg BID & 76% for 100 mg BID]

24/57 (42%)

[50% for 50 mg BID & 33% for 100 mg BID]

• Generally well tolerated

– Nausea/vomiting, diarrhea, and fatigue

– < 10% of patients experienced grade 2 or grade 3 events at doses ≤ 100 mg/day

– Hematologic toxicity was uncommon

– Fischer et al, JCO, 2010

PKC412 plus chemo in newly diagnosed,

previously untreated AML: Treatment Plan

– Induction Chemotherapy

• DNR 60 mg/m2 d1, 2, 3 plus ara-C 100 mg/m2 IVCI d1-7

– Post-remisssion chemotherapy

• ara-C 3 gm/m2 over 3h q 12h d1, 3, and 5 x 3 cycles

– PKC412

• 100 mg po bid begin d1 (simultaneous) OR d8 (sequential) of each cycle

• give continuously during induction and post-CR

Study Induction Scheme by Cohort PKC412 dosed bid

Dauno 60mg/sqm i.v.

ara-C 100mg/sqm c.i.v.

Continuous 100mg*

14-day treatment 100mg*

14-day treatment 50mg+

28-day cycle

7 pts

8 pts

7 pts

8 pts

20 pts

20 pts

21 281471

DDD

*AE rate, dt GI tox

too high

Efficacy

• 80% Complete Response (CR) rate

(32/40)

• 92% of FLT3mut patients had a CR

• Trend toward higher CR in FLT3mut

patients

• No significant difference in response

rates or duration of remission between

the sequential and concomitant schedules

0

10

20

30

40

50

60

70

80

90

100

FLT3wt

(n=27)

FLT3mut

(n=13)

74%

92%

Com

ple

te R

esponse (

%)

Complete Response Rate in Patients with FLT3wt and FLT3mut Blasts

90% CR rate also w

soraf+IA; Ravandi et al

JCO, 2010 Stone, Leukemia, 2012

Similar Survival Seen in Previously Untreated Patients With

FLT3mut and FLT3wt Blasts

FLT3-mutant

FLT3-wild type

FLT3-mutant censored

FLT3-wild type censored

Mutation Med OS, mo (range) Alive

ITD (n=9) 20 (4-56) 3

TKD (n=4) 50 (39-54) 4

1.0

Surv

ival P

robabili

ty

Months After Treatment Start

X X XXXXXXXXX

X X XXX

0.9

0.8

0.7

0.6

0.5

0.4

0.3

0.2

0.1

0.0

0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 54 57 60

13

27

13

27

12

24

12

22

11

21

9

20

9

17

8

14

8

14

8

14

8

14

8

12

6

12

6

11

5

11

5

9

4

5

3

3

1

2

0

1

0

0

Patients at risk

X

X

X

XStone, Leukemia, 2012

CALGB 10603: Prospective Phase III, double-blinded

randomized study of induction and consolidation

+/- Midostaurin (PKC412) in newly diagnosed patients

< 60 years old with FLT3 mutated AML

R

E

G

I

S

T

E

R

R

A

N

D

O

M

I

Z

E

DNR

ARA-C

PKC412

DNR

ARA-C

PLACEBO

HiDAC

PKC412

HiDAC

PLACEBO

PKC412

MAINTENANCE

12 months

PLACEBO

MAINTENANCE

12 months

FLT3

ITD

or

TKD

Not on STUDY:

FLT3 WILD TYPE

X 4

X 4

CR

CR

Study drug is given on Days 8-21 after each course

of chemotherapy, and Days 1-28 (note change) of each 28 day

Maintenance cycle.

Key Eligibility Criteria

• Age 18-60, normal end-organ function

• Documented AML (non-APL)

• FLT3 mutation centrally determined prior to

enrollment

– Assessed at one of 9 academic labs around the world

– Results within 48h

• Up to 5 days of hydroxyurea allowed prior to start of

treatment while awaiting results of mutation analysis

14

Protocol Therapy

• Transplant not specifically mandated

Induction(2nd cycle given based

on d21 marrow)

daunorubicin 60 mg/m2 IVP days 1-3

cytarabine 200 mg/m2/d d 1-7 via IVCI

midostaurin 50 mg po bid days 8-21

or placebo

Consolidation(up to 4 cycles)

cytarabine 3 gm/m2 over 3h q 12h

days 1, 3, and 5

midostaurin 50 mg po bid days 8-21

or placebo

Maintenancemidostaurin 50 mg po bid

or placebo days 1-28 x 12 cycles

Stone R, et al. Blood. 2015;126: Abstract 6.

Patient Characteristics

MIDO

(N = 360)

PBO

(N = 357)P value

Age (years), median (range)47.1

(19.0-59.8)

48.6

(18.0-60.9).27

Gender .045

Female 187 (51.9%) 212 (59.4%)

Male 173 (48.1%) 145 (40.6%)

FLT3 stratification Groups .995

FLT3 TKD (No ITD) 81 (22.5%) 81 (22.7%)

ITD allelic ratio <0.7 (+/- FLT3 TKD) 171 (47.5%) 170 (47.6%)

ITD allelic ratio ≥0.7 (+/- FLT3 TKD) 108 (30.0%) 106 (29.7%)

MIDO, midostaurin

Stone RM, et al. Blood. 2015;126: Abstract 6.

Consort Diagram

Activated May 2008; completed accrual:

Oct 2011 Screened 3279 patients

Total FLT3(+): N = 887 (27% of screened)

Total randomized: N = 717 (81% of FLT3(+))

Midostaurin (MIDO), N = 360 Placebo (PBO), N = 357

Induction 1, N = 355 Induction 1, N = 354

Consolidation, N = 231

Maintenance, N = 120

Consolidation, N = 210

Maintenance, N = 85

Induction 2, N = 81 Induction 2, N = 101

Stone RM, et al. Blood. 2015;126: Abstract 6.

Complete Response Rates

MIDO

(N = 360)

PBO

(N = 357)P*

CR by day 60 212 191

Rate 59% 53% .15

Time to CR, median (range) 35 days (20-60) 35 days (20-60)

CR in induction/consolidation**239 211

Rate 66% 59% .045

Time to CR, median (range) 37 days (20-99) 36 days (20-112)

*2-sided Fisher’s exact P

**Includes all CRs reported within 30 days of ending protocol therapy

Stone RM, et al. Blood. 2015;126: Abstract 6.

All Grade 3-4 Non-Hematologic Events,

reported during treatment in ≥ 10% of patients

MIDO PBO p *

%

(n= 360)

%

(n=357)

Febrile Neutropenia 81% 82% 0.92

Infection 40% 38% 0.49

Diarrhea 15% 16% 1.00

Hypokalemia 13% 17% 0.17

Pain 13% 13% 0.91

Infection - Other 12% 12% 1.00

ALT, SGPT 12% 9% 0.23

Rash/desquamation 13% 8% 0.02

Fatigue (asthenia, lethargy, malaise) 9% 11% 0.53

* 2-sided Fisher’s Exact p

19

Grade 5 events during Treatment

*2-sided Fisher’s Exact p = 1.0

Adverse EventInduction Post Induction

MIDO PBO MIDO PBO

Death NOS 5 3 0 3

Infection 3 6 1 1

Hemorrhage, CNS 1 1 0 1

Pneumonitis 3 0 0 0

Cardiac ischemia 1 0 0 1

Colitis 1 0 1 0

Hypotension 1 0 0 0

Febrile Neutropenia 0 1 0 0

Perforation, GI 0 0 0 1

Potassium serum 0 1 0 0

Renal failure 1 0 0 0

Total 16 12 2 7

Total Deaths: MIDO 18 (5%) vs PBO 19 (5.3%)*

20

Overall Survival (Primary Endpoint)23% Reduced Risk of Death in the MIDO Arm

• Median OS: MIDO 74.7 (31.7-NE); PBO 25.6 (18.6-42.9) monthsNE, not estimable

*Controlled for FLT3 subtype (TKD, ITD-Low, ITD-High)

Arm 4-year Survival

MIDO 51.4% (95%CI: 46, 57)

PBO 44.2% (95%CI: 39, 50)

+ Censor

Hazard Ratio*: 0.77

1-sided log-rank P value*: .0074

Stone RM, et al. Blood. 2015;126: Abstract 6.

Event-Free Survival (Key Secondary Endpoint)

• Event: first of no CR within 60 days, relapse or death

• 4 year EFS rate: MIDO 27.6% vs PBO 20.2%

* controlled for FLT3 subtype (TKD, ITD-Low, ITD-High))

Arm Median (95%CI)

MIDO 8.0 mo (5.1-10.6)

PBO 3.0 mo (1.9-5.9)

+ Censor

1-sided log-rank p-value*: 0.0025

Hazard Ratio*: 0.79

22

Consistent Effect on EFS by FLT3 status

162

341

214

717

0.66

0.85

0.77

0.79

0.44

0.67

0.57

0.66

0.98

1.09

1.04

0.93

0.2 0.4 0.6 0.8 1.0 1.2

FLT3-TKD

FLT3-ITD-Low

FLT3-ITD-High

Overall (strat)

N HR LL UL

Favors Treatment Favors Placebo

23

Confidence intervals are two-sided; p-values are one-sided.

HR: Hazard Ratio, LL: Lower Limit; UL: Upper Limit;

strat: stratified by FLT3 status; ITD-low: Allelic ratio < 0.7; ITD-high: Allelic ratio ≥ 0.7

Overall (strat)

p=0.003

FLT3-ITD-High

p=0.04

FLT3-ITD-Low

p=0.10

FLT3-TKD

p=0.02

Disease-Free Survival CR in Induction/Consolidation

• 4 year DFS rate: MIDO 46.4% vs. PBO 37.4%

• Event: first of relapse or death among CRNE: not estimable

* controlled for FLT3 subtype (TKD, ITD-Low, ITD-High).

211 111 82 72 51 17 2Placebo-239 163 117 103 73 30 7Midostaurin-

number at risk

0 12 24 36 48 60 72 80

time (months)

0

10

20

30

40

50

60

70

80

90

100

% d

ise

as

e f

ree

Censor

14.4 (11.0-22.2)Placebo

25.9 (19.4-NE)Midostaurin

Median (95% CI)Arm

0 12 24 36 48 60 72 80

time (months)

0

10

20

30

40

50

60

70

80

90

100

% d

ise

as

e f

ree

Censor

14.4 (11.0-22.2)Placebo

25.9 (19.4-NE)Midostaurin

Median (95% CI)Arm

1-sided log-rank p-value*: 0.002

Hazard Ratio*: 0.70

Arm Median (95%CI)

MIDO 25.9 mo (19.4, NE)

PBO 14.4 mo (11.0, 22.2)

+ Censor

24

Overall Survival: Post-TransplantTreatment With MIDO Increases OS After SCT in CR1

+ Censor

SCT in CR1

HR 0.61

SCT outside CR1

HR 0.98

Midostaurin Placebo

Stone RM, et al. Blood. 2015;126: Abstract 6.

Conclusions

• Midostaurin, a mult-itargeted kinase inhibitor,

improves OS when added to standard chemo

with one year maintenance in newly diagnosed

pts aged 18-60 with ITD and TKD FLT3 mutant

AML, and represents a new standard of care

• OS and EFS benefit was consistent in uncensored

as well as censored analyses, despite high SCT rate

• Safety profile similar in each arm

• An international academic-industry collaborative

AML study based on genotype at dx is feasible

Stone R, et al. Blood. 2015;126: Abstract 6.

* Patients may receive hydroxyurea during screening phase

** Optional 2nd cycle in patients achieving PR after cycle I

*** Cytarabine: 18-65 years, 3g/m², q12hr, day 1,3,5; >65 years, 1g/m², q12hr, day 1,3,5;

optional for patients before allogeneic HSCT

Daunorubicin

Cytarabine**

High-Dose

Cytarabine***

FLT3

Mutation

Screening

Within

48 Hours*

1-yr Maintenance

3x High-Dose

CytarabinePKC412

PKC412

1-yr MaintenanceStart: Day 30 after allo HSCT

1st priority

Allogeneic

HSCT

PKC412

2nd priority

AMLSG 16-10 (Clinicaltrials.gov identifier, NCT01477606; EudraCT Number, 2011-003168-63 )

Study Design

Schlenk RF, et al. Blood. 2015;126: Abstract 322.

AML With FLT3-ITDAMLSG 16-10 Compared to Historical Controls

Age 18-<60 yrs

AMLSG 16-10 n = 79

Historical-control AMLSG

N = 481

Age 60-70 yrs

AMLSG 16-10 n = 37

Historical-control AMLSG

N = 97

P = .014 P = .036

Time (Months)

0 6 12 18 24 30 36

Time (Months)

Rela

pse-F

ree S

urv

iva

l (%

)

0 6 12 18 24 30 36

0

25

50

75

100

Rela

pse-F

ree S

urv

iva

l (%

)

0

25

50

75

100

Schlenk RF, et al. Blood. 2015;126: Abstract 322.

Midostaurin in AML: FAQs

– When will the drug be approved?• I don’t know; ask Novartis

– What will the approval look like?• Untreated mutant FLT3 age to 60, ? higher

– When will C10603/RATIFY be published• Second revision under review

– Is there enough data to justify adding midostaurin to all newly diagnosed mutant FLT3 AML pts who can receive 3+7?

•YES

Midostaurin in AML: Final Points

– Midostaurin plus chemo leads to a lower death rate than chemo alone in initial rx of mutant FLT3 AML

• Is benefit due to FLT3 inhibition at all or in part?

• Chemo +/- mido in upfront FLT 3 WT AML trial planned

– Will also be approved in aggressive systemic mastocytosis

– SWOG trial will test azacitidine +/- mido in chemo unfit older AML

Many thought C10603 would be negative

Many thought the Pats would lose when down by

25 in third quarter

Acknowledgements

• DFCI Adult Leukemia Team

– MDs: DeAngelo, Garcia, Steensma, Wadleigh. Luskin, Winer

– MD Scientists: Abel, DeCaprio, Ebert, Frank, Griffin, Lane,

Letai, Lindsley, Weinstock

– Midlevel practitioners: Galinsky, Cahill, Edmonds, Gerard,

Penicaud

– Research RN Toomey-Matthews, CRCs, administrative

support

• Other Key Local Colleagues

– DFCI SCT: Alyea, Antin, Cutler, Ho, Koreth, Soiffer

– DFHCC ( MGH) : Amrein, Fathi, Graubert, Hobbs

– DFHCC ( BIDMC): Avigan, Rosenblatt

• National and International (partial list) ( Novartis)

– Dohner, Fischer, Larson, Marcucci, Schiffer, CTEP/NCI 32

Design considerations

• Primary Endpoint: Overall Survival, not censoring for transplantation

• Sample size: 714 evaluable patients

– HR = 0.78 for OS

– Power = 84%

– Type I error rate= 0.025 (1-sided)

– Final analysis after 509 events (deaths) observed

– Interim analysis conducted at 50% of events

33

Revised Analysis Plan

• Alliance DSMB and CTEP approved the primary analysis of OS

to occur with a cut off of April 1, 2015 with 357 events

• Event rate reached a plateau: 6 events 2014, 3 in 2015.

– 509 events predicted to occur in 2025

• Higher than expected transplant rate: 25% in CR 1,

57% overall

• increased TKD incidence: 23%

• Median follow-up time for survivors: 56.7 mo

(range: 0.1, 79.2 mo)

• Critical value to declare statistical significance: 0.0239

(1-sided).

34

Event-Free SurvivalCR in Induction/Consolidation

MIDO PBO

1-sided

Log-rank

p-value*

Hazard Ratio

MIDO vs. PBO

(95%CI)*

N 360 357

0.00020.73

(0.61, 0.87)

# of events 242 271

Median (mo)

(95% CI)

11.3

(8.4, 15.1)

6.1

(4.7, 7.5)

*controlled for FLT3 subtype (TKD, ITD-Low, ITD-High).

• Event: first of either no CR in induction/consolidation, or

relapse or death

35

FLT 3 inhibitors in prior studies

Lestaurtinib(CEP-701)

Midostaurin(PKC-412)

Sorafenib Quizartinib(AC220)

a – Molm-14 cells incubated in RPMI/10% FBS b - Molm-14 cells incubated in plasmaPratz et al. Blood 2010;115(7):1425-32 Human kinome image generated using TREEspot™ software tool and reprinted with permission from KINOMEscan™, a division of DiscoveRx Co. 36