Agenda. 6:30pm 7:00pm. Dinner. 7:00pm 7:15pm. NSCLC Treatment in 2014: Focus on Use of 2nd Generation TKIs in Clinical Practice.

Similar documents
Treatment of EGFR mutant advanced NSCLC

Treatment of EGFR mutant advanced NSCLC

Management Guidelines and Targeted Therapies in Metastatic Non-Small Cell Lung Cancer: An Oncologist s Perspective

Overall survival with afatinib versus chemotherapy in patients with NSCLC harboring common EGFR

Inhibidores de EGFR Noemi Reguart, MD, PhD Hospital Clínic Barcelona IDIPAPS

Targeted Therapies for Advanced NSCLC

Targeted Agents as Maintenance Therapy. Karen Kelly, MD Professor of Medicine UC Davis Cancer Center

Next Generation EGFR Inhibitors

Slide 1. Slide 2 Maintenance Therapy Options. Slide 3. Maintenance Therapy in the Management of Non-Small Cell Lung Cancer. Maintenance Chemotherapy

Sequencing in EGFR-Mutated NSCLC: Does Order Matter?

EGFR inhibitors in NSCLC

Maintenance therapy in advanced non-small cell lung cancer. Egbert F. Smit MD PhD Dept Thoracic Oncology Netherlands Cancer Institute

EGFR Mutation-Positive Acquired Resistance: Dominance of T790M

EGFR TKI sequencing: does order matter?

Target therapy nel NSCLC con EGFR M+ Cesare Gridelli Division of Medical Oncology S.G. Moscati Hospital Avellino (Italy)

PROGNOSTIC AND PREDICTIVE BIOMARKERS IN NSCLC. Federico Cappuzzo Istituto Toscano Tumori Ospedale Civile-Livorno Italy

Management Strategies for Lung Cancer Sensitive or Resistant to EGRF Inhibitors

MAINTENANCE TREATMENT CHEMO MAINTENANCE OR TARGETED OF BOTH? Martin Reck Department of Thoracic Oncology LungenClinic Grosshansdorf

Quale sequenza terapeutica nella malattia EGFR+

Best of ASCO 2014: Highlights in Metastatic Non-Small Cell Lung Cancer

Maintenance Therapy for Advanced NSCLC: When, What, Why & What s Left After Post-Maintenance Relapse?

Maintenance paradigm in non-squamous NSCLC

Improving outcomes for NSCLC patients with brain metastases

Metastatic NSCLC: Expanding Role of Immunotherapy. Evan W. Alley, MD, PhD Abramson Cancer Center at Penn Presbyterian

Antiangiogenic Agents in NSCLC Where are we? Which biomarkers? VEGF Is the Only Angiogenic Factor Present Throughout the Tumor Life Cycle

Maintenance Therapy for Advanced NSCLC: Which Patients, Which Approach?

J. C.-H. Yang 1, L.V. Sequist 2, S. L. Geater 3, C.-M. Tsai 4, T. Mok 5, M. H. Schuler 6, N. Yamamoto 7, D. Massey 8, V. Zazulina 8, Yi-Long Wu 9

Recent Advances in Lung Cancer: Updates from ASCO 2017

Sequence or intercalation of use of targeted agents and Chemotherapy Definition of progression under TKI

Biomarkers of Response to EGFR-TKIs EORTC-NCI-ASCO Meeting on Molecular Markers in Cancer November 17, 2007

1st line chemotherapy and contribution of targeted agents

2 nd line Therapy and Beyond NSCLC. Alan Sandler, M.D. Oregon Health & Science University

Emerging Algorithm for Optimal Sequencing of EGFR TKIs in EGFR Mutation Positive NSCLC

Treatment of EGFR-Mutation+ NSCLC in 1st- and 2nd-Line

Optimum Sequencing of EGFR targeted therapy in NSCLC. Dr. Sema SEZGİN GÖKSU Akdeniz Univercity, Antalya, Turkey

Choosing Optimal Therapy for Advanced Non-Squamous (NS) Non-Small Cell Lung Cancer

Frequency of Epidermal Growth Factor Mutation Status and Its Effect on Outcome of Patients with Adenocarcinoma of the Lung

Changing demographics of smoking and its effects during therapy

IMPORTANT PATHWAYS TO TARGET IN (ADVANCED) NSCLC:

Afatinib in patients with EGFR mutation-positive NSCLC harboring uncommon mutations: overview of clinical data

EGFR MUTATIONS: EGFR PATHWAY AND SELECTION OF FIRST-LINE THERAPY WITH TYROSINE KINASE INHIBITORS

LONDON CANCER NEW DRUGS GROUP RAPID REVIEW. Erlotinib for the third or fourth-line treatment of NSCLC January 2012

PERIOPERATIVE TREATMENT OF NON SMALL CELL LUNG CANCER. Virginie Westeel Chest Disease Department University Hospital Besançon, France

Virtual Journal Club: Front-Line Therapy and Beyond Recent Perspectives on ALK-Positive Non-Small Cell Lung Cancer.

Next-Generation Covalent Irreversible Kinase Inhibitors in NSCLC: Focus on Afatinib

Plotting the course: optimizing treatment strategies in patients with advanced adenocarcinoma

Osimertinib Activity in Patients With Leptomeningeal Disease From Non-Small Cell Lung Cancer: Updated Results From the BLOOM Study

Immune Checkpoint Inhibitors for Lung Cancer William N. William Jr.

Best of ASCO 2014 Lung

Second-line treatment for advanced NSCLC

Osimertinib as first-line treatment of EGFR mutant advanced nonsmall-cell

Management of EGFR-Mutation Positive Metastatic Non-Small Cell Lung Cancer

The Rapidly Changing World of EGFR Mutation-Positive Acquired Resistance

State of the Art Treatment of Lung Cancer Ravi Salgia, MD, PhD

INNOVATION IN LUNG CANCER MANAGEMENT. Federico Cappuzzo Department of Oncology-Hematology, AUSL della Romagna, Ravenna, Italy

NSCLC: Terapia medica nella fase avanzata. Paolo Bidoli S.C. Oncologia Medica H S. Gerardo Monza

11/21/2009. Erlotinib in KRAS Mt patients. Bevacizumab in Squamous patients

Sponsor / Company: Sanofi Drug substance(s): Docetaxel (Taxotere )

Nivolumab: esperienze italiane nel carcinoma polmonare avanzato

Afatinib dose adjustment: Effect on safety, efficacy and patient-reported outcomes in the LUX-Lung 3/6 trials in EGFRm+ NSCLC

Antiangiogenici in combinazione a chemioterapia in prima linea: bevacizumab

Practice changing studies in lung cancer 2017

Second-line treatment for advanced NSCLC

Kazuhisa TAKAHASHI, MD, PhD

TKI en primera línea EGFR mutado:importancia del equilibrio eficacia tolerabilidad

Squamous Cell Carcinoma Standard and Novel Targets.

Original Article. Abstract

Joachim Aerts Erasmus MC Rotterdam, Netherlands. Drawing the map: molecular characterization of NSCLC

Lung Cancer Case. Since the patient was symptomatic, a targeted panel was sent. ALK FISH returned in 2 days and was positive.

Conversations in Oncology. November Kerry Hotel Pudong, Shanghai China

Molecular Targets in Lung Cancer

Do You Think Like the Experts? Refining the Management of Advanced NSCLC With ALK Rearrangement. Reference Slides Introduction

CURRENT STANDARD OF CARE OF LUNG CANCER. Maroun El-Khoury, MD Consultant Oncologist/Hematologist American Hospital Dubai President of Medical staff

Immunotherapy in the clinic. Lung Cancer. Marga Majem 20 octubre 2017

Targeted Therapy for NSCLC: EGFR and ALK Fadlo R. Khuri, MD

OUR EXPERIENCES WITH ERLOTINIB IN SECOND AND THIRD LINE TREATMENT PATIENTS WITH ADVANCED STAGE IIIB/ IV NON-SMALL CELL LUNG CANCER

Introduction. Methods. Patient and study design

Slide 1. Slide 2. Slide 3. Disclosures. Personalized Medicine for Advanced NSCLC in East Asia. No conflicts related to this presentation

First line erlotinib for NSCLC patients not selected by EGFR mutation: keep carrying the TORCH or time to let the flame die?

Overview of Lung Cancer :Perspectives from Cancer Genotype. Ji-Youn Han, MD, PhD. Center for Lung Cancer National Cancer Center

EGFR mutations in early-stage and advanced-stage lung adenocarcinoma: Analysis based on large-scale data from China

Targeted therapy in lung cancer : experience of NIO-RABAT

Diagnostic with alternative sample types (liquid biopsy)

Targeting Acquired Resistance to EGFR Kinase Inhibitors: Beyond T790M Mutation

Non-small Cell Lung Cancer: Multidisciplinary Role: Role of Medical Oncologist

EGFR Tyrosine Kinase Inhibitors Prolong Overall Survival in EGFR Mutated Non-Small-Cell Lung Cancer Patients with Postsurgical Recurrence

RESEARCH ARTICLE. Ryosuke Hirano 1, Junji Uchino 1 *, Miho Ueno 2, Masaki Fujita 1, Kentaro Watanabe 1. Abstract. Introduction

Combined Modality Therapy State of the Art. Everett E. Vokes The University of Chicago

Strategies in the therapy of advanced NSCLC SAMO Winter-Conference 2008 on Chest tumors

GIOTRIF (AFATINIB*) For journalists outside the US/UK/Canada only 1. WHAT IS GIOTRIF (AFATINIB*)? 2. HOW DOES GIOTRIF (AFATINIB*) WORK?

Retrospective analysis of Gefitinib and Erlotinib in EGFR-mutated non-small-cell lung cancer patients

Yan Zhang 1*, Zheng Wang 2*, Xuezhi Hao 1, Xingsheng Hu 1, Hongyu Wang 1, Yan Wang 1, Jianming Ying 3. Original Article. Abstract

Editorial Process: Submission:10/27/2017 Acceptance:05/25/2018

Correspondence should be addressed to Kumar Prabhash;

Malignant pleural Mesothelioma: A Year In Review

IRESSA (Gefitinib) The Journey. Anne De Bock Portfolio Leader, Oncology/Infection European Regulatory Affairs AstraZeneca

First-line treatment of EGFR-mutated nonsmall cell lung cancer: critical review on study methodology

ASCO Highlights Lung Cancer

Biomarkers in oncology drug development

AURA 3: the last word on chemotherapy as a control arm in EGFR mutant NSCLC?

Transcription:

Agenda 6:30pm 7:00pm Dinner 7:00pm 7:15pm Welcome and Introductions Natasha Leighl, MD 7:15pm 7:50pm 7:50pm 8:00pm NSCLC Treatment in 2014: Focus on Use of 2nd Generation TKIs in Clinical Practice Questions Tony Mok, MD 8:00pm 9:00pm Closing Remarks/Interactive Discussion Natasha Leighl, MD & Tony Mok, MD

Welcome and Introductions Natasha Leighl, MD, FRCP(C) Princess Margaret Hospital Associate Professor, Department of Medicine University of Toronto

Welcome & Introductions The CARE (Community Academic Research Education) Faculty is a Pan-Canadian group of medical oncologists from across Canada who meet and discuss various topics from key conferences. The vision of the CARE Lung Cancer Faculty is to share opinions and update Canadian specialists with key news and developments from key conferences framed in a Canadian perspective. The mission of the CARE Lung Cancer Faculty is to enhance medical education with the explicit goal of improving patient outcomes. The CARE Lung Cancer Faculty is proud to host globally recognized lung cancer specialist Dr. Tony Mok. He previously spoke in Vancouver, Montreal and Toronto in January of this year, as part of a three-day CARE Lung Cancer Speaker Tour.

NSCLC Treatment in 2014: Focus on Use of 2nd Gen TKIs in Clinical Practice Tony Mok, MD, FRCP(C) Professor, Department of Clinical Oncology, Chinese University of Hong Kong, Prince of Wales Hospital President, IASLC

Optimal Strategy in EGFR Inhibition: 2014 Professor Tony Mok Li Shu Fun Medical Foundation Professor of Clinical Oncology The Chinese University of Hong Kong

Optimal Strategies Finding the EGFR Mutation Choosing a EGFR TKI Stopping the EGFR TKI Treating the resistance Adding to EGFR TKI

Optimal Strategies Finding the EGFR Mutation How good are we?

Mutation testing in Asia 2011 Country (N diagnosed with NSCLC * ) Proportion tested for EGFR mutations % (95% CI ) Proportion of males/females, smokers and non-smokers, and histological subtypes that were tested for EGFR mutations Gender Smoking status Histology ADC / Males / Females Current + ex-smoker All non-adc / Only (%) / Never smoker (%) SCC (%) Total (22,193) 31.8 (31.2 32.5) 26.9 / 40.2 47.0 / 57.4 50.4 / 12.5 / 12.5 China (12,086 ) 18.3 (17.6 19.0) 15.2 / 25.3 N.D. 30.3 / 8.0 / 9.4 Hong Kong (795) 42.0 (38.6 45.5) 36.2 / 52.3 34.1 / 52.1 55.4 / 9.0 / 6.4 Japan (2,379) 64.8 (62.9 66.7) 63.6 / 67.0 68.8 / 68.3 69.2 / 55.0 / 50.3 Korea (3,794) 33.5 (32.0 35.0) 26.1 / 38.1 27.1 / 42.9 62.7 / 9.8 / 8.3 Taiwan (2,890) 54.3 (52.5 56.1) 47.1 / 64.3 37.0 / 56.8 69.3 / 15.5 / 8.5 Thailand (249 ) 57.8 (51.6 63.8) 51.6 / 69.3 49.5 / 84.7 83.6 / 7.1 / 6.9

Sequencing was used in 40% of tumor tissue Methods of Testing

Optimal strategy 1wk 2wk 3wk Circulating 4wk Free Plasma DNA (cfplasma DNA) Core needle biopsy Pathologic diagnosis of adeno carcinoma EGFR or ALK testing Circulating Tumor Cell (CTC)

Targeting EGFR mutation -cobas 4800 Blood test -COLD PCR -Digital PCR

cobas EGFR _ blood Test _ Kit Components Utilizing most of the reagents in the cobas EGFR_FFPET test and requiring additional reagents and the blood-specific data analysis software Blood ctdna Preparation Kit Cobas EGFR_Blood ctdna SP 2 ml Plasma cobas cell-free DNA Preparation Kit (To be used for other blood based assays) cobas EGFR _ blood Test cobas 4800 v 2.0 cobas EGFR _ Blood HPEA x25 PK x2 PBB x6 Additional reagents added to cobas DNA preparation Kit Tube FAM HEX JA270 1 EX 19Del S768I 2 L858R T790M 3 G719X L861Q* EX 20Ins *New primer and probe for L861Q Blood-specific cutoffs; Blood-specific data analysis software Mok et al ASCO 2013

FASTACT 2: Tumor versus plasma DNA for EGFR Mutation Analysis TISSUE SAMPLES 397 (88%) patients consented 301 (66.7%) samples available PLASMA SAMPLES 241 (53.4%) samples analyzable 224 patients with matched tumor and plasma samples 451 (100%) patients consented 427 (94.6%) samples available 427 (94.6%) samples analyzable

Concordance between tumor and plasma samples Total of 224 patients had both tumor and baseline plasma samples with available EGFR mutation analysis results (Table 3) Sensitivity: 77% (69/90) Specificity: 96% (129/134) Positive predictive value: 93% (69/74) Negative predictive value: 86% (129/150) Overall concordance: 88% (198/224) EGFR Activating Mutations p-egfr Mut+ (Plasma) p-egfr Mut- (Plasma) Total t-egfr Mut+ (Tumor) 69 21 90 t-egfr Mut- (Tumor) 5 129 134 Total 74 150 224

Droplet digital PCR (ddpcr) Hindson et al. Analytical chemistry 2011

Positive result on exon 19 deletion assay Plasma sample 214 Mutant concentration:72 copies/ml plasma Fraction concentration : 3.2%

Negative result on exon 19 deletion assay

Analyzing plasma and tumor sample from ASPIRATION study and matched control (n=197) Tumor sample: COBAS EGFR Mutation Test Plasma sample: Droplet digital PCR Lee et al WCLC 2013

Diagnostic utility of digital PCR for detection of EGFR mutation POS in plasma NEG in plasma POS in tumor 117 27 144 NEG in tumor 0 53 53 Droplet digital PCR Sensitivity 81% Specificity 100% Positive Predictive Value 100% Concordance 86% 117 80 197

Blood-based molecular analysis may optimize the timing and coverage

First line TKI Finding the EGFR Mutation Choosing a EGFR TKI

Where we are? Author Study N (EGFR mut +) RR Median PFS Mok et al IPASS 132 71.2% vs 47.3 9.8 vs 6.4 months Lee et al First-SIGNAL 27 84.6% vs 37.5% 8.4 vs 6.7 months Mitsudomi et al WJTOG 3405 86 62.1% vs 32.2% 9.2 vs 6.3 months Maemondo et al NEJGSG002 114 73.7% vs 30.7% 10.8 vs 5.4 months Zhou et al OPTIMAL 154 83% vs 36% 13.1 vs 4.6 months Rosell et al EURTAC 135 56% vs 18% 9.2 vs 4.8 months Yang et al LUX Lung 3 345 56% vs 22% 11.1 vs 6.9 months Wu et al LUX Lung 6 364 67% vs 23% 11.0 vs 5.6 months Mok et al NEJM 2009, Lee et al WCLC 2009, Mitsudomi et al Lancet Oncology 2010, Maemondo NEJM 2010 Zhou et al Lancet Oncol 2010; Yang et al JCO epub; Wu et al Lancet Oncol: In Press

Katakami et al ASCO 2014 ASCO 2014: WJOG5108 Study Register Randomize Lung adenoca. Evaluable 2nd line & latter Age > 20 y.o PS 0-2 Stage III, IV, recurrence No interstitial lung disease R Stratify gender PS stage smoking history mutation status institution prior regimen A Erlotinib 150mg/day B Gefitinib 250mg/day PD

EGFR mutation (+) % PFS 95% CI p E 10.09 mo 8.54-11.60 mo 0.532 G 8.90 mo 7.59-10.38 mo EGFR mutation (-) % PFS 95% CI p E 2.10 mo 1.35-3.22 mo 0.221 G 2.07 mo 1.64-3.68 mo E G EGFR mutation unknown % PFS 95% CI p E 2.53 mo 1.54-5.22 mo 0.878 G 3.61 mo 2.27-6.14 mo 26

LUX Lung 3 Stage IIIB (wet)/iv lung adenocarcinoma (AJCC version 6) EGFR mutation in tumor (central lab testing; Therascreen EGFR29* RGQ PCR) Randomization 2:1 stratified by EGFR mutation (Del19/L858R/other) and race (Asian/non-Asian) Afatinib 40 mg/day Cisplatin + Pemetrexed 75 mg/m 2 + 500 mg/m 2 i.v. q21days, up to 6 cycles Primary endpoint: PFS (RECIST 1.1, independent review) Secondary endpoints: ORR, DCR, DoR, tumor shrinkage, OS, PRO, safety, PK Primary PFS analysis (independent review) Sample size: 217 independent events needed to detect HR of 0.64 (or median increase in PFS from 7 to 11 months) at two-sided 5% significance level with 90% power Yang JC, et al. Yang et al JCO 2013

Progression-free survival (probability) 1.0 0.8 0.6 Primary endpoint: PFS Independent review all randomized patients Afatinib n=230 Cis/pem n=115 PFS event, n (%) 152 (66) 69 (60) Median PFS (months) 11.1 6.9 Hazard ratio (95% confidence interval) 0.58 (0.43 0.78) p=0.0004 0.4 47% 0.2 22% 0.0 0 3 6 9 12 15 18 21 24 27 Number at risk Progression-free survival (months) Afatinib 230 180 151 120 77 50 31 10 3 0 Cis/Pem 115 72 41 21 11 7 3 2 0 0

Progression-free survival (probability) Common mutations: PFS Independent review patients with Del19/L858R (n=308) 1.0 0.8 0.6 LUX lung 2: PFS 13.7m Afatinib n=204 Cis/pem n=104 PFS event, n (%) 130 (64) 61 (59) Median PFS (months) 13.6 6.9 Hazard ratio (95% confidence interval) 0.47 (0.34 0.65) p<0.0001 0.4 51% 0.2 21% 0.0 0 3 6 9 12 15 18 21 24 27 Number at risk Progression-free survival (months) Afatinib 204 169 143 115 75 49 30 10 3 0 Cis/Pem 104 62 35 17 9 6 2 2 0 0

LUX Lung 6 Asian patients (China, South Korea, Thailand) Stage IIIB (wet)/iv lung adenocarcinoma EGFR mutation in the tumor* No prior treatment for advanced disease, no prior EGFR TKI ECOG PS 0 or 1 Randomization Stratified by mutation (Del19/L858R/other) 2:1 Afatinib 40 mg/day Gemcitabine + Cisplatin 1000 mg/m 2 D1, D8 + 75 mg/m 2 i.v. q21 days, up to 6 cycles Primary endpoint: PFS (RECIST 1.1, independent review) Secondary endpoints: ORR, DCR, DoR, tumor shrinkage, OS, PRO, safety *Central lab testing; Therascreen EGFR29 RGQ PCR detecting 19 deletions in exon 19, 3 insertions in exon 20, L858R, L861Q, T790M, G719S, G719A and G719C (or G719X), S768I. Dose escalated to 50 mg if limited AE observed in cycle 1. Dose reduced by 10 mg decrements in case of related G3 or prolonged G2 AE. Tumor assessments: q6 weeks until Week 48 and q12 weeks thereafter until progression/start of new therapy. Patient-reported outcomes: EQ-5D, EORTC QLQ-C30 and QLQ-LC13 at randomization and q3 weeks until progression or new anti-cancer therapy. Presented by: WU YL et al ASCO 2013

Primary endpoint PFS by independent review Presented by:

Study 1017: Clinical Activity of Dacomitinib (PF-00299804) in First-line Advanced NSCLC with an EGFR-activating Mutation Cohort A: Non- or former light-smoker or EGFR-mutation (1 st -line) n=89 (fully enrolled) Dacomitinib 45 mg QD (amended to 30 mg for selected patients b ) Until progression Cohort B: HER2 mutation or HER2 amplification a Target n=25 (still recruiting) Optional biopsy on progression a [gene]/[centromere of chromosome 17] ratio >2 b Starting dose changed to 30 mg QD with dose escalation to 45 mg QD after 8 weeks of treatment in absence of grade >1 toxicity for 1 month for 30 patients in Cohort A, and patients in Cohort B who had no prior lines of therapy Janne and Mok et al Lancet Oncology In Press

Change from baseline (%) Waterfall Plot for Patients with EGFR-mutant Lung Cancers with Exon 19 and Exon 21 Mutations (N=45) 20 10 0 10 20 30 40 50 60 70 80 90 100 110 PR Exon 19 Exon 21 RR=76%

From PFS to OS

Estimated OS probability Estimated OS probability LUX-Lung 3 and 6: OS in common mutations LUX-Lung 3 LUX-Lung 6 1.0 Median, months Afatinib n=203 Pem/Cis n=104 31.57 28.16 1.0 Median, months Afatinib n=216 Gem/Cis n=108 23.6 23.5 0.8 HR (95%CI), p-value 0.78 (0.58 1.06), p=0.1090 0.8 HR (95%CI), p-value 0.83 (0.62 1.09), p=0.1756 0.6 0.6 0.4 0.4 0.2 0.2 0 0 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 Time (months) No of patients Afatinib 203 197 188 181 171 162 143 133 121 108 101 90 58 49 32 9 1 0 Pem/Cis 104 98 92 86 81 71 63 55 52 47 40 35 26 20 10 5 1 0 Time (months) No of patients Afatinib 216 214 202 190 172 158 141 118 104 93 80 51 19 9 1 0 Gem/Cis 108 101 93 87 81 70 61 55 49 36 30 17 8 3 0 0 Presented by: James Chih-Hsin Yang

Estimated OS probability Combined OS analysis: common mutations (n=631) 1.0 0.8 0.6 Afatinib n=419 Chemo n=212 Median, months 27.3 24.3 HR (95%CI), p-value 0.81 (0.66 0.99), p=0.0374 0.4 0.2 0 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 Time (months) No of patients Afatinib 419 411 390 371 343 320 284 251 225 201 181 141 77 58 33 9 1 0 Chemo 212 199 185 173 162 141 124 110 101 83 70 52 34 23 10 5 1 0 Presented by: James Chih-Hsin Yang

Estimated OS probability Estimated OS probability Two subgroups of LUX Lung 3 and 6 OS analysis Del19 L858R 1.0 Median, months Afatinib n=236 Chemo n=119 31.7 20.7 1.0 Median, months Afatinib n=183 Chemo n=93 22.1 26.9 0.8 HR (95%CI), p-value 0.59 (0.45 0.77), p=0.0001 0.8 HR (95%CI), p-value 1.25 (0.92 1.71), p=0.1600 0.6 0.6 0.4 0.4 0.2 0.2 0 0 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 Time (months) No of patients Afatinib 236 230 223 217 202 192 173 160 145 131 117 90 50 38 22 6 1 0 Chemo 119 113 103 95 87 72 63 55 51 43 38 27 14 9 1 1 0 0 Time (months) No of patients Afatinib 183 181 167 154 141 128 111 91 80 70 64 51 27 20 11 3 0 0 Chemo 93 86 82 78 75 69 61 55 50 40 32 25 20 14 9 4 1 0 Exon 19 Exon 21

Why???

Estimated OS probability Estimated OS probability Afatinib: OS in Del19 subgroup 1.0 0.8 Median, months HR (95%CI), p-value LUX-Lung 3 Afatinib n=112 Pem/Cis n=57 33.3 21.1 0.54 (0.36 0.79), p=0.0015 1.0 0.8 Median, months HR (95%CI), p-value LUX-Lung 6 Afatinib n=124 Gem/Cis n=62 31.4 18.4 0.64 (0.44 0.94), p=0.0229 0.6 0.6 0.4 0.4 0.2 0.2 0 0 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 Time (months) No of patients Afatinib 112 108 105 102 96 93 83 80 72 62 58 51 34 30 21 6 1 0 Pem/Cis 57 55 50 46 43 37 33 27 25 22 20 16 10 6 1 1 0 0 Time (months) No of patients Afatinib 124 122 118 115 106 99 90 80 73 69 59 39 16 8 1 0 Gem/Cis 62 58 53 49 44 35 30 28 26 21 18 11 4 3 0 0 Presented by: James Chih-Hsin Yang

Treatment beyond 1st line in patients with common mutations LUX-Lung 3 LUX-Lung 6 Afatinib (n=203) Pem/Cis (n=104) Afatinib (n=216) Gem/Cis (n=108) Discontinued treatment, n (%) 184 (100) 104 (100) 194 (100) 108 (100) Subsequent systemic therapy, n (%)* 144 (78) 88 (85) 123 (63) 70 (65) Chemotherapy, n (%) 131 (71) 49 (47) 114 (59) 29 (27) EGFR TKI therapy, n (%) 81 (44) 78 (75) 50 (26) 61 (56) Erlotinib Gefitinib Afatinib AZD9291 Dacomitinib Icotinib EGFR TKI combinations 61 (33) 28 (15) 2 (1) 2 (1) 5 (3) 46 (42) 44 (42) 7 (7) 1 (1) 1 (1) 9 (9) 21 (11) 19 (10) 11 (6) 5 (3) 22 (20) 39 (36) 3 (3) 3 (3) Other, n (%) 5 (3) 2 (2) 3 (2) 4 (4) Radiotherapy, n (%) 32 (17) 21 (20) 4 (2) 0 (0) *Collection of data on subsequent therapies still ongoing. Presented by: James Chih-Hsin Yang 419/419 pt had exposure to first line TKI plus 131/419 had further TKI 131/212 pt had exposure to TKI

More TKI exposure during the course of illness may be associated with longer survival 419/419 pt had exposure to first line TKI plus 131/419 had further TKI 131/212 pt had exposure to TKI

Median progression-free survival (months) Exon 21: a weaker cousin? 1 2 3 4 5 6 7 8 1. Mok et al. N Engl J Med. 2009;361:947; 2. Maemondo et al. N Engl J Med. 2010;362:2380; 3. Mitsudomi et al. Lancet Oncol. 2010;11:121; 4. Rosell et al. Lancet Oncol. 2012;13:239; 5. Wu et al. J Thorac Oncol. 2013;8:suppl 2 (P1.11-021); 6. Zhou et al. Lancet Oncol. 2011;12:735; 7. Sequist et al. J Clin Oncol. 2013;31:3327; 8. Wu et al. Lancet Oncol. 2014;15:213. Presented by: James Chih-Hsin Yang

Direct comparison: LUX Lung 7 Randomized IIb Study Is Afatinib better than Gefitinib in patients with EGFR mutation? Advanced NSCLC Adenocarcinoma EGFR mut+ First-line treatment PS 0-1 N= 264 patients R A N D O M I Z E 1 1 Afatinib 40mg qd Gefitinib 250mg qd PFS 13.7 vs 10 months HR 0.73 Sample size increased to 319

Direct comparison: LUX Lung 7 Randomized IIb Study Is Afatinib better than Gefitinib in patients with EGFR mutation? Advanced NSCLC Adenocarcinoma EGFR mut+ First-line treatment PS 0-1 N= 264 patients R A N 1 D Aug O 16 2013 M I 1 Z E Complete accrual on Afatinib 40mg qd Gefitinib 250mg qd PFS 13.7 vs 10 months HR 0.73 Sample size increased to 319

ARCHER 1050: Randomized Phase III Study Dacomitinib vs Gefitinib Advanced NSCLC Adenocarcinoma EGFR exon 19/21 mut+ First-line treatment PS 0-1 R A N D O M I Z E 1 1 Dacomitinib 45mg qd Gefitinib 250mg qd Primary endpoint in PFS 14.8 vs 9.5 months N= 440 patients Stratification -Race -Exon 19 v 21

ARCHER 1050: Randomized Phase III Study Dacomitinib vs Gefitinib Advanced NSCLC Adenocarcinoma EGFR exon 19/21 mut+ First-line treatment PS 0-1 N= 440 patients R A N D O M I Z E 1 303 accrued in Sept 2014 1 Stratification -Race -Exon 19 v 21 Dacomitinib 45mg qd Gefitinib 250mg qd Primary endpoint in PFS 14.8 vs 9.5 months

Second generation TKI may optimize the PFS and possibly the OS (for exon19 deletion)

Optimal Strategy Finding the EGFR Mutation Choosing a EGFR TKI Stopping the EGFR TKI

Options at RECIST progression RECIST PD to first line TKI Continue with TKI Change to doublet chemo Add doublet chemo and continue TKI

Options at RECIST progression RECIST PD to first line TKI Continue with TKI ASPIRATION ESMO Abst 5892 Sept.27 at 15:45

ASPIRATION: Study design Patients 18 years with stage IV, EGFR mut+ NSCLC Erlotinib PFS 1 PD by RECIST Erlotinib PD by physician assessment PFS 2 Inclusion criteria: patients 18 years with confirmed stage IV or recurrent NSCLC with exon 18 21 mutations (except T790M) with measurable disease and ECOG PS 0 2 Exclusion criteria: T790M mutations, prior chemotherapy, prior treatment with anti-her agents, uncontrolled systemic conditions, pre-existing lung conditions, warfarin use Primary endpoint: PFS1 (time to RECIST PD or death) Secondary endpoints: PFS2 (time to off-erlotinib PD if erlotinib was extended beyond RECIST PD) PFS1 in exon 19 deletion/l858r subsets OS ORR/DCR/BOR safety ESMO 2014 Park et al

Survival probability PFS1 (time to RECIST PD or death) PFS1 assessed by investigator 1.0 N=176 PFS events 0.8 0.6 0.4 0.2 0.0 0 10.8 months (95% CI 9.2 11.1) 10 20 30 Progression-free survival time (months) Secondary endpoints, ITT population ORR : 66.2% DCR : 82.6% median OS: 31.0 months (95% CI 27.3 not reached) CI = confidence interval Data cut-off 14 Feb 2014

Erlotinib continuation post-pd Screened n=359 Enrolled n=208 ITT n=207 Safety n=207 Per protocol * n=148 ITT n=207 Patients with PFS1 RECIST PD event n=171 Patients not receiving post-pd erlotinib n=78 Patients receiving post-pd erlotinib n=93 Patients with PFS1 event of death n=5 Patients without PFS1 (still receiving firstline treatment) n=21 Patients withdrawn without PFS1 n=10 AE n=2 Withdrew consent n=6 Refused treatment n=2 Patients still on post-pd treatment n=14 Patients with PFS2 event n=70 Patients withdrawn n=9 AE n=1 Withdrew consent n=3 Other n=5 *Per-Protocol (PP) population is defined as those patients who have EGFR mutations confirmed by study designated central laboratory. Data cut-off 14 Feb 2014

Baseline characteristics according to post-pd erlotinib Characteristic Continued erlotinib beyond PD N=93 Did not continue erlotinib beyond PD * N=78 * Median age, years (range) 60.5 (36 83) 59.9 (31 89) Gender, n (%) Male Female 33 (35.5) 60 (64.5) 35 (44.9) 43 (55.1) Smoking status, n (%) Never smoker Former smoker Current smoker 68 (73.1) 16 (17.2) 9 (9.7) 54 (69.2) 15 (19.3) 9 (11.5) EGFR mutation Exon 19 deletion Exon 21 L858R Other (exon 18/21) 52 (55.9) 36 (38.7) 13 (14.0) 36 (46.2) 38 (48.7) 16 (20.5) Histology, n (%) Adenocarcinoma Squamous-cell Other 90 (96.8) 0 (0.0) 3 (3.2) 76 (97.4) 1 (1.3) 1 (1.3) Disease stage ECOG PS baseline 0 1 2 Recurrent Stage IV 15 (16.1) 78 (83.9) 15 (16.1) 73 (78.5) 5 (5.4) 3 (3.8) 75 (96.2) 14 (17.9) 54 (69.2) 10 (12.8) * only consider patients who had a PD event; n=9 in each group (n=93 and n=78) were evaluated by cytology not histology Data cut-off 14 Feb 2014

Survival probability PFS2 (time to off-erlotinib PD if erlotinib was extended beyond RECIST PD) Median PFS2 (n=93; 79 PD events): 14.1 months (95% CI 12.2 15.9) 1.0 0.8 0.6 0.4 0.2 0.0 0 14.1 months (95% CI 12.2 15.9) 10 20 30 Progression-free survival time (months) Data cut-off 14 Feb 2014

Survival probability Continuation of erlotinib post-pd extended PFS In patients receiving post-pd erlotinib (n=93) PFS1 was 11.0 months the difference between PFS1 and PFS2 was an additional 3.1 months 1.0 0.8 PFS1 PFS2 0.6 0.4 0.2 0.0 0 11.0 months 14.1 months 10 20 30 Progression-free survival time (months)

Options at RECIST progression RECIST PD to first line TKI IMPRESS ESMO LBA6544 Presidential Symposium 1 Sept 28 16:00 Change to doublet chemo Add doublet chemo and continue TKI

IMPRESS: Study Design Enrollment period: March 2012-December 2013 Patients Age 18 years ( 20 years in Japan) WHO PS 0-1 Histologically confirmed stage IIIB / IV EGFR mutationpositive advanced NSCLC Chemotherapy naive Achieved CR/PR 4 months or SD >6 months with first-line gefitinib Disease progression (RECIST) <4 weeks prior to study randomisation Cisplatin 75 mg/m 2 + Pemetrexed 500 mg/m 2 ( 6 cycles) + Gefitinib 250 mg 1:1 randomisation Cisplatin 75 mg/m 2 IV + Pemetrexed 500 mg/m 2 IV ( 6 cycles) + Placebo 250 mg Objectives Endpoints Primary Progression-free survival Secondary Overall survival Objective response rate Disease control rate Safety and tolerability Health-related quality of life b Mok et al EMSO 2014

Patient demographics (ITT) Demographic characteristic Gefitinib (n=133) Placebo (n=132) 65 years 32% 26% Mean age, years (range) 59 (33-79) 57 (35-79) Female 65% 64% PS 0, 1 41%, 59% 40%, 60% Never-smoker 66% 69% Adenocarcinoma histology 95% 99% Metastatic (baseline) 93% 90% Brain metastases (baseline) 33% 23% CR or PR/SD to first-line gefitinib 68% / 32% 76% / 24% Time to progression after initial gefitinib treatment 10 months >10 months 39% 61% 44% 56% Exon 19 deletion, exon 21 L858R 64%, 30% 65%, 32%

ORR (%) DCR (%) ORR and DCR (ITT population) Odds ratio (95% CI) = 0.92 (0.55, 1.55); p=0.760 Odds ratio (95% CI) = 1.39 (0.74, 2.62); p=0.308 (n=133) (n=132) (n=133) (n=132) Odds ratio >1 favours gefitinib Odds ratio and p-value from logistic regression with covariates CI, confidence interval

Probability of PFS PFS (primary endpoint; ITT) 1.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.0 Gefitinib (n=133) Placebo (n=132) Gefitinib (n=133) 0 2 4 6 8 10 12 14 Patients at risk: Time of randomisation (months) Gefitinib 133 110 88 40 25 12 6 0 Placebo 132 100 85 39 17 5 4 0 Placebo (n=132) Median PFS, months 5.4 5.4 Number of events, n (%) 98 (73.7) 107 (81.1) HR a (95% CI) = 0.86 (0.65, 1.13); p=0.273 a Primary cox analysis with covariates A HR <1 implies a lower risk of progression with gefitinib

Probability of OS 1.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.0 OS (ITT; 33% of events) Gefitinib (n=133) Placebo (n=132) Gefitinib (n=133) Placebo (n=132) Median OS, months 14.8 17.2 Number of events, n (%) 50 (37.6) 37 (28.0) HR a (95% CI) = 1.62 (1.05, 2.52); p=0.029 0 2 4 6 8 10 12 14 16 18 20 22 24 26 Patients at risk: Time of randomisation (months) Gefitinib 133 125 111 88 64 43 27 19 12 8 4 2 0 0 Placebo 132 129 119 94 76 55 39 27 16 10 7 4 2 0 a Primary cox analysis with covariates A HR <1 implies a lower risk of death with gefitinib

Post-discontinuation therapy (ITT) Anti-cancer therapy Gefitinib (n=133) ITT population Placebo (n=132) Any, n (%) 61 (45.9) 72 (54.5) Platinum alone, a n (%) 0 (0) 2 (1.5) Platinum-based regimen (doublet or triplet), n (%) Single-agent cytotoxic, b n (%) 5 (3.8) 17 (12.9) 27 (20.3) 27 (20.5) EGFR TKI, c n (%) 30 (22.6) 44 (33.3) Others, n (%) 16 (12.0) 14 (10.6) a Platinum: carboplatin, cisplatin, nedaplatin b Single agent: docetaxel, paclitaxel, pemetrexed, gemcitabine, vinorelbine c EGFR TKI (gefitinib, erlotinib, AZD9291 [n=2 gefitinib arm; n=3 placebo arm])

Optimal Strategy TKI Chemo 2 nd /3 rd line therapy Death TKI Treatment beyond progression (RECIST)

Optimal Strategy TKI Chemo 2 nd /3 rd line therapy Death TKI Treatment beyond progression (RECIST)

Optimal Strategies Finding the EGFR Mutation Choosing a EGFR TKI Adding to EGFR TKI Stopping the EGFR TKI

Optimal Strategies Finding the EGFR Mutation Choosing a EGFR TKI Adding to EGFR TKI Stopping the EGFR TKI

FASTACT-2 (MO22201; CTONG0902) study design Screening Study treatment Maintenance phase Previously untreated stage IIIB/IV NSCLC, PS 0/1 (n=451) Primary endpoint: PFS with IRC confirmation R Gemcitabine 1,250mg/m 2 (d1, 8) + carboplatin AUC=5 or cisplatin 75mg/m 2 (d1) + erlotinib 150mg/day (d15 28); q4wks x 6 cycles GC-erlotinib (n=226) 1:1; stratified by stage, histology, smoking status and chemo regimen Gemcitabine 1,250mg/m 2 (d1, 8) + carboplatin AUC=5 or cisplatin 75mg/m 2 (d1) + placebo (d15 28); q4wks x 6 cycles GC-placebo (n=225) Erlotinib 150mg/day Placebo Secondary endpoints: subgroup analyses, OS in all patients and subgroups, ORR, duration of response, TTP, NPR at 16 weeks, safety, QoL PD PD Erlotinib 150mg/day Wu and Mok Lancet Oncology 2013

Improved overall survival outcomes Wu and Mok Lancet Oncology 2013

PFS benefit confined to patients with EGFR mutations EGFR Mutation EGFR Wild type Wu and Mok Lancet Oncology 2013

OS benefit confined to patients with EGFR mutations EGFR Mutation EGFR Wild type Wu and Mok Lancet Oncology 2013

OS benefit confined to patients with EGFR mutations Need confirmation with FASTACT 3 using EGFR TKI as control arm EGFR Mutation EGFR Wild type Wu and Mok Lancet Oncology 2013

JO25567: Randomized phase II study on erlotinib + bevacizumab Assessed for eligibility Randomized (n = 154) EB combination (n = 77) E monotherapy (n = 77) Withdrew before treatment started Thrombosis: 1 Pleural effusion: 1 Received EB and eligible for analysis (n = 75) Received E and eligible for analysis (n = 77) Kato et al ASCO 2014

Tumor volume change from baseline (%) Tumor volume change from baseline (%) Objective tumor response EB (n = 75) E (n = 77) *P value CR 3 (4%) 1 (1%) PR 49 (65%) 48 (62%) SD 22 (29%) 19 (25%) PD 0 (0%) 6 (8%) NE 1 (1%) 3 (4%) ORR 69% 64% 0.4951 DCR 99% 88% 0.0177 *Fisher s exact test 100 100 60 EB combination 40 20 0 20 30 40 60 80 60 E monotherapy 40 20 0 20 30 40 60 80 Responder (CR or PR) Non-responder (SD, PD or NE) Median duration of response: 13.3 months with EB vs 9.3 months with E Presented by: Terufumi Kato

PFS probability Primary endpoint: PFS by independent review 1.0 0.8 0.6 0.4 EB E Median (months) HR 0.54 (95% CI: 0.36 0.79) P value* EB E *log-rank test, two-sided 0.2 0 0 9.7 16.0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 Number at risk Time (months) EB 75 72 69 64 60 53 49 38 30 20 13 8 4 4 0 E 77 66 57 44 39 29 24 21 18 12 10 5 2 1 0 Presented by: Terufumi Kato

PFS probability PFS probability 1.0 PFS by EGFR mutation type Exon 19 deletion EB E Median (months) 18.0 10.3 HR 0.41 (95% CI: 0.24 0.72) 1.0 Exon 21 L858R EB E Median (months) 13.9 7.1 HR 0.67 (95% CI: 0.38 1.18) 0.8 0.8 0.6 0.6 0.4 0.4 Number at risk EB E 0.2 0 0 40 EB E 2 4 6 8 10 12 14 16 18 20 22 24 26 28 Time (months) 39 38 36 33 29 27 24 19 12 8 5 2 2 0 40 35 29 26 22 16 12 9 9 5 3 1 0 0 0 E 0.2 Number at risk EB 0 0 35 37 EB E 2 4 6 8 10 12 14 16 18 20 22 24 26 28 Time (months) 33 31 28 27 24 22 14 11 8 5 3 2 2 0 31 28 18 17 13 12 12 9 7 7 4 2 1 0 Presented by: Terufumi Kato

PFS probability PFS probability 1.0 PFS by EGFR mutation type Exon 19 deletion EB E Median (months) 18.0 10.3 HR 0.41 (95% CI: 0.24 0.72) 1.0 Exon 21 L858R EB E Median (months) 13.9 7.1 HR 0.67 (95% CI: 0.38 1.18) 0.8 0.6 0.8 Chance of a phase III study is remote 0.6 0.4 0.4 Number at risk EB E 0.2 0 0 40 EB E 2 4 6 8 10 12 14 16 18 20 22 24 26 28 Time (months) 39 38 36 33 29 27 24 19 12 8 5 2 2 0 40 35 29 26 22 16 12 9 9 5 3 1 0 0 0 E 0.2 Number at risk EB 0 0 35 37 EB E 2 4 6 8 10 12 14 16 18 20 22 24 26 28 Time (months) 33 31 28 27 24 22 14 11 8 5 3 2 2 0 31 28 18 17 13 12 12 9 7 7 4 2 1 0 Presented by: Terufumi Kato

Addition of chemotherapy or bevacizumab to TKI may prolong PFS but need to be validated by phase III study

Optimal Strategies Finding the EGFR Mutation Choosing a EGFR TKI Stopping the EGFR TKI Treating the resistance Adding to EGFR TKI

What we know about the mechanism of resistance? Oxnard et al CCR 17:5530, 2011

Gatekeeper Mutation: T790M Acquired point mutation resulting in threonine-tomethioine amino acid change at positive 790 Kobayashi S NEJM 352:786, 2006

SLCG: Implication of de-novo T790M Rosell et al ASCO 2010

Implication of acquired T790M Median T790M pos = 19 months Median T790M neg = 12 months Median T790M pos = 39 months Median T790M neg = 26 months Oxnard et al CCR 17:1616, 2011

Targeting T790M T790M

Third generation EGFR TKI targeting the resistant T790M mutation Erlotinib Gefitinib CO1686 WZ4002

In vitro and In vivo activity of AZD9291 AZD9291 is a potent oral, irreversible inhibitor of EGFR that contains EGFR-TKI-sensitising (EGFR+) and resistance mutations (T790M) Updated long-term dosing of H1975 (L858R/T790M) xenograft with indicated doses of AZD9291 Good potency and high selectivity demonstrated in enzymatic and cellular in vitro assays 1 Model AZD9291 phospho- EGFR IC 50 nm Wildtype LoVo cells EGFR+ PC9 cells EGFR+/ T790M H1975 cells 480 17 15 1. Cross et al. Abstract A109, AACR-EORTC-NCI conference, Boston, 2013 Profound regression in EGFR-mutant tumour models, showing sustainable complete macroscopic tumour response out to at least 200 days Ranson et al WCLC 2013

Phase I/II study on AZ9291 Phase I, open-label, multicenter study of AZD9291 administered once daily in Asian and Western patients with advanced NSCLC who have documented radiological progression while on prior therapy with an EGFR-TKI (AURA; NCT01802632) Objectives Primary: safety and tolerability in EGFR-TKI-refractory patients Secondary include: define maximum tolerated dose, pharmacokinetics, preliminary efficacy Escalation Not preselected by T790M status Cohort 1 20 mg Cohort 2 40 mg Rolling six design Cohort 3 80 mg Cohort 4 160 mg Cohort 5 240 mg Expansion Enrollment by local testing followed by central laboratory confirmation* of T790M status or by central laboratory testing alone T790M+ T790M+ T790M- T790M+ T790M- 1st-line EGFRm+ T790M+ T790M- 1st-line EGFRm+ T790M+ Biopsy Biopsy *cobas EGFR Mutation Test (Roche Molecular Systems) Janne ASCO 2014 Tablet

Response rate* in overall population 40 20 ####### Best percentage change from baseline in target lesion: all evaluable patients, escalation and expansion (N=205) 0-20 -40-60 Complete response Partial response* Non-response -80-100 First patient dosed Mar 6, 2013 Longest response >9 months ongoing at time of data cutoff ORR* = 53% (109/205; 95% CI 46%, 60%); no difference in ORR by race Overall disease control rate (CR+PR+SD) = 83% (171/205; 95% CI 78%, 88%) 20 mg 40 mg 80 mg 160 mg 240 mg N (205) 20 57 61 55 12 ORR 55% 44% 54% 58% 67% *Includes confirmed responses and responses awaiting confirmation; # represents imputed values. Population: all dosed patients with a baseline RECIST assessment and an evaluable response (CR, PR, SD or PD), N=205 (from 232 dosed patients, 27 patients with a current nonevaluable response are not included). CI, confidence interval; CR, confirmed complete response; ORR, overall response rate; PR, confirmed partial response; PD, progressive disease; RECIST, Response Evaluation Criteria In Solid Tumors; SD, stable disease Presented by: Pasi A. Jänne

40 20 # # D D Response rate* in central T790M+ Best percentage change Best from percentage baseline in change target lesion: from baseline all evaluable in target T790M+ lesion: patients, Part B T790M+ evaluable patients, expansion cohorts only (N=107) 0-20 D D D D D D D D D -40-60 -80 20 mg QD 40 mg QD 80 mg QD 160 mg QD 240 mg QD D D D D D D D D D D D D -100 ORR* = 64% (69/107; 95% Cl 55%, 73%) in patients with EGFR T790M+ NSCLC Overall disease control rate (CR+PR+SD) = 94% (101/107; 95% CI 88%, 98%) 20 mg 40 mg 80 mg 160 mg 240 mg N (107) 10 29 34 28 6 ORR 50% 62% 68% 64% 83% *Includes confirmed responses and responses awaiting confirmation; # represents imputed values. Population: all dosed central T790M+ patients with a baseline RECIST assessment and an evaluable response (CR/PR, SD or PD), N=107 (from 120 T790M+ patients, 13 patients with a current non-evaluable response are not included). QD, once daily; central T790M+, T790M positive by central laboratory testing Presented by: Pasi A. Jänne

CO-1686 Phase I Study: Dose Levels Presented By Lecia Sequist at 2014 ASCO Annual Meeting

CO-1686 Phase I patient characteristics Presented By Lecia Sequist at 2014 ASCO Annual Meeting

Adverse events Presented By Lecia Sequist at 2014 ASCO Annual Meeting

Best response in Phase 1 and early <br />Phase 2 expansion cohort patients Presented By Lecia Sequist at 2014 ASCO Annual Meeting

Sum of target lesions over time Presented By Lecia Sequist at 2014 ASCO Annual Meeting

Both AZ9291 and CO1686 are on fast track approval by FDA TIGER 2 study: Single arm phase II study of CO1686 for patient with T790M resistant mutation after first TKI failure AURA 2 study: Single arm phase II study of AZ 9291 for patient with T790M resistant mutation after first line TKI failure

AURA 3 Study Design Randomise ~610 patients 2:1 Central testing of ~ 1540 biopsy samples T790M+ (n=610) AZD9291 (80 mg p.o. qd) (n=407) Platinum-based doublet chemotherapy* every 3 weeks (n=203) Primary endpoint: PFS T790M- Not eligible for enrolment *Pemetrexed 500 mg/m 2 + carboplatin AUC5 or Pemetrexed 500 mg/m 2 + cisplatin 75 mg/m 2 P PI: T Mok YL Wu AUC5, area under the plasma concentration time curve 5 mg/ml 1 per minute; EGFRm+, EGFR mutation-positive; EGFR-TKI, EGFR tyrosine kinase inhibitor; NSCLC, non-small cell lung cancer; p.o., orally; qd, once daily; T790M+, T790M mutation-positive; T790M-, T790M mutation-negative

Summary Advanced stage NSCLC (2014) EGFR mut Gefitinib or Erlotinib or Afatinib PD Chemo +/- Bev

Summary Advanced stage NSCLC EGFR mut (by tissue or blood) Gefitinib or Erlotinib or Afatinib or Dacomitinib Continue TKI beyond RECIST PD T790m+ AZ9291/ CO1686 PD T790m- Chemo +/- Bev Chemo +/- Bev

Optimal shape

Questions

CARE Lung Cancer Faculty The CARE (Community Academic Research Education) Faculty is a Pan-Canadian group of medical oncologists from across Canada who meet and discuss various topics from key conferences. The CARE Lung Cancer Faculty is comprised of leading medical oncologists from across Canada and includes Drs. Barbara Melosky (BC), Anil A. Joy (AB), Denis Soulieres (QC), Natasha Leighl (ON), Sunil Verma (ON) & Ron Burkes (ON)

Canadian Perspectives & Interactive Discussion

Thank You!