1 / 37

Acquired Resistance to EGFR TKIs: What Is It and How Do We Treat?

Acquired Resistance to EGFR TKIs: What Is It and How Do We Treat?. Vincent A Miller, MD Thoracic Oncology Service Memorial Sloan-Kettering Cancer Center. Disclosure Slide. Background. Patients with CML and GIST have a high rate of response to treatment with imatinib.

nelson
Download Presentation

Acquired Resistance to EGFR TKIs: What Is It and How Do We Treat?

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Acquired Resistance to EGFR TKIs: What Is It and How Do We Treat? Vincent A Miller, MDThoracic Oncology ServiceMemorial Sloan-Kettering Cancer Center

  2. Disclosure Slide

  3. Background • Patients with CML and GIST have a high rate of response to treatment with imatinib. • These patients commonly acquire resistance after an initial response. • There are a small number of conserved changes in the BCR-ABL tyrosine kinase domain which confer resistance in vivo. • Other patients with acquired resistance have amplification of the BCR-ABL gene. • Virtually all patients with response to gefitinib or erlotinib eventually have progression of disease.

  4. Acquired Resistance to TKI Pre-rx 4M 19M 23M 25M 21M bx

  5. Clinical Definition of Acquired Resistance to EGFR TKIs in Lung Adenocarcinoma • Prior treatment with single agent TKI • Either: • EGFR ex 19 del, L858R, G719X, L861Q • PR or CR with TKI or SD lasting ≥6 months • PD by RECIST or WHO in previous 30 days • No therapy after TKI before start of new agent • “Washout” after stopping TKI not >5 half-lives of the TKI (14 days for gefitinib or erlotinib) • Obtain baseline scan on day 1 Jackman J Clin Oncol 2009

  6. Trials to Overcome Acquired Resistance * The number of patients enrolled into the trial with AR to EGFR TKIs was low.

  7. T790M in Acquired Resistance • Acquired exon 20 mutation found in >50% of pts with acquired resistance to TKI • Increases relative affinity of mutant EGFR for ATP, may also cause steric hindrance • Less commonly detected in CNS, thought to be due to poor CNS penetrance of TKI

  8. Overall Survival from Start of TKI 100 80 60 40 20 0 Survival of TKI_OS: Survival proportions OS_t790MOS_neg HR 0.46, p = 0.006 Percent survival Median OS T790M-positive = 47 months Median OS T790M-negative = 26 months 0 50 100 150 TKI_OS Courtesy of VA Miller.

  9. Survival from Progressive Disease 100 80 60 40 20 0 Survival of PPS: Survival proportions PPS_t790MPPS_neg HR 0.59, p = 0.057 Percent survival Post-PD survival T790M-positive = 22 months Post-PD survival T790M-negative = 14 months 0 20 40 60 80 PPS Courtesy of VA Miller.

  10. T790M as a Biomarker T790M-type resistance Longer survival Later metastases Indolent growth Sensitivity to 2nd-line EGFR inhibitors? Non-T790M-type resistance Poorer survival Earlier metastases More aggressive? Needs mechanisms better elucidated and alternate therapies?

  11. Afatinib (BIBW 2992) — Irreversible Dual EGFR/HER2 Inhibitor • Anilino quinazoline derivative • Activity versus T790M (100 nM) • Binds covalently to Cys773 of the EGFR and Cys805 of HER2 F F S N C l N C l N N N N N N N S O O O O N O N N O O O EGFR - Cys773 HER2 - Cys805 Solca F et al. 17th EORTC-NCI-AACR Symposium on “Molecular Targets and Cancer Therapeutics” Philadelphia, PA, USA, November 14 – 18, 2005.

  12. LUX-Lung 1: Trial Design • Patients with: • Adenocarcinoma of the lung • Stage IIIB/IV • Progressed after one or two lines of chemotherapy (incl one platinum-based regimen) and ≥12 weeks of treatment with erlotinib or gefitinib • ECOG 0–2 • N = 585 Randomization 2:1 (Double blind) Oral afatinib 50 mg once daily plus BSC Oral placebo once daily plus BSC Primary endpoint: Overall survival (OS) Secondary: PFS, RECIST response, QoL (LC13 & C30), safety • Radiographic assessments at 4, 8, 12 wks and every 8 wks thereafter • Exploratory biomarkers: • Archival tissue testing for EGFR mutations (optional; central lab) • Serum EGFR mutational analysis (all patients)

  13. PFS by Independent Review 1.0 0.8 0.6 0.4 0.2 0.0 Placebo, PFS events = 133, median = 1.1 months (95% CI: 0.95-1.68) Afatinib, PFS events = 275, median = 3.3 months (95% CI: 2.79-4.40)Hazard ratio (95% CI) = 0.38 (0.306, 0.475)Log-rank test p-value <0.0001 Estimated PFS probability 0 3 6 9 12 15 18 PFS time since randomization (months) Number at risk 195 26 4 2 390 152 65 16 9 3 Miller VA et al. Presentation. ASTRO Thoracic Symposium 2010.

  14. Primary Analysis: Overall Survival Placebo, deaths = 114 (58.5%), median = 11.96 months (95% CI: 10.15-14.26)Afatinib, deaths = 244 (62.6%), median = 10.78 months (95% CI: 9.95-11.99)Hazard ratio (afatinib vs placebo) = 1.077 (0.862, 1.346)Log-rank test p-value (one-sided) = 0.7428 1.0 0.8 0.6 0.4 0.2 0.0 Estimated survival probability 0 3 6 9 12 15 18 21 24 Time to death since randomization (months) Number at risk 195 169 142 112 65 33 18 5 390 344 283 217 122 69 32 12 Miller VA et al. Presentation. ASTRO Thoracic Symposium 2010.

  15. Summary – HOPP Lab Animal Studies (number in parentheses = number of mice treated and assessed)

  16. Hypothesis We hypothesized that the combination of afatinib and cetuximab would overcome acquired resistance to erlotinib or gefitinib in patients with non-small cell lung cancer (NSCLC)

  17. Methods: Study Design • Phase Ib, open-label, multicenter trial in the US and the Netherlands • Primary objective: Maximum tolerated dose (MTD) and recommended Phase II dose (RPIID) for afatinib and cetuximab in patients with acquired resistance to erlotinib and gefitinib • Primary endpoint: • Occurrence of dose-limiting toxicity (DLT) • Secondary endpoints include: • Safety as assessed by National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) grading system • Objective response by Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 assessed at weeks 4, 8 and 12, and every 8 weeks thereafter

  18. Methods • Definitions of DLTs (CTCAE V3): • Grade ≥2 decrease in cardiac left ventricular function • Grade ≥2 diarrhea lasting for ≥7days, despite anti-diarrheal therapy • Grade ≥3 rash or nausea/vomiting despite medical management • Grade ≥3 fatigue lasting for ≥7 days • Grade 3 or 4 hypomagnesemia with clinically significant sequelae • All other toxicities of CTCAE Grade ≥3 (except alopecia and allergic reaction) leading to an interruption of afatinib/cetuximab for ≥14 days until recovery

  19. Study Schema Dose escalation schema 3-6 patients per cohort Afatinib PO daily + escalating doses of intravenous (IV) cetuximab q 2 weeks Dose levels starting at: afatinib 40 mg + cetuximab 250 mg/m2 Predefined maximum dose: afatinib 40 mg +cetuximab 500 mg/m2 NSCLC withEGFR mutation AND Stable disease(SD) ≥6 months on erlotinib/gefitinib OR Partial orcomplete responseto erlotinib/gefitinib Stop erlotinib/gefitinib for≥72 hours Diseaseprogression Expansion cohort part MTD cohort expandedup to 80 EGFR mutation-possible patients:40 T790M-positive and 40 T790M-negative

  20. Inclusion criteria: Pathologically confirmed NSCLC Presence of EGFR drug-sensitizing mutations1 or RECIST response, or SD ≥6 months Systemic progression of disease on continuous treatment with erlotinib or gefitinib within 30 days No systemic therapy between cessation of gefitinib/erlotinib and initiation of the study treatment Eastern Cooperative Oncology Group (ECOG) performance status (PS) 0-2 Age ≥18 years Exclusion criteria: Prior treatment with EGFR targeting antibodies Prior severe infusion reaction to a monoclonal antibody Patients with disease progression only in the central nervous system Symptomatic brain metastases Patient Eligibility 1 EGFR G719X, exon 19 deletion, L858R, L861Q mandated in MTD expansion cohort

  21. Patient Characteristics *Ongoing trial – T790M status not available in all patients

  22. Dose cohorts tested in the dose escalation: Afatinib 40 mg + 250 mg/m2 cetuximab (n = 4) Afatinib 40 mg + 500 mg/m2 cetuximab (n = 6) No DLT occurred in cycle 1 (28 days) Pre-defined MTD = RPIID: Afatinib 40 mg daily + cetuximab 500 mg/m2 47 patients have been enrolled in the MTD expansion cohort to date out of 80 planned 12 (26%) patients discontinued due to progression of disease Six (13%) patients discontinued due to toxicity Results

  23. Afatinib + Cetuximab at MTD: Responses by Mutation T790M+ T790M- No mutation Uninformative 70 60 50 40 30 20 10 0 -10 -20 -30 -40 -50 -60 -70 -80 -90 -100 Maximum percentage decrease from baseline (%) 0 4 8 12 16 20 24 28 32 36 40 44 48 Patient index sorted by maximum % decrease With permission from Janjigian Y et al. Proc ASCO 2011;Abstract 7525.

  24. Afatinib + Cetuximab at MTDResponses by Mutation *Two patients were not evaluable for efficacy

  25. Case Presentation • Middle-aged female diagnosed with Stage III-A lung adenocarcinoma in 2004 • 10 pack-yr smoker, no symptoms • Treated with chemotherapy, lobectomy and adjuvant RT followed by adjuvant erlotinib • May 2007 isolated bone met, treated with bisphosphonate, analgesics • May 2008 XRT L hip • June 2008 renal metastasis Jackman J Clin Oncol 2009

  26. Case Presentation • Erlotinib continued • February 2009 clinical trial with dasatinib – POD • May 2009 pemetrexed added and then bevacizumab added – initial response then POD in October 2010 • KPS 60-70% • Left hip pain, nausea, malignant pleural effusion, nausea, hematuria • Opiates, oxygen required

  27. Case Presentation (cont’d) • Commenced afatinib and cetuximab • Nausea resolved; weight gain • Opiates stopped, no need for wheelchair or cane • Hematuria absent • KPS 80% • Tox – Grade 1 rash, xerosis, paronychia

  28. 9/14/10 10/11/10

  29. What Causes Non-T790M Resistance? • 47 of 93 patients (51%) had tissue available for MET FISH • 9 patient specimens failed testing • 4 of 38 patients (11%) had MET:CEP7 ratio >2 • 1 pt with high level amplification + T790M • 2 of 3 pts with low level amplification had T790M • Is FISH the optimal way to test for MET amplification?

  30. ARQ 197-209: Study DesignRandomized, placebo-controlled, double-blind clinical trial R A N D O M I Z E Erlotinib 150 mg PO QD + ARQ 197 360 mg PO BID 28-day cycle • NSCLC • Inoperable locally adv/metastatic dz • ≥1 prior chemo (no prior EGFR TKI) PD Erlotinib 150 mg PO QD + placebo 28-day cycle • Endpoints • 1° PFS • 2° ORR, OS • Subset analyses • Crossover: ORR • 33 sites in 6 countries • Study accrual over 11 months (10/08-9/09) • Randomization stratified by prognostic factors incl sex, age, smoking, histology, performance status, prior therapy and best response, and geography (US vs ex-US) With permission from Schiller JH et al. Proc ASCO 2010;Abstract LBA7502.

  31. ARQ 197-209: Progression-Free Survival (ITT Population) 1.0 • HR = 0.81 (95% CI: 0.57, 1.15); p = 0.24 • Adjusted HR = 0.68 (95% CI: 0.47, 0.98); p < 0.05* 0.9 0.8 0.7 Erlotinib + ARQ 197 16.1 wks(n = 84) 0.6 Proportion of patients progression-free 0.5 0.4 0.3 0.2 Erlotinib + placebo 9.7 wks(n = 83) 0.1 0 50 0 10 20 30 40 Time from randomization (weeks) • * Cox regression model • PFS also measured by independent radiographic review: • - median 15.6 vs 8.4 wks • - unadjusted/adjusted HR = 0.74/0.51 With permission from Schiller JH et al. Proc ASCO 2010;Abstract LBA7502.

  32. ARQ 197-209: Overall Survival (ITT Population) 1.0 • HR = 0.88 (95% CI: 0.60, 1.3); p = 0.50 • Adjusted HR = 0.88 (95% CI: 0.6, 1.3); p = 0.52* 0.9 0.8 Erlotinib + ARQ 197: 36.6 wks(n = 84) 0.7 0.6 Proportion of patients surviving 0.5 0.4 Erlotinib + placebo: 29.4 wks(n = 83) 0.3 0.2 0.1 0.0 0 10 20 30 40 50 60 70 Survival time (weeks) *Cox regression model With permission from Schiller JH et al. Proc ASCO 2010;Abstract LBA7502.

  33. MetMAb Is an Anti-MetMonovalent Antibody That Inhibits HGF-Mediated Activation • Rationale for targeting Met: • Met is amplified, mutated, overexpressed in many tumors • Met expression is associated with a worse prognosis in many cancers including NSCLC • Met activation is implicated in resistance to erlotinib/gefitinib in pts with activating EGFR mutations • MetMAb: • One-armed format designed to prevent HGF-mediated stimulation of pathway • Preclinical activity across multiple tumor models HGF HGF MetMAb Met Met Growth, migration, survival No activity With permission from Spigel DR et al. Proc ESMO 2010;Abstract LBA15.

  34. OAM4558g Study Design: Global, Double-Blind, Placebo-Controlled, Phase II Study n = 64 Arm AErlotinib (150 qd-oral) + MetMAb (15 mg/kg IV q3w) n = 128 1:1 RANDO M IZ A T I O N • Key eligibility: • Stage IIIB/IV NSCLC • 2nd/3rd-line NSCLC • Tissue required • PS 0-2 • Stratification factors: • Tobacco history • Performance status • Histology n = 64 Arm BErlotinib (150 qd-oral) +placebo (IV q3w) • Co-primary objectives: • PFS in “MET High” patients • PFS in overall ITT population Other key objectives: • OS in “MET High” patients • OS in overall ITT patients • Overall response rate • Safety/tolerability PD Addition of MetMAb* n = 23 • Enrollment from 3/2009 to 3/2010 • Data cut-off: June 8, 2010 * If eligible With permission from Spigel DR et al. Proc ESMO 2010;Abstract LBA15.

  35. PFS and OS: MET High Population Erlotinib +MetMAb(n = 35) Erlotinib +Placebo(n = 30) OS, HR = 0.55 PFS, HR = 0.56 Erlotinib +Placebo(n = 30) Erlotinib +MetMAb(n = 35) Median OS (mo)Hazard ratiop-value# Events 7.7 7.4 1.0 1.0 0.550.1113 0.8 0.8 20 13 Median PFS (wk)Hazard ratiop-value# Events 6.4 12.4 0.560.0547 0.6 0.6 Probability of Survival 25 Probability of Progression Free 19 0.4 0.4 0.2 0.2 0 0 0 3 6 9 12 15 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 51 Time to Progression (weeks) Overall Survival (months) Number at Risk: Number at Risk: Erlotinib + placebo Erlotinib + placebo 3035 1726 910 33 01 00 3035 1822 59 35 33 21 21 01 01 Erlotinib + MetMAb Erlotinib + MetMAb • MetMAb + erlotinib improves both PFS and OS in • MET high NSCLC patients With permission from Spigel DR et al. Proc ESMO 2010;Abstract LBA15.

  36. PFS and OS: MET Low Population Erlotinib +MetMAb(n = 27) Erlotinib +Placebo(n = 29) PFS, HR = 2.01 OS, HR = 3.02 Median OS (mo)Hazard ratiop-value# Events 9.2 5.5 Erlotinib +Placebo(n = 29) Erlotinib +MetMAb(n = 27) 3.020.0212 1.0 1.0 14 9 11.4 6.0 Median PFS (wk)Hazard ratiop-value# Events 0.8 0.8 2.010.0354 20 23 0.6 0.6 Probability of Survival Probability of Progression Free 0.4 0.4 0.2 0.2 0 0 0 3 6 9 12 15 48 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 51 Overall Survival (months) Time to Progression (weeks) Number at Risk: Number at Risk: Erlotinib + placebo Erlotinib + placebo 2927 2312 93 30 00 00 2927 2215 95 61 20 20 00 00 00 Erlotinib + MetMAb Erlotinib + MetMAb • MET low NSCLC patients do worse with • MetMAb + erlotinib With permission from Spigel DR et al. Proc ESMO 2010;Abstract LBA15.

  37. Approach to Therapy of Acquired Resistance to EGFR TKIs • Rebiopsy the patient • T790M prognostic and possibly predictive biomarker • Rare transformation to small cell phenotype • Continue an EGFR TKI • “Second generation” EGFR/ErbB2 TKIs • Rational combination strategies • BIBW 2992 + cetuximab • HSP-90 inhibitor + chemo or EGFR-TKI • Add MET inhibitor - best diagnostic unclear

More Related