Histologic Distinctions in the Management of Non-Small ... · PDF fileManagement of Non-Small...

26
1/28/14 1 A Collabora)ve Prac)ce Approach to Managing Pa)ents With Advanced NonSmall Cell Lung Cancer Clarifying the Selec)on of Newer Treatment Op)ons Corey Langer, MD, FACP Beth Eaby-Sandy, CRNP, OCN ® Abramson Cancer Center University of Pennsylvania Histologic Distinctions in the Management of Non-Small Cell Lung Cancer Corey Langer, MD, FACP Disclosure Dr. Langer discloses the following: Grant/Research: Genentech, Merck, GSK, Nektar, Daiichi-Sankyo Scientific Advisor: Novartis, Genentech, Bayer/Onyx, Abraxis, Abbott, Biodesix, Clarient, Caris Dx, Vertex, Synta, Celgene, Boehringer- Ingelheim DSMC: Eli Lilly, Amgen, Synta, Peregrine

Transcript of Histologic Distinctions in the Management of Non-Small ... · PDF fileManagement of Non-Small...

1/28/14

1

A  Collabora)ve  Prac)ce  Approach  to  Managing  Pa)ents  With  Advanced  

Non-­‐Small  Cell  Lung  Cancer  Clarifying  the  Selec)on  of  Newer  Treatment  Op)ons  

Corey Langer, MD, FACP Beth Eaby-Sandy, CRNP, OCN®

Abramson Cancer Center University of Pennsylvania

Histologic Distinctions in the Management of Non-Small Cell Lung

Cancer

Corey Langer, MD, FACP

Disclosure

Dr. Langer discloses the following:

Grant/Research: Genentech, Merck, GSK, Nektar, Daiichi-Sankyo Scientific Advisor: Novartis, Genentech, Bayer/Onyx, Abraxis, Abbott,

Biodesix, Clarient, Caris Dx, Vertex, Synta, Celgene, Boehringer-Ingelheim

DSMC: Eli Lilly, Amgen, Synta, Peregrine

1/28/14

2

Therapeutic Implications Histologic Prism

•  Tendency over past 25 years to “lump” non-small cell lung cancer (NSCLC) histologies – Little difference in treatment outcome

• Therapeutic empiricism –  Increasing NOS designation

•  Insufficient tissue • Pathologic “laziness”

ECOG 1594: Overall Survival by Treatment Group

•   1,207  pa)ents,  stage  IIIB/IV  (15/85%),  PS  0–2,  median  age  63,    M/F  (64%/36%)  

Schiller, J. H., et al. (2002). N Engl J Med, 346(2), 92-98.

Personalized Therapy

•  Previous “one size fits all” approach •  Eclipsed by “personalized” or “patient-specific”

treatment – Histology – Molecular markers

1/28/14

3

NSCLC

•  Adenocarcinoma – Glandular pattern – Mucin positivity (50%) – CK7+/CK20- – TTF-1+ (75%)

•  Squamous cell carcinoma – Cellular keratinization – Intercellular bridges – Keratin “pearl” formation – CK7-/CK20- – TTF-1 neg – P63+ p40+ CK5/6+

Common, but not 100%

WHO

Common, but not 100%

WHO

Emergence of Histology as Determinant of Therapy

•  Sandler: Paclitaxel-carboplatin +/- bevacizumab •  Scagliotti: Gem-DDP vs. pem-DDP •  Socinski: Nab-paclitaxel–carboplatin vs.

paclitaxel-carboplatin

ECOG 4599: Phase III Trial of Bevacizumab in Non-Squamous NSCLC

Sandler, A., et al. (2006). N Engl J Med, 355, 2542-2550.

Parameter PC PCB p value

RR (%) 15 35 <.001

PFS (mo) 4.5 6.2 <.001

Median survival (mo) 10.3 12.3 p = .003 1-year survival (%) 44 51

2-year survival (%) 15 23

PC  Paclitaxel  200  mg/m2    Carbopla)n  AUC  6  mg/m2  q3wk  Eligibility  

•  Non-­‐squamous  NSCLC  •  No  Hx  of  hemoptysis  •  No  CNS  metastases  

Stra)fica)on  variables    RT  vs.  no  RT    Stage  IIIB  or  IV  vs.  recurrent    Wt  loss  <  5%  vs.  ≥  5%    Measurable  vs.  nonmeasurable  

No crossover to bevacizumab permitted

RT = radiotherapy PD = progressive disease

PCB  Paclitaxel  x  6  cycles  +    bevacizumab    (15  mg/kg  q3wk)  to  PD  

1/28/14

4

ECOG 4599 Adverse Events

*Pulmonary  hemorrhagic  events  are  included  in  the  hemorrhagic  events.    **p  <  .05    

Patients, %

Event PC

(n = 441) B/PC

(n = 427) GI perforation 0 0.9 Hemorrhage* (grade 3-5)1 1.1 4.7** Pulmonary hemorrhage2 0.5 2.3 Arterial thromboembolic events 1.4 3 Hypertension (grade 3/4) 0.7 8** Neutropenia and infection Neutropenia 17 27** Infection without neutropenia 3 7 Infection with neutropenia (grade 3/4) 2 4 Febrile neutropenia 2 5**

Treatment-related deaths (actual numbers) 2 15**

1Sandler, A. B., et al. (2006). N Engl J Med, 355, 2542-2550. 2Bevacizumab prescribing information, Genentech, 2008.

Advanced Stage IIIB/IV NSCLC First-Line Subsets

•  Bevacizumab-ineligible (~60%-70%) – Squamous histology – Brain metastases – Ongoing anticoagulation – Antecedent hemoptysis – Poor PS (2+)

•  Bevacizumab-eligible (~20%-30%) – Non-squamous

•  EGFR mutants (~10%) Socinski, M., et al. (2009). J Clin Oncol, 27, 5255-5261; Reck, M., et al. (2009). J Clin Oncol, 27, 1227-1234.

PASSPORT Study: Bevacizumab in Patients With Treated Brain Metastasis

Socinski, M., et al. (2009). J Clin Oncol. 27, 5255-5261.

1/28/14

5

Median Survival (mo) 3 age cut-points  

Age Cohort   < 45 yr   45-59 yr   ≥ 60 yr  Women control   5.8   12.5   13.8  Women + BEV   16.8   15.5   12.8  Men control   3.4   9.5   8.5  Men + BEV   12.6   12.3   11.0  

Wakelee, H., et al. (2012). Lung Cancer, 76, 410-415.

Conclusion: The only group that failed to have a conclusive benefit was women over 60.

Median  Survival  by  Sex  and  Age  With  or  Without  Bevacizumab  on  E4599  

Overall Survival in the Pooled Population of E4599 and POINT-BREAK

Unadjusted Kaplan-Meier estimates for OS among patients (A) < 75 years and (B) ≥ 75 years receiving PC + bevacizumab in the pooled population of E4599 and PointBreak relative to patients receiving PC alone in E4599

Langer, C., et al.,15th World Conference on Lung Cancer (WCLC), Sydney, 2013.

Pemetrexed and Histology: The Scagliotti Study

1/28/14

6

Study Design

Gemcitabine  1250  mg/m2    days  1  +  8    Cispla)n  75  mg/m2  day  1  

Stage  IIIB/IV  NSCLC  

PS  0  -­‐  1  

No  prior  chemo  

Randomiza)on:  gender,  PS,  stage,  histo  vs  cyto  dx,  brain  mets  

Pemetrexed  500  mg/m2  +    Cispla)n    75  mg/m2  day  1  

Primary  objec)ve:  Overall  survival  

15%  Non-­‐inferiority  margin  (HR  1.17)  

N  =  1,700  pa)ents,  power  80%  

B12, folate, and dexamethasone given in both arms

Scagliotti, G. V., et al. (2008). J Clin Oncol, 26(21), 3543-3551.

Overall Survival: All Patients Cisplatin + Gemcitabine vs. Cisplatin + Pemetrexed

1.176 was non-inferiority margin Scagliotti, G. V., et al. (2008). J Clin Oncol, 26, 3543-3551.

months

Overall Survival in Patients With Nonsquamous Histology

Scagliotti, G. V., et al. (2008). J Clin Oncol, 26, 3543-3551. months

1/28/14

7

What  Op)ons  Exist  for  NSCLC  With  Squamous  Histology?  

•  Gemcitabine •  Nab-paclitaxel •  Cetuximab •  Necitumumab •  Ipilimumab

Overall Survival in Patients With Squamous Cell Carcinoma

months Scagliotti, G. V., et al. (2008). J Clin Oncol, 26, 3543-3551.

Abstract 7511 Phase III Trial: Nab-paclitaxel–Carbo vs Carbo-Paclitaxel

Chemo-naive NSCLC

IIIB/IV

ECOG PS 0-1

Baseline peripheral neuropathy ≥ grade 2

N = 1,050

Nab-paclitaxel 100 mg/m2 d1, 8, 15 Carbo AUC 6 d1 No premeds

Paclitaxel 200 mg/m2 d1 Carbo AUC 6 d1 Premeds: dex, antihistamines

RANDOM I ZED

•  Stratification factors: stage IIIb vs. IV, age < 70 or ≥ 70, gender, histology (SCC vs. non-SCC), geography •  Primary endpoint: ORR •  Secondary endpoint: PFS, OS, DCR, safety (NCI CTCAE v3)

Socinski, M., et al. (2011). ASCO Abstract 7551; Socinski, M., et al. (2012). J Clin Oncol, 30, 2055-2062.

1/28/14

8

Res

pons

e R

ate

(%)

Objective Responses by Histology*

p < .001 RR =1.680

p =.808 RR=1.034

* Not a pre-specified subgroup analysis

41%  

26%  24%   25%  

0  

10  

20  

30  

40  

50   Ab-­‐P/C  P/C  

Interac)on  p  value  for  histology:    .036  

Squamous Histology

Non-Squamous Histology

Socinski, M., et al. (2011). ASCO Abstract 7551; Socinski, M., et al. (2012). J Clin Oncol, 30, 2055-2062.

PFS: ITT Population

Prop

ortio

n N

ot P

rogr

esse

d

Months

Pt at risk Ab-P P

521 531

330 321

167 162

86 75

38 48

23 19

10 10

4 4

0 2

0 1

0 0

Ab-P/carboplatin (N=521) paclitaxel/carboplatin (N=531)

0 3 6 9 12 15 18 21 24 27 30 33 0.00

0.25

0.50

0.75

1.00 Ab-P/ Carbo  

Paclitaxel/ Carbo   HR  

P value  

N/Events   521/297   531/312  

Median PFS (mo)*  

6.3   5.8   0.902   0.214  

95% CI   5.6-7.0   5.6-6.7   0.767-1.060  

* PFS based on independent assessment

Overall Survival: ITT Population

Prob

abili

ty o

f Sur

viva

l

Months

Pt at risk Ab-P Pac

521 531

469 470

381 389

313 308

246 243

200 191

163 148

98 89

23 24

0 5

0 1

Ab-P/carboplatin (N=521) Paclitaxel/carboplatin (N=531)

0 3 6 9 12 15 18 21 24 27 30 33 0.00

0.25

0.50

0.75

1.00

0 0

Ab-P/Carbo  

Paclitaxel/ Carbo   HR   P value  

N/Events   521/360   531/384  

Median OS (mo)   12.1   11.2   0.922   0.271  

95% CI   10.8-12.9   10.3-12.6   0.797-1.066  

1/28/14

9

Secondary Endpoint: OS

Median OS (mo) Events / N HR ab-P/C P/C

744 / 1052

86 / 149 521 / 724 127 / 165 589 / 789 155 / 263 639 / 896 105 / 156 343 / 450 401 / 602 142 / 218 602 / 834

0.922 0.950 1.019 0.622 0.894 0.995 0.999 0.583 0.890 0.950 0.896 0.917

12.1 16.7 11.0 12.7 11.4 16.8 11.4 19.9 10.7 13.1 12.4 12.0

11.2 17.2 11.1 9.8

10.0 16.0 11.3 10.4 9.5

13.0 13.6 11.0

Favors ab-P/C

Socinski, M., et al. (2011). ASCO Abstract 7551; Socinski, M., et al. (2012). J Clin Oncol, 30, 2055-2062.

Paclitaxel + Carboplatin Show Significant Benefit in Pts ≥ 70 y/o With Advanced NSCLC

Phase III study in 451 patients 70-89 yo •  Arm A: Carboplatin AUC 6 every 4 weeks + paclitaxel 90 mg/m² (d1,8,15) Q 4wk vs •  Arm B: Single agent gemcitabine 1150 mg/m² or vinorelbine 30 mg/m², d1, d8

Conclusions: Paclitaxel + carboplatin provides a significantly longer survival in elderly patients with advanced NSCLC than current standard single-agent therapy, with acceptable toxicity

Parameter Arm A

Carbo + Pacl Arm B

Gem + Vin Median OS, mo 10.4 6.2 1 yr OS % 44 25 Median PFS, mo 6.3 3.2 Grade 3-4 hematologic tox 54.1% 17.9%

Quoix, et al. (2010). J Clin Oncol, 28(suppl), ASCO abstract; Quoix, et al. (2011). Lancet, 378, 1079-1088.

Chemotherapy-­‐naive  advanced  NSCLC  

Vinorelbine  25  mg/m2  d1,8  

+  cispla)n  80  mg/m2  d1  q3wk  

Phase III FLEX: Vinorelbine/Cisplatin ± Cetuximab in 1st-Line Advanced NSCLC

Primary endpoint: OS

Secondary endpoints: PFS, ORR, DCR, QoL, safety

n=557  

n=568  

Cetuximab  400  mg/m2  d1  wk1,  then  250  mg/m2  qwk  

 +  vinorelbine  25  mg/m2  d1,8  

+  cispla)n  80  mg/m2  d1  q3wk  

RANDOMIZE

Up to 6 cycles of chemotherapy; patients not progressing continue on cetuximab maintenance

Pirker, R. et al. (2008). ASCO, oral presentation and abstract 3; Pirker, R., et al. (2009). Lancet, 373(9674), 1525.

Stra)fied  by    IIIB  or  IV    ECOG  PS  0,1  

or  2  

1/28/14

10

*Including: large cell, adenosquamous, and undifferentiated carcinoma

Characteristic CT + Cetuximab (n=557)

CT (n=568)

Male/female, % 69/31 71/29 Median age, years (range) ≥ 65 years, %

59 (18–78) 31

60 (20–83) 32

ECOG PS, % 0 1 2

24 60 17

21 60 18

Stage, % IIIB wet IV

6 94

6 94

Histology, % Adenocarcinoma Squamous cell carcinoma Other*

46 34 20

49 33 18

Never smoker, % 22 22

Ethnicity, % Caucasian Asian Other

84 11 5

85 10 5

FLEX Patient baseline characteristics

Pirker, R. et al. (2008). ASCO, oral presentation and abstract 3; Pirker, R., et al. (2009). Lancet, 373(9674), 1525.

FLEX Overall Survival

Months  Pa)ents  at  risk  CT  +  Cetuximab  557  383  251  155  53  3  CT  568  383  225  134  48  0  

Overall  Survival  (%

)  

p value = stratified log-rank test (2-sided)

Median OS 1-year survival

▬ CT + Cetuximab (n=557) 11.3 months 47 %

▬ CT (n=568) 10.1 months 42 %

HR=0.871 (95% CI 0.762–0.996), p=.044

Pirker, R. et al. (2008). ASCO, oral presentation and abstract 3; Pirker, R., et al. (2009). Lancet, 373(9674), 1525.

Median OS (months)

CT + Cetuximab CT HR [95% CI] p value

All (n=1125) 11.3 10.1 0.871 [0.762–0.996]

.044

Caucasian (n=946)

Adenoca. (n=413)

SCC (n=347)

10.5 9.1 0.803 [0.694–0.928]

.003

12.0 10.3 0.815 [0.649–1.023]

.077

10.2 8.9 0.794 [0.626–1.007]

.057

Asian (n=121) 17.6 20.4 1.179 [0.730–1.905]

.499

Pirker, R. et al. (2008). ASCO, oral presentation and abstract 3; Pirker, R., et al. (2009). Lancet, 373(9674), 1525.

FLEX:  OS  by  Subgroups    Ethnic  Origin  and  Histology  (ITT)  

1/28/14

11

High  and  Low  EGFR  Expression  Low EGFR expression IHC score < 200

High EGFR expression IHC score ≥ 200

IHC score=270 IHC score=30 IHC score=0*

IHC, immunohistochemistry; *IHC score=7 for tumor overall

O’Byrne, K., et al. (2010). JTO, 12(suppl), S558 (LBOA1).

FLEX H Score Elevated Cohort (> 200)

Arm   C225   Control  Number   178   176  OR%   44*   28  PFS (mo)   5.0   4.4  Med survival (mo)   12**   9.6  1 yr OS%   50   37  2 yr OS%   24   15  

* p = .002 **HR = 0.73, p = .011

O’Byrne, K., et al. (2010). JTO, 12(suppl), S558 (LBOA1).

Elevated Cohort (> 200) Breakdown by Histology

C225   Control  Adenocarcinoma *  MS (mo)   20.2   13.3  Arm   65   52  Squamous cell ca**  MS (mo)   11.2   8.9  1 yr OS %   45   25  

*HR = 0.72 **HR = 0.62

O’Byrne, K., et al. (2010). JTO, 12(suppl), S558 (LBOA1).

1/28/14

12

Perspectives on First-Line Therapy Based on Histology

•  Based on E4599 and Scagliotti trials, use of Pemetrexed and Bevacizumab is limited to NON-squamous histology

•  Standard cytotoxic platform for Adenocarcinoma – Platinum plus either pemetrexed or taxane

•  Standard platform for Squamous cell carcinoma – Platinum plus either gemcitabine or taxane

Histologic Distinctions in the Treatment of NSCLC: Conclusions

•  Prognosis is generally inferior in squamous vs. non-squamous NSCLC

•  Gemcitabine appears superior to pemetrexed in the first-line setting in squamous NSCLC (in combination with cisplatin), while pemetrexed appears superior in non-squamous cell carcinoma

•  It is NOT safe to use bevacizumab in squamous NSCLC patients, its use should be limited to patients with non-squamous histology

Molecular Distinctions in the Management of

Non-Small Cell Lung Cancer

1/28/14

13

Molecular Determinants of Treatment in NSCLC

•  EGFR mutation •  EML4/ALK •  ROS-1 •  KRAS (?) •  BRAF •  cMET •  ERCC1 and RRM1

Ac)onable    

Possibly  Ac)onable    

An Evolving View of Adenocarcinoma Emergence of Molecular Markers

KRAS    

2000  

Pending  

EGFR    BRAF  

PIK3CA    

EML4-­‐ALK  

ROS1    HER2    

2013  

Lung Cancer Mutation Consortium Patients and Study Plan

1000 patients Stage IV ECOG PS 0-2 Lung Adenocarcinoma Sufficient Tissue (Paraffin) Informed Consent

Central Confirmation of Adenocarcinoma Diagnosis (1 slide)

Mutational Analysis CLIA-Certified lab at LCMC site: KRAS, EGFR, EML4-ALK, BRAF, HER2, PIK3CA, NRAS, MEK1, AKT1, MET amplification

Use Data to Select Therapy (Erlotinib with EGFR mutation)

Report to Physician

Report to LCMC Virtual Database

Recommend Clinical Trial of Agent Specific for Target NCI’s Lung Cancer Mutation Consortium (LCMC)

Kris, M. G., et al. (2011). J Clin Oncol, 29(18 suppl), Abstract CRA7506.

1/28/14

14

Incidence  of  Muta)ons  Detected  

Muta)on  found  in  54%  (280/516)  of  tumors  completely  tested    (CI  50-­‐59%)  

Johnson  et  al  on  behalf  of  LCMC  inves)gators,  WLCC  July  2011  Abstract  O16.01.  Kris  et  al  on  behalf  of  LCMC  inves)gators,  ASCO  June  2011  Abstract  #CRA7506.  

Biomarkers (BM) France: Results Biomarkers Assessment (n=9911)

EGFR act mut 9.5% EGFR res mut

0.8%

HER2 mut 0.9%

KRAS mut 27%

BRAF mut 1.7%

PI3K mut 2.6%

ALK rearrangement

3.7%

UKN/Other 53.8%

Results expressed in % on available analyses

Barlesi , F., et al. (2013). J Clin Oncol, 31(suppl), abstract 8000.

Results: Biomarkers by Smoking Status (n=9911*)

33.2

3.1 9.6

1.8 9.7 3.6

3.8

35.2

Never smokers

EGFR activ EGFR resist KRAS BRAF ALK PI3K HER2 UNK

4.2 0.3 31.7

1.6 3.5 1.7 0.2

56.8

Smokers

* Including 2664 with full clinical data available at the time of this analysis.

Barlesi , F., et al. (2013). J Clin Oncol, 31(suppl), abstract 8000.

1/28/14

15

Detecting EGFR Mutations in NSCLC

•  NSCLC patients with the somatic activation mutation of EGFR are hyper-responsive to EGFR TKIs

•  Most common NSCLC-associated EGFR mutations: – In-frame deletion in exon 19 (E746-A750del) – Point mutation in exon 21 (L858R)

•  These account for 85-90% of EGFR mutations •  Most other mutations, especially those on exon 20,

are associated with resistance to EGFR TKIs

Yu, et al. (2008). Molecular Markers Meeting, Abstract No 64.

6 FEB 2002 11 FEB 2002

NSCLC Pt Treated with an EGFR Inhibitor

IPASS Study Design

Gefitinib (250 mg/day)

N=609

Carboplatin (AUC 5 or 6) / paclitaxel (200 mg/m2) 3 weekly#

n=608

1:1 randomization

Never smokers = <100 cigarettes in lifetime; light ex-smokers = stopped ≥15 years ago and smoked ≤10 pack-years; #limited to a maximum of 6 cycles; carboplatin / paclitaxel was offered to gefitinib patients upon progression PS = performance status; EGFR = epidermal growth factor receptor.

Pa)ents  • Chemonaive  

• Age  ≥  18  years    

• Adenocarcinoma  histology  • Never  or  light  ex-­‐smokers*  

• Life  expectancy  ≥  12  weeks  

• PS  0-­‐2  

• Measurable  stage  IIIB  /  IV  disease  

Primary  •   Progression-­‐free  survival  (non-­‐inferiority)  

Secondary  •   Objec)ve  response  rate  •   Overall  survival    •   Quality  of  life  •   Disease-­‐related  symptoms    •   Safety  and  tolerability  

Exploratory  •   Biomarkers  

•   EGFR  muta)on  •   EGFR-­‐gene-­‐copy  number  •   EGFR  protein  expression  

•     

Endpoints

Mok, et al. (2009). N Engl J Med, 361, 947-957.

1/28/14

16

Progression-Free Survival in ITT Population

609 453 (74.4%)

608 497 (81.7%)

N Events

HR (95% CI) = 0.741 (0.651, 0.845) p<0.0001

Gefitinib

Primary Cox analysis with covariates HR <1 implies a lower risk of progression on gefitinib

Carboplatin / paclitaxel

Carboplatin / paclitaxel

Gefitinib

Median PFS (months) 4 months progression-free 6 months progression-free 12 months progression-free

5.7 61% 48% 25%

5.8 74% 48% 7%

609 212 76 24 5 0 608 118 22 3 1 0

363 412

0 4 8 12 16 20 24 Months

0.0

0.2

0.4

0.6

0.8

1.0 Probability of PFS0

At risk :

Mok, et al. (2009). N Engl J Med, 361, 947-957.

Progression-Free Survival in EGFR Mutation Positive and Negative Patients (n=437)

EGFR mutation positive EGFR mutation negative

Treatment by subgroup interaction test, p < .0001

HR (95% CI) = 0.48 (0.36, 0.64) p<0.0001 No. events gefitinib, 97 (73.5%) No. events C / P, 111 (86.0%)

Gefitinib (n=132) Carboplatin / paclitaxel (n=129)

ITT population – Mutation rate ~60% Cox analysis with covariates

HR (95% CI) = 2.85 (2.05, 3.98) p<0.0001 No. events gefitinib , 88 (96.7%) No. events C / P, 70 (82.4%)

0 4 8 12 16 20 24 0.0

0.2

0.4

0.6

0.8

1.0

Pro

babi

lity

of p

rogr

essi

on-fr

ee s

urvi

val

0 4 8 12 16 20 24 0.0

0.2

0.4

0.6

0.8

1.0

Pro

babi

lity

of p

rogr

essi

on-fr

ee s

urvi

val

Gefitinib (n=91) Carboplatin / paclitaxel (n=85)

Months Months

Mok, et al. (2009). N Engl J Med, 361, 947-957.

Objective Response Rate in EGFR Mutation Positive and Negative Patients

Gefitinib Carboplatin / paclitaxel

EGFR M+ odds ratio (95% CI) = 2.75 (1.65, 4.60), p=0.0001

EGFR M- odds ratio (95% CI) = 0.04 (0.01, 0.27), p=0.0013

Overall response rate (%)

(n=132) (n=129) (n=91) (n=85)

Odds ratio >1 implies greater chance of response on gefitinib

71.2%

47.3%

1.1%

23.5%

Mok, et al. (2009). N Engl J Med, 361, 947-957.

1/28/14

17

Impact on QOL

Mok, et al. (2009). N Engl J Med, 361, 947-957.

Gefitinib vs Chemotherapy in EGFR Mutated Tumors: A Prospective Study

Maemondo, et al. (2010). N Engl J Med, 362(25), 2380-2388.  

Gefitinib

N=160

CBDCA + PAC n=169

Randomization balanced for institution, sex, stage

           Pa)ents  NSCLC  with  sensi)ve  EGFR  muta)ons  • Stage  IIIB  ,  IV  

• No  prior  chemo  

• PS  0-­‐1  

• Age  20-­‐75  y/o  

Primary  •   Progression-­‐free  survival  

Secondary  • Overall  survival    •   Objec)ve  response  rate  • Safety  and  tolerability  •   Quality  of  life  

•     

Endpoints

Efficacy

Gefitinib Chemotherapy p value

N 98 100

Response rate 73.7% 30.7% <0.001

Median PFS 10.8 m 5.4 m <0.001

Median OS (mo) 30.5 23.8 0.31

Maemondo, et al. (2010). N Engl J Med, 362(25), 2380-2388.

1/28/14

18

The 7503—EURTAC Trial: Erlotinib in First-Line Therapy

Primary endpoint •  Progression-free survival (PFS)

–  interim analysis planned at 88 events Secondary endpoints •  Objective response rate •  Overall survival (OS) •  Location of progression •  Safety •  EGFR mutation analysis in serum •  Quality of life

*Cisplatin 75mg/m2 d1 / docetaxel 75mg/m2 d1; cisplatin 75mg/m2 d1 / gemcitabine 1250mg/m2 d1,8; carboplatin AUC6 d1 / docetaxel 75mg/m2 d1; carboplatin AUC5 d1 / gemcitabine 1000mg/m2 d1,8

Patients - Chemonaive - Stage IIIB/IV NSCLC -EGFR exon 19 deletion or exon 21 L858R mutation - PS 0–2 (n=174)

R

Platinum-based doublet chemotherapy q3wk x 4 cycles*

PD

Erlotinib 150 mg/day PD

Stratification •  Mutation type •  ECOG PS (0 vs 1 vs 2)

*1,139 patients screened

Rosell, R., et al. (2012). Lancet Oncol, 13, 239-246.

Best  Response  Erlo)nib  N=86  

Chemotherapy  N=87  

ORR   58%   13%  

Stable  disease   79%   66%  

Progressive  disease   7%   11%  

No  response  assessment   14%   19%  

Response and PFS in the ITT

Rosell, R., et al. (2012). Lancet Oncol, 13, 239-246.

Overall Survival

Rosell, R., et al. (2012). Lancet Oncol, 13, 239-246.

1/28/14

19

Randomized Studies of First Line EGFR TKI in Patients With EGFR Mutation (Through 2012)

Author Study N    (EGFR  mut  +) RR   Median  PFS  

(months)

Mok  et  al IPASS 261 71.2%  vs  47.3 9.8  vs  6.4  

Lee  et  al First-­‐SIGNAL 42 84.6%  vs  37.5% 8.4  vs  6.7  

Mitsudomi  et  al WJTOG  3405 177 62.1%  vs  32.2% 9.2  vs  6.3  

Maemondo  et  al NEJGSG002 230 73.7%  vs  30.7% 10.8  vs  5.4

Zhou  et  al OPTIMAL 154 83%  vs  36% 13.1  vs  4.6  

Rosell  et  al EURTAC 154 54.5%  vs  10.5% 9.2  vs  5.4

1018 Mok, et al. (2009). N Engl J Med, 361, 947-957; Lee, et al. (2009). Proc IASL WCLC; Mitsudomi, et al. (2010). Lancet Oncol, 11, 121-128; Maemondo, et al. (2010). N Engl J Med, 362, 2380-2388; Zhou, et al. (2010). ESMO; Rosell, R., et al. (2012). Lancet Oncol, 13, 239-246.

Afatinib in EGFR Mutated NSCLC

•  N=129 patients •  Median PFS 10.1m •  Median OS 24.8 m

Yang, et al. (2012). Lancet Oncol, 13, 539-548.

LUX-Lung 3: Afatinib in Treatment-Naive Adenocarcinoma With EGFR Mutation • Randomized, open-label phase III trial with 2:1 randomization • Afatinib 40 mg/d vs chemo; pem [500 mg/m2] –DDP [75 mg/m2] • Projected: 2,200 screened for 330 enrolled • Actual: 1,269 patients screened; 345 were randomized • Demographics: 72% East Asian, 68% never-smokers; 65% female; Exon 19

deletions (49%) and L858R point mutations (40%) • Toxicities: rash, diarrhea, mucositis, xeroderma

Conclusions: • Afatinib superior to state-of-the-art chemo in EGFR mut (+) NSCLC wrt OR%,

PFS, and QoL (PROs) • Led to FDA approval in this setting on 8/13

Arm   Afatinib   Pem-DDP   p value   HR  

OR % (independent eval)   56%   23%   < .001  PFS (mo)   11.1   6.9   < .001   0.58  PFS (mo) Exon 19 del, pt mut exon 21   13.6   6.9   < .001   0.47  

Sequist, L. V., et al. (2013). J Clin Oncol, 31(27), 3327-3334; Yang et al ASCO 2012. .

1/28/14

20

Why Is LUX-Lung 3 Important? •  Largest trial in the first-line EGFR mutant setting (at the time) •  1st to use a 2nd generation irreversible EGFR TKI •  State-of-the-art comparator (pem-DDP) •  Global trial •  Registration trial

What are the outstanding questions? •  Survival data? •  How does afatinib stack up against 1st gen EGFR TKIs? With

respect to toxicity, OR% PFS? •  Can we predict who is more likely to benefit? Proteomics? •  Appropriate approach in more obscure mutations?

Randomized Studies of First-Line EGFR TKI in Patients With EGFR Mutations

Author   Study   N    (EGFR    

mut  +)  

RR     Median  PFS  

(mo)  

Mok  et  al   IPASS   261   71.2%  vs  47.3   9.8  vs  6.4    

Lee  et  al   First-­‐SIGNAL   42   84.6%  vs  37.5%   8.4  vs  6.7    

Mitsudomi  et  al   WJTOG  3405   177   62.1%  vs  32.2%   9.2  vs  6.3    

Maemondo  et  al   NEJGSG002   230   73.7%  vs  30.7%   10.8  vs  5.4  

Zhou  et  al   OPTIMAL   154   83%  vs  36%   13.1  vs  4.6    

Rosell  et  al   EURTAC   154   54.5%  vs  10.5%   9.2  vs  5.4  

Yang  et  al   LUX-­‐Lung  3   345   56%  vs  23%   13.6  vs  6.9  

1363 Mok, et al. (2009). N Engl J Med, 361, 947-957; Lee, et al. (2009). Proc IASL WCLC; Mitsudomi, et al. (2010). Lancet Oncol, 11, 121-128; Maemondo, et al. (2010). N Engl J Med, 362, 2380-2388; Zhou, et al. (2010). ESMO; Rosell, R., et al. (2012). Lancet Oncol, 13, 239-246.

Author   Study   N    (EGFR    

mut  +)  

RR     Median  PFS  

(mo)  

Mok  et  al   IPASS   261   71.2%  vs  47.3   9.8  vs  6.4    

Lee  et  al   First-­‐SIGNAL   42   84.6%  vs  37.5%   8.4  vs  6.7    

Mitsudomi  et  al   WJTOG  3405   177   62.1%  vs  32.2%   9.2  vs  6.3    

Maemondo  et  al   NEJGSG002   230   73.7%  vs  30.7%   10.8  vs  5.4  

Zhou  et  al   OPTIMAL   154   83%  vs  36%   13.1  vs  4.6    

Rosell  et  al   EURTAC   154   54.5%  vs  10.5%   9.2  vs  5.4  

Yang  et  al   LUX-­‐Lung  3   345   56%  vs  23%   13.6  vs  6.9  

Wu  et  al   LUX-­‐Lung  6   364   67%  vs  23%   11.0  vs  5.6  

1727

Randomized  Studies  of  First-­‐Line  EGFR  TKI                                  in  Pa)ents  With  EGFR  Muta)ons  

Mok, et al. (2009). N Engl J Med, 361, 947-957; Lee, et al. (2009). Proc IASL WCLC; Mitsudomi, et al. (2010). Lancet Oncol, 11, 121-128; Maemondo, et al. (2010). N Engl J Med, 362, 2380-2388; Zhou, et al. (2010). ESMO; Rosell, R., et al. (2012). Lancet Oncol, 13, 239-246.

1/28/14

21

Author   Study   Agent  

N    (EGFR    

mut  +)   RR    

Median  PFS  

(mo)   OS  (mo)  

Mok  et  al   IPASS   Gef   261   71.2%  vs  47.3   9.8  vs  6.4     21.6  vs  21.9  

Lee  et  al   First-­‐SIGNAL   Gef   42   84.6%  vs  37.5%   8.4  vs  6.7     27.2  vs  25.6  

Mitsudomi  et  al   WJTOG  3405   Gef   177   62.1%  vs  32.2%   9.2  vs  6.3     35.5  vs  38.8  

Maemondo  et  al   NEJGSG002   Gef   230   73.7%  vs  30.7%   10.8  vs  5.4   30.0  vs  23.6  

Zhou  et  al   OPTIMAL   Erl   154   83%  vs  36%   13.1  vs  4.6     22.6  vs  28.8  

Rosell  et  al   EURTAC   Erl   154   54.5%  vs  10.5%   9.2  vs  5.4   19.3  vs  19.5  

Yang  et  al   LUX-­‐Lung  3   Afat   345   56%  vs  23%   13.6  vs  6.9   HR  1.12  

Wu  et  al   LUX-­‐Lung  8   Afat   364   67%  vs  23%   11.0  vs  5.6   HR  0.95  

Crossover to an EGFR TKI in the control group nullifies any chance of an OS benefit

Randomized Studies of First Line EGFR TKI in Patients with EGFR Mutations

Mok, et al. (2009). N Engl J Med, 361, 947-957; Lee, et al. (2009). Proc IASL WCLC; Mitsudomi, et al. (2010). Lancet Oncol, 11, 121-128; Maemondo, et al. (2010). N Engl J Med, 362, 2380-2388; Zhou, et al. (2010). ESMO; Rosell, R., et al. (2012). Lancet Oncol, 13, 239-246.

Resistance to EGFR TKI •  Regardless of extent of initial response, resistance to

TKI develops invariably due to –  T790 mutation (~ 50% to 60%) –  MET amplification (~ 5% to 20%) –  Conversion to SCLC (< 5%)

•  EGFR mutated tumors might progress rapidly upon withdrawal of TKI

•  Role of continuation of TKI beyond progression is under evaluation

•  Afatinib: Targets T790 mutation, and we have seen promising activity for the afatinib/C225 combination in pts with acquired resistance

Sequist, L., et al. (2011). Sci Transl Med, 3(75), 75ra26; Pao, W., et al. (2005). PLoS Med, 2(3), e73.

•  141 pts were screened for ALK fusions, 13% positive

•  F=M, young, non-smokers, usually adenocarcinoma

•  No overlap between EGFR and Kras mutants

•  Refractory to EGFR TKIs •  Typical chemo responses

(not increased as with EGFR mutations)

•  3% to 8% of general NSCLC population Signet-ring cells

Shaw,  A.  T.,  et  al.  (2009).  J  Clin  Oncol,  27(26),  4247-­‐4253.  

1/28/14

22

Pre-Treatment Crizotinib x 12 weeks

Response to Crizotinib (ALK Inhibitor)

Crino et al. ASCO 2011 Abstract 7514

Clinical Activity of the Oral ALK Inhibitor, Crizotinib (PF-02341066), in Patients With ALK-positive NSCLC

Key entry criteria • Positive for ALK by central laboratory

• 1 prior chemotherapy (platinum based)

Crizotinib 250 mg BID (n=159) administered on a continuous dosing schedule

Pemetrexed 500 mg/m2 or docetaxel 75 mg/m2 (n=159) infused on day 1 of a 21-day cycle

RANDOMIZE

PROFILE 1007

Key entry criteria • Positive for ALK by central laboratory

• Chemo-naive

PROFILE 1006 N=318

Current crizotinib clinical trials

Crizotinib 250 mg BID (n=159) administered on a continuous dosing schedule

Pemetrexed 500 mg/m2 and cisplatin 75 mg/m2 (n=159) infused on day 1 of a 21-day cycle

RANDOMIZE

N=400 PROFILE 1007-NCT00932893; PROFILE 1005-NCT00932451. Bang  Y  et  al.  J  Clin  Oncol.  2010;28(18s).  Abstract  3.    Shaw, et al. (2013). N Engl J Med, 368, 2385-2394.

1/28/14

23

Primary Endpoint: PFS by Independent Radiologic Review (ITT Population)

Shaw, et al. (2013). N Engl J Med, 368, 2385-2394.

PFS Subgroup Analysis

Shaw, et al. (2013). N Engl J Med, 368, 2385-2394.

aRECIST v1.1; bITT population; cas-treated population

Improved Response Rate

65.3

19.5 OR

R (%

)

ORR ratio: 3.4 (95% CI: 2.5 to 4.7); P<0.0001

Crizotinib (n=173)

Chemotherapy (n=174) 80

60

40

20

0 Treatment

65.7

29.3

6.9

Crizotinib (n=172) Pemetrexed (n=99) Docetaxel (n=72)

Treatment

80

60

40

20

0

Shaw et al. ESMO 2012.

1/28/14

24

Interim Analysis of OS

Shaw, et al. (2013). N Engl J Med, 368, 2385-2394.

Patient Reported Outcomes Symptoms and Quality of Life

Shaw, et al. (2013). N Engl J Med, 368, 2385-2394.

Crizotinib Adverse Events in PROFILE 1007

*AEs reported in 15% or more of patients in either treatment group

Shaw, et al. (2013). N Engl J Med, 368, 2385-2394.

1/28/14

25

Summary of PROFILE 1007 Compared to single-agent chemotherapy, crizotinib:

•  Prolongs progression-free survival

•  Improves response rate

•  Improves symptoms and quality of life

Side effects are generally mild and manageable

•  Monitor liver function tests

•  Treat GI side effects

First and Second Generation ALK TKIs in Clinical Development

Name Company Status Comments

Crizotinib Pfizer 1st and 2nd line registration trials are ongoing. 2nd line trial is completed

Accelerated approval granted August 26, 2011 Regular approval granted November 20, 2013

LDK378* Novartis Phase I dose escalation Phase II

Activity observed at 400 mg. Enrolling in the US and Europe.

AF802 Chugai Phase I/II in Japan and starting in US

Activity observed in crizotinib-naive pts

AP26113* Ariad Phase I ongoing Some activity against EGFR T790M. Enrolling in the US.

ASP3026 Astellos Phase I Similar to TAE684. Enrolling in Japan.

CEP-28122 Cephalon Preclinical

NMS-E628 Nerviano Preclinical

X276/396 Xcovery Preclinical

*Activity seen in crizotinib exposed patients

ALK Inhibitors: RR%

Compound   Company   Phase   No.   ALKI naive   ALKI exposed  Crizotinib   Pfizer   I/II/III   261   60%   NA  LDK 378*   Novartis   I/II   130   60%   57%  

CH5424802*   Chugai   I/II   46   94%   NA  AP 26113*   Ariad   I/II   21   50%   73%  

TSR011   Tesaro   I/II   NA   NA   NA  

Outstanding Questions • Do different ALK fusions lead to variable response? • Can we heighten repsonse and PFS up front with combinatorial Tx?

•  HSP 90, MEKI, mTORI • Can we overcome resistance?

Shaw, et al. (2013). N Engl J Med, 368, 2385-2394; Shaw, et al. (2013). ASCO, Abstract 8010; Nakagawa, et al. (2013). ASCO A-8033. Camidge et al. (2013). ASCO A-8031. Tesaro, European Cancer Congress 2013.

1/28/14

26

Advances in Recurrent and Metastatic NSCLC: Past 25 yr

Endpoint 2nd Gen Era

< 1990 3rd Gen Era 1990-2000

Bev Era 2000-2005

Targeted Era [>2005]

Overall response 15% 25% 35% 35%-60% Time to progression 2 mo 4 mo 6 mo 7-12 mo

Median survival 6 mo 8-10 mo 12-14 mo 15+ mo

1-yr survival 15%-20% 30% 40%-50% 50%-60%

2-yr survival < 5% 10%-15% 20% 25%-50%

First-­‐Line  Therapy  in  NSCLC      

NSCLC    

1st  line  

EGFR    mt  +    

Gefi)nib    or  Erlo)nib    or  Afa)nib  

small  molecule  inhibitors  of  EGFR;    

EGFR  nega)ve    or  unknown    

Nonsquamous  

Pla)num  double-­‐based  therapy,  Cispla)n  or  Carbopla)n  plus  

pemetrexed,  taxane,  gemcitabine  or  vinorelbine  

Bevacizumab-­‐eligible  

Pla)num  doublet  with  bevacizumab    

Con)nua)on  or  Switch  

maintenance    

Squamous  Pla)num  /gemcitabine;  

Pla)num/taxane  

Clinical  Trial  

Azzoli, C. G., et al. (2009). J Clin Oncol, 27(36), 6251-6266.

Alk+  or  Ros1  

Crizo)nib  

Molecular Therapy for Advanced NSCLC: Conclusions

•  Clear role for first line EGFR TKI in patients harboring EGFR mutations (10%-15%)

•  Crizotinib highly active in patients with EML4/ALK translocation (4%-7%), now approved by FDA (independent of line of treatment); also active in ROS-1 positive patients

•  Dabrafenib has activity in BRAF V600 mutants •  MET, KRAS, and other molecular targets may prove

“actionable” in the near future •  Ongoing studies with PD1 MAb and Ipilimumab will

establish their role, if any, in advanced NSCLC