Molecular Basis of Lung Cancer Therapy James R. Rigas Comprehensive Thoracic Oncology Program Traditional View of Lung Cancer Small Cell Large Cell Adenoca Squamous Presentation • EGFR mutations – RADIANT adjuvant results in EGFR mutations – Uncommon EGFR mutations – Resistance in EGFR mutations (+) patients • ALK gene rearrangements – Resistance in ALK gene rearrangement (+) patients • BRAF mutations • Other mutations, fusions and amplifications Locations and Types of the 134 EGFR Gene Mutations Detected in Lung Cancers Del 19 T790M L858R Shigematsu H et al. JNCI J Natl Cancer Inst 2005;97:339-346 Journal of the National Cancer Institute, Vol. 97, No. 5, © Oxford University Press 2005, all rights reserved. EURTAC Study Design Stage IIIB/IV NSCLC EGFR exon 19 deletion or exon 21 L858R mutation (DNA sequencing/Genescan and Taqman) Chemonaive ECOG PS 0–2 Measurable or evaluable disease Primary endpoint • Progression-free survival (PFS) US FDA approval May 14, 2013 Cobas® EGFR Mutation Test 41 mutations in Exons 18, 19, 20 and 21 Erlotinib 150 mg/day R PD Stratification • Mutation type • ECOG PS (0 vs 1 vs 2) Platinum-based doublet chemotherapy q3wks x 4 cycles* Secondary endpoints • Objective response rate • Overall survival (OS) • Location of progression • Safety • EGFR mutation analysis in serum • Quality of life ECOG = Eastern Cooperative Oncology Group; PS = performance status; PD = progressive disease *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 PD Primary endpoint: PFS in ITT population (updated analysis 26 Jan 2011) 1.0 Erlotinib (n=86) Chemotherapy (n=87) PFS probability 0.8 HR=0.37 (0.25–0.54) 0.6 Log-rank p<0.0001 0.4 0.2 5.2 0 0 9.7 3 6 9 12 63 49 54 20 32 8 21 5 15 18 Time (months) 21 24 27 30 33 7 1 4 0 2 0 2 0 0 0 Patients at risk Erlotinib Chemo 86 87 17 4 9 3 Data cut-off: 26 Jan 2011 US FDA approval May 14, 2013 Study design 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) Race (Asian/non-Asian) Afatinib 40 mg/day† Cisplatin + Pemetrexed 75 mg/m2 + 500 mg/m2 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, PK *EGFR29:19 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: Q-5D, EORTC QLQ-C30 and QLQ-LC13 at randomization and q3 weeks until progression or new anti-cancer therapy. US FDA approval July 12, 2013 Yang JC, et al. EGFR Gene Mutations in Adenocarcinoma Lung Cancer 1% 1% 2% 3% 2% 0% 1% 1% 0% 0% n = 345 40% 49% N=138 N=170 Del 19 89% L858R Exon 20 ins L858R + T790M L861Q G719X Del19 + T790M T790M L858R + S768I G719X + S768I G719X + T790M S768I Therascreen ® EGFR (29) RGQ PCR Kit PFS: Common mutations (Del19/L858R) Independent review – patients with Del19/L858R (n=308) Progression-free survival (probability) 1.0 Afatinib n=204 Cis/pem n=104 PFS event, n (%) 130 (64) 0.8 Median PFS (months) 13.6 Hazard ratio (95% confidence interval) 0.6 61 (59) 6.9 0.47 (0.34–0.65) p<0.0001 51% 0.4 0.2 21% 0.0 0 Number at risk Afatinib 204 Cis/Pem 104 3 6 169 62 143 35 US FDA approval July 12, 2013 9 12 15 18 Progression-free survival (months) 115 17 75 9 49 6 Yang JC, et al. 30 2 21 24 27 10 2 3 0 0 0 RADIANT Study Schematic P a tie n ts w ith S ta g e IB -IIIA N S C L C T u m o r T is s u e A n a ly s is EG FRN e g a tiv e b y b o th IH C a n d F IS H In e lig ib le C o m p le te S u rg ic a l R e s e c tio n S c re e n in g C o n s e n t EG FRP o s itiv e b y IH C a n d /o r F IS H N o C h e m o th e ra p y o r U p to 4 C y c le s o f a S ta n d a rd (n o n -in v e s tig a tio n a l), P la tin u m -b a s e d , A d ju v a n t C h e m o th e ra p y R e g im e n T re a tm e n t C o n s e n t R a n d o m iz a tio n (2 :1 ) 1 5 0 m g /d a y T a rc e v a P la c e b o 2 y rs o r u n til o n e o f th e fo llo w in g : re la p s e , d e a th , p a tie n t re q u e s t o r in v e s tig a to r d e c is io n to d is c o n tin u e s tu d y d ru g th e ra p y , o r in to le ra b le to x ic ity F o llo w -u p V is it O n c e E v e r y 6 M o n th s fo r U p to 5 Y e a rs a fte r R a n d o m iz a tio n , th e n Y e a rly RADIANT Study Results Hierarchical testing rendered all secondary endpoints non-significant. LUX-Lung clinical trials and eligibility 2 LUX-Lung 2 LUX-Lung 3 LUX-Lung 6 Phase II Phase III Phase III N=129 N=345 N=364 Treatment Afatinib Afatinib vs. Pemetrexed/ cisplatin Afatinib vs. Gemcitabine/ cisplatin Line of treatment First- and second-line (after chemo) First-line First-line Mutation test Direct sequencing (central) EGFR29* (central) EGFR29* (central) *EGFR mutations detected by TheraScreen EGFR29 test: – Common: 19 deletions in exon 19 and L858R in exon 21 – Uncommon: 3 insertions in exon 20, L861Q, T790M, G719S, G719A and G719C, S768I EGFR mutation-positive patients in LUX-Lung trials 3 LUX-Lung 2 LUX-Lung 3 LUX-Lung 6 Phase II Phase III Phase III N=129 N=345 N=364 Del19 n=408 n=52 n=170 n=186 L858R n=330 n=54 n=138 n=138 Uncommon n=100 n=23 n=37 n=40 Patients with uncommon mutations treated with afatinib Uncommon n=75 n=23 n=26 n=26 Subgroups of patients with uncommon mutations 5 Categories n= Mutations (n) De novo T790M Exon 20 insertions Other (exon 18, 19, 20, 21) 14 23 38 T790M alone (3) T790M+Del19 (3) T790M+L858R (6) T790M+G719X (1) T790M+L858R+G719X (1) n/a L861Q alone (12) G719X alone (8) G719X+S768I (5) G719X+L861Q (3) E709G or V+L858R (2) S768I+L858R (2) S768I alone (1) L861P alone (1) P848L alone (1) R776H+L858R (1) L861Q+Del19 (1) K739_1744dup6 (1) Tumour shrinkage in patients with uncommon mutations Independent review (n=67†) 120 * 7 De novo T790M (n=14): T790M alone(*), T790M+Del19, T790M+L858R, T790M+G719X, T790M+L858R+G719X Maximum change from baseline (%) 100 Exon 20 insertions (n=20) Other (n=33): 80 L861Q, G719X, G719X+S768I, G719X+L861Q, E709G or V+L858R, S768I+L858R, S768I, L861P, P848L, R776H+L858R, L861Q+Del19, K739_1744dup6 60 40 20 0 * * -20 -40 -60 -80 -100 †8 patients were not included due to insufficient data T790M+ EGFR- TKI failures 2nd/3rd line NSCLC An area of high unmet medical need There are no current proven treatment options for these patients. Chemotherapy or EGFR-TKIs are used, but their benefit isn’t proven EGFR TKI resistance mechanisms AZD9291 or CO1686 “Patients with EGFR-mutant lung adenocarcinoma develop acquired resistance to EGFR tyrosine kinase inhibitors (TKIs) after a median of 10-16 months. In half of these cases, a second EGFR mutation, T790M, underlies acquired resistance” Oxnard, et al. Clin Cancer Res Dec 6, 2010 Clinical activity AZD9291 in T790M+ Fast Track status granted by FDA in October 2013 Breakthrough Therapy Designation submission early February 2014 Best % change from baseline in target lesions in central testing T790M+ T790M+ Response Rate* = 64% (95% CI 53%-74%) For the 89 T790M+ evaluable patients there are 57 PRs (35 confirmed), 28 SDs, 4 PDs Population: T790M+ patients with observed or imputed target lesion data (n=89) *Confirmed+ unconfirmed response Response ongoing for 34/35 T790M+ patients with confirmed PR D Discontinued treatment * Imputed T790M status is assigned by central testing of a recent tumour sample Independent review of scans is underway as per FDA request 41/43 confirmed so far Preliminary data, data cut off 16th January 2014 CO1686 FISH Assay for ALK Rearrangement* p25.2 p25.2 p24.3 p24.1 p23.2 p22.3 p22.1 p16.3 ALK 29.3 p24.3 EML4 42.3 p16.3 Telomere 2p23 region p24.1 p23.2 p22.3 p22.1 p16.1 p16.1 p14 p13.2 p14 p13.2 p12 p12 q12.1 q12.3 q12.1 q12.3 q14.1 q14.1 q14.3 q21.2 q14.3 q21.2 q22.1 q22.2 q23.2 q22.1 q22.2 q23.2 q24.1 q24.1 q24.3 q24.3 q31.3 q31.3 q32.1 q32.1 q32.3 q32.3 q33.2 q33.2 q34 q34 q36.1 q36.3 q37.2 q36.1 q36.3 q37.2 Centromere t(2;5) ALK gene breakpoint region 3’ 5’ ~250 kb ~300 kb Break-apart FISH assay for ALK-fusion genes1 Non-split signal Split signal ALK break-apart FISH assay [Courtesy John Iafrate, Massachusetts General Hospital] *Assay is positive if rearrangements can be detected in ≥15% of cells FISH = fluorescence in situ hybridization 1Shaw AT et al. J Clin Oncol 2009;27:4247–4253 Crizotinib: First-in-human/Patient Trial Cohort 5 (n=6) 2 DLTs: grade 3 fatigue 300 mg BID Part 1: Dose escalation Cohort 6 (n=9) Cohort 4 (n=7) 250 mg BID MTD/RP2D 200 mg BID Cohort 3 (n=8) 1 DLT: grade 3 ALT elevation 200 mg QD Cohort 2 (n=4) 100 mg QD Cohort 1 (n=3) 50 mg QD ALT = alanine aminotransferase Part 2: Molecularly enriched cohorts (ALK and c-MET) Enrolling patients with ALK-positive NSCLC after preliminary observation of impressive activity in a few patients • Data from database April 7, 2010 • Data presented for 82 patients, study ongoing Tumor Responses to Crizotinib for Patients with ALK-positive NSCLC Maximum change in tumor size (%) 60 Progressive disease Stable disease 40 Confirmed partial response Confirmed complete response 20 0 –20 –30% –40 –60 –80 –100 US FDA accelerated approval Aug 26, 2011 Full approval Nov 21, 2013 * *Partial response patients with 100% change have non-target disease present U Expansion Phase NCT01283516 Advanced ALK-rearranged malignancies Completed n=59 Dose escalation started at 50 mg/day Escalate to MTD (750 mg/day) ALK-rearranged NSCLC Additional n=71 enrolled Crizotinib naive Crizotinib pretreated Other ALKactivated tumors Primary objective: determination of MTD Secondary objectives: characterize safety, PK, and antitumor activity Patients received treatment until disease progression, unacceptable toxicity, or withdrawal of consent Continuous oral dosing 21-day cycles Dose-Escalation Phase Study Design – LDK378 (Ceritinib) Shaw AT et al. N Engl J Med. 2014;370:1189-1197. Expires April 2015 MED.ONC.LDK.U.EISEX 25 U Best Change (%) in Tumor Response from Baseline in NSCLC Patients Treated with Ceritinib Best Change (%) from Baseline 100 Crizotinib Pretreated Crizotinib Naive PFS Event 80 60 40 20 0 -20 -40 -60 -80 US FDA accelerated approval Apr 30, 2014 -100 Based on investigator assessment of response. PFS, progression-free survival. Shaw AT et al. N Engl J Med. 2014;370:1189-1197. From N Engl J Med, Shaw AT, Kim DW, Mehra R, et al, Ceritinib in ALK-Rearranged Non–Small-Cell Lung Cancer, Vol 370, Page 1194. Copyright © 2014 Massachusetts Medical Society. Reprinted with permission from Massachusetts Medical Society. Expires April 2015 MED.ONC.LDK.U.EISEX 26 BRAF-Mutations in NSCLC W. Pao, N. Girard Lancet Oncology, February 2011 A. Marchetti et al., J. Clin. Oncol. 29, 1, 2011 • 1,046 NSCLC samples • Frequency of BRAF-mutations 3.5% • BRAF-mut. in 4.9% adeno- and 0.3% SCC • 57% BRAFV600E-mutations • 43 % non-activating BRAF mutations P. Paik et al., J. Clin. Oncol. 29, 2046, 2011 • 697 NSCLC-adeno. ca. Samples • Frequency of BRAF-mutations 3% 1.5-2.5 % of NSCLC harbor BRAF V600E-mutations • 50% BRAFV600E-mutations • 50% non-activating BRAF mutations BRF113928: Initial Study Design • Single arm, Phase II, open label • Green-Dahlberg 2-stage: H(0): ORR ≤ 10% versus H(1): ORR ≥30% Primary objective: Investigator-assessed ORR NSCLC (Adenocarcinoama) BRAFV600Emutation N = 40 dabrafenib 150 mg twice daily ≥ 2nd Line Secondary objectives: PFS, duration of response, OS, safety and tolerability, population PK Interim Analysis (ASCO 2013) US FDA granted Breakthrough Therapy Jan 13, 2014 Investigator based response assessments demonstrated: 7PRs (5 confirmed—duration 29 and 49 weeks for 2 pts and remaining 3 patient 6+ to 24+ weeks) 1 stable disease 4 progressive disease Planchard et. al. 2013 ASCO Annual Meeting Proceedings Rationale for the Combination Sustained target inhibition Delay and potentially to observe more prolonged prevent the development of and durable anti-tumor resistance effect RAS BRAFi mutBRAF + MEKi MEK pERK Proliferation, survival Invasion , Metastasis Prevent/delay hyperproliferative lesions and secondary malignancies (Cu SCC) BRAF V600 BRAF WT Molecular Subsets Adenocarcinoma Lung Cancer NRAS MAP2K1 ROS1 fusions AKT1 KIF5B-RET PIK3CA BRAF HER2 ALK Fusions Unknown EGFR KRAS 1. Mascaux C et al. Br J Cancer 2005;92:131–9; 2. Winton et al. N Engl J Med 2005;352:2589–97; 3. Eberhard et al JCO 2005;23:5900–9; 4. Pao et al. PLOS Medicine 2005 2(1): e17; 5. Pao et al. Nat Med 2012;18:349–51; 6. Kris et al. ASCO 2011 Structural variants • • • Translocations Fusions Inversion Copy number alterations • • • Amplifications Deletions LOH Point mutations & indels • • • • Missense Nonsense Splice site Frameshift Gene expression • • • Outlier expression Isoform usage Pathways & signatures Wild type AGTGA Mutant AGAGA Adapted from: Roychowdhury et al. Sci Transl Med; 20122 32 Conclusion • Molecular selection of patients improves therapy outcomes for patients with advanced adenocarcinoma of the lung Future Directions • Extent patient selection and targeted therapies to squamous cell and earlier stages of NSCLC (i.e. RADIANT) • Next generation sequencing needed • Molecular characterization of resistance • Development of a structure to conduct studies in uncommon molecular selected populations (BRAF, HER2, RET, etc) • Clinical integration of tumor genomic and proteomic testing to better direct therapy for NSCLC (Gerber)