Advanced Non-Small Cell Lung Cancer: State of the Art Primo N. Lara, Jr., MD Professor of Medicine University of California Davis Cancer Center Frontline therapy of advanced NSCLC: “Back in the day” dogma (ie, 2009) Prognostic factors for survival (PS, Weight Loss, Gender) are known and clinically apparent Platinum-based chemotherapy results in prolongation of life, symptom control, & superior QOL compared with supportive care alone – Four to six cycles are sufficient Inhibitors of epidermal growth factor and angiogenesis pathways improve outcomes in select patient subsets ECOG 1594: Platinum-based doublets have similar survival outcomes Cis/Gem Control arm: Cisplatin/paclitaxel Schiller JH, et al. N Engl J Med. 2002; 346:92–98 Cis/Docetaxel Carbo/Paclitaxel Duration of chemotherapy: 4-6 cycles are sufficient Author Smith JCO 2001 Socinski JCO 2002 Eberhardt W, et al. (2007) ASCO Study design Patients Median survival (months) MVP x 3 155 6 P value .2 MVP x 6 153 7 Carbo/Pac x 4 114 6.6 .63 Carbo/Pac till PD 116 8.5 Bevacizumab in NSCLC (E4599) Proportion surviving 1.0 12 mo 24 mo Median PC 44.4% 15.4% 10.3 mo BV/PC 51.0% 22.0% 12.3 mo 0.8 Median 12.3 mo 0.6 HR: 0.80, P = .013 Median 10.3 mo 0.4 0.2 0.0 0 6 12 18 24 30 36 42 48 PC 444 318 190 104 36 9 5 1 0 BV/PC 434 340 216 127 54 25 8 3 0 Patients at risk Months Non-squamous histology, no hemoptysis, no CNS mets, no anticoagulation Sandler A, et al. N Engl J Med. 2006; 356:2542–2550 Cisplatin/vinorelbine + cetuximab (FLEX) in EGFR-positive* (by IHC) NSCLC Patients surviving, % ▬ Cetuximab +CT ▬ CT HR = 0.871, 0.762–0.996 Log-rank P =.044 11.3 10.1 1-year OS 47% vs 42% Months Pirker R, et al. (2008) ASCO * One or more IHC positive cell(s) Metastatic NSCLC: The New Issues (i.e., October 2010) Maintenance therapy – After completing platinum-based chemo, should all patients receive maintenance therapy? Tumor histology – Should we routinely use histology to select therapy? Frontline biologics – Can we abandon frontline platinum therapy in favor of single agent EGFR TKI? Maintenance therapy: Considerations Continuing same chemotherapy past 4–6 cycles yields no clear benefit E4599 and FLEX trials included maintenance therapy of bevacizumab and cetuximab, respectively Maintenance therapy with non-cross resistant agents may be more efficacious Maintenance pemetrexed Double-blind, Placebo-controlled, Multicenter, Phase III Trial Stage IIIB/IV NSCLC Pemetrexed 500 mg/m2 (d1,q21d) + BSC (N = 441)* PS 0-1 4 prior cycles of gem, doc, or tax + cis or carb, with CR, PR, or SD Randomization factors: gender PS stage best tumor response to induction non-platinum induction drug brain mets 2:1 Randomization Primary Endpoint = PFS Placebo (d1, q21d) + BSC (N = 222)* *B12, folate, and dexamethasone given in both arms Ciuleanu TE, et al. (2008) ASCO; Belani CP, et al. (2009) ASCO Drug-related adverse events Pemetrexed n = 441 Placebo n = 222 P value Drug-related deaths* 0.0% 0.0% 1.000 ≥ 1 serious AE (SAE) 4.3% 0.0% .001 14.3% 3.6% < .001 ≥ 1 grade 3/4 AE Drug-related CTCAE grade 3/4 toxicity (≥ 2% of pts) Anemia 2.7% 0.5% .070 Neutropenia 2.7% 0.0% .011 Fatigue 4.3% 0.5% .004 * on-study or within 30 days post-study Ciuleanu TE, et al. (2008) ASCO Progression-free Survival Progression-free Probability JMEN: “Maintenance” Pemetrexed vs Placebo After Platinum-based Chemotherapy 1.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.0 HR = 0.60 (95% CI: 0.49-0.73) P <0.00001 Pemetrexed 4.0 mos Placebo 2.0 mos Overall Survival Belani CP, et al. ASCO 2009. CRA8000. Survival Probability 0 1.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.0 3 6 9 12 15 Time (months) 18 21 24 HR = 0.79 (95% CI: 0.65-0.95) P = 0.012 Pemetrexed 13.4 mos Placebo 10.6 mos 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 Time (months) JMEN: “Maintenance” Pemetrexed vs Placebo After Platinum-based Chemotherapy Pemetrexed N = 309 Placebo N = 178 Any systemic therapy 51% 67% Docetaxel 22% 29% Erlotinib 22% 21% Gefitinib 13% 10% 1% 19% Pemetrexed • • • • Post-study therapy was not balanced between the arms 51% of patients on the pemetrexed arm received further therapy (equates to third-line therapy) Only 19% of placebo-arm patients received pemetrexed Would survival benefits have been preserved if more patients on the placebo arm received pemetrexed? Belani CP, et al. ASCO 2009. CRA8000. JMEN: “Maintenance” Pemetrexed vs Placebo: Survival by Histology Non-squamous (n = 481) Squamous (n = 182) Survival Probability HR = 0.70 (95% CI: 0.56-0.88); P = 0.002 HR = 1.07 (95% CI: 0.49–0.73); P = 0.678 1.0 1.0 0.9 0.9 0.8 0.8 0.7 0.7 0.6 0.6 Pemetrexed 15.5 mos 0.5 0.4 Pemetrexed 9.9 mos 0.5 0.4 0.3 0.3 Placebo 10.3 mos 0.2 0.1 0.1 0.0 0.0 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 Time (months) Belani CP, et al. ASCO 2009. CRA8000. Placebo 10.8 mos 0.2 0 3 6 9 12 15 18 21 24 27 30 33 36 39 42 45 48 Time (months) Maintenance chemotherapy: Points to Ponder • Definition of “maintenance” varies • Benefit for pemetrexed confined to nonsquamous histology • Many patients never received secondline therapy! – Survival in control arms may have diminished due to lack of life-prolonging second line therapy • Difficult to reconcile with maintenance bevacizumab, cetuximab, and erlotinib Maintenance chemotherapy: Is it time for routine clinical use? • Issues of cost, convenience, toxicity, and QOL need to be weighed against (modest) PFS benefit in the palliative care setting – Many patients desire a “drug holiday” • Maintenance chemotherapy can be considered only for selected, highly motivated patients – Bottom line: It’s not for everyone Maintenance biologics SATURN trial (N = 889) – Phase III erlotinib vs. placebo following initial treatment with platinum-based chemotherapy ATLAS trial (N = 1157) – Phase III bevacizumab ± erlotinib following initial treatment with chemo + bevacizumab – Brain mets, non-centrally located squamous, anticoagulation allowed 1. Cappuzzo F, et al. (2009) ASCO; 2. Miller VA, et al. (2009) ASCO SATURN study design Cappuzzo et al, ASCO 2009, # 8001 Erlotinib 150mg/day Chemonaïve advanced NSCLC n=1,949 4 cycles of firstline platinum doublet chemotherapy* Non-PD n=889 PD 1:1 Placebo PD Mandatory tumour sampling Co-primary endpoints: Stratification factors: • PFS in all patients • PFS in patients with EGFR IHC+ tumours • EGFR IHC (positive vs negative vs indeterminate) • Stage (IIIB vs IV) Secondary endpoints: • ECOG PS (0 vs 1) • OS in all patients and those with • CT regimen (cis/gem vs carbo/doc vs EGFR IHC+ tumours, OS and PFS in others) EGFR IHC– tumours; biomarker analyses; safety; time to symptom • Smoking history (current vs former vs progression; QoL never) *Cisplatin/paclitaxel; cisplatin/gemcitabine; cisplatin/docetaxel cisplatin/vinorelbine; • Region carboplatin/gemcitabine; carboplatin/docetaxel carboplatin/paclitaxel SATURN Trial: Efficacy Summary Erlotinib (n = 438) Placebo (n = 451) (n = 437) (n = 447) PFS at 12 weeks 53% 40% PFS at 24 weeks 31% 17% 12.3 weeks 11.1 weeks 12 months 11 months (n = 436) (n = 445) ORR 12% DCR 61% PFS Median PFS Median OS HR P Value 0.71 < .0001 0.81 .0088 5% NA .0006 51% NA .0035 • PFS benefit with erlotinib observed regardless of gender, race, histology or smoking history Cappuzzo et al. J Thorac Oncol 2009; 4(suppl 1):S289 (abstract A2.1). PFS in EGFR wild-type tumors PFS probability 1.0 Erlotinib (n=199) Placebo (n=189) 0.8 0.6 HR=0.78 (0.63–0.96) Log-rank p=0.0185 0.4 0.2 0 0 8 Capuzzo, ASCO 2009 16 24 32 40 48 56 64 Time (weeks) 72 80 88 96 PFS in EGFR mutation+ tumors* PFS probability 1.0 Erlotinib (n=22) Placebo (n=27) 0.8 HR=0.10 (0.04–0.25) Log-rank p<0.0001 0.6 0.4 0.2 0 0 8 Capuzzo, ASCO 2009 16 24 32 40 48 56 64 Time (weeks) 72 80 88 96 *60% censored SATURN: Post-study Treatment Erlotinib* (n = 438) Placebo* (n = 451) All classes 55% 64% Taxanes (including docetaxel) 26% 27% Antimetabolites (including pemetrexed) 18% 20% Antineoplastic agents 11% 15% Tyrosine-kinase inhibitors 5% 16% Platinum compounds 8% 11% *% receiving ≥1 treatment Cappuzzo F, et al. ASCO 2009. Abstract 8001. ATLAS Phase III Study Design Miller et al, ASCO 2009, #8002 Bevacizumab (15mg/kg) + erlotinib (150mg) to PD Chemo-naïve advanced NSCLC N=1,160 4 cycles of 1st-line chemotherapy* + bevacizumab Non-PD n=768 (66%) 1:1 Unblind at PD Bevacizumab + placebo to PD Eligibility Primary endpoint • Stage IIIB**/IV NSCLC • PFS in all randomized pts • ECOG performance status 0-1 Secondary endpoints Stratification factors • Overall survival • Gender • Safety • Smoking history (never vs former/current) Exploratory endpoints • ECOG performance status (0 v >1) Post progression therapy • Biomarker analyses (IHC, FISH, EGFR & K-Ras mutation) • Chemotherapy regimen *Carbo/paclitaxel; cis/vinorelbine; carbo or cis/gemcitabine; carbo or cis/docetaxel. **IIIB with pleural effusion Progression-Free Survival Proportion Without Event 1.0 Bev + Placebo (n=373) Bev + Erlotinib (n=370) 0.8 0.6 HR=0.722 (0.592-0.881) Log-rank P=0.0012 0.4 0.2 0.0 0 3 6 9 12 15 18 21 Progression-Free Survival (months) Miller et al, ASCO 2009, #8002 ATLAS: Toxicity Bev + Placebo, n (%) (n=368) Bev + Erlotinib, n (%) (n=367) Any Grade AE 313 (85.1%) 349 (95.1%) Grade 3–4 AE 112 (30.4%) 162 (44.1%) 4 (1.1%) 8 (2.2%) 60 (16.3%) 84 (22.9%) Grade 5 AE SAE The most common adverse events were rash and diarrhea Miller et al, ASCO 2009, #8002 Formal statistical comparison testing between treatment arms was not done. Take Home Points: SATURN and ATLAS trials • Trials confirm that erlotinib is an active agent in NSCLC (duh!) • Patients on maintenance erlotinib had more toxicities than those on placebo • Only a minority (16% in SATURN, 40% in ATLAS) of placebo patients ever received subsequent EGFR TKI therapy!!! • As expected, PFS benefit was best in patients with EGFR-mutated tumors • Need to balance consequences of increased toxicity and cost in the context of modest PFS benefit Metastatic NSCLC: The New Issues (i.e., October 2010) Maintenance therapy – After completing platinum-based chemo, should all patients receive maintenance therapy? Tumor histology – Should we routinely use histology to select therapy? Frontline biologics – Can we abandon frontline platinum therapy in favor of single agent EGFR TKI? Complexities of lung cancer pathogenesis result in diverse histologic subtypes Sun S, et al. Nat Rev Cancer. 2007; 7:778–790 SCLC (~15%) Squamous Cell Ca (~25%) Adenocarcinoma (~45%) BAC (~5-10%) Large Cell (~5-10%) NOS (Not Otherwise Specified (~10-30%) NSCLC histology: Treatment Considerations Bevacizumab is FDA-approved for non-squamous histology due to SAFETY issues – Severe hemoptysis Pemetrexed is FDA-approved for non-squamous histology due to EFFICACY issues – PFS and OS benefit confined to non-squamous EGFR TKIs traditionally viewed as more efficacious in adenocarcinoma – Likely due to higher rate of EGFR mutants in adeno JMDB trial: Cisplatin/pemextexed (CP) vs cisplatin/gemcitabine (CG) in Adv NSCLC Randomization Factors • • • • • Stage PS Gender Histo vs cyto dx Brain mets hx Cisplatin 75 mg/m2 day 1 plus Pemetrexed 500 mg/m2 day 1 R Cisplatin 75 mg/m2 day 1 plus Gemcitabine 1250 mg/m2 days 1 & 8 Vitamin B12, folate, and dexamethasone given in both arms Scagliotti GV, et al. J Clin Oncol. 2008; 26:3543–3551 Cisplatin/pemetrexed (CP) vs cisplatin/gemcitabine (CG) in NSCLC No difference in PFS or OS CP improves survival over CG in non-SCCA (HR 0.81, P = .005) CG improves survival over CP in SCCA (HR 1.23, P = .05) Scagliotti GV, et al. J Clin Oncol. 2008; 26:3543–3551 Pemetrexed: Influence of histology on efficacy In two other phase III trials (docetaxel vs. pemetrexed; maintenance pemetrexed vs. placebo), benefit was confined to patients with non-squamous histology Scagliotti GV, et al. J Clin Oncol. 2008; 26:3543–3551 Take Home Points: Histology-based therapy NSCLC histology is now a consideration in therapeutic selection for chemotherapy This is but one (primitive) step in the direction of “personalized therapy” Still unclear how “NSCLC NOS” or cytologic diagnoses (by FNA) should be treated True personalized therapy will rely on molecular profiling rather than histology Metastatic NSCLC: The New Issues (i.e., October 2010) Maintenance therapy – After completing platinum-based chemo, should all patients receive maintenance therapy? Tumor histology – Should we routinely use histology to select therapy? Frontline biologics – Can we abandon frontline platinum therapy in favor of single agent EGFR TKI? IPASS Study design Conducted in China, Japan, Thailand, Taiwan, Indonesia, Malaysia, Philippines, Hong Kong and Singapore Randomization period: March 2006 – October 2007 Patients • • • • • • • Chemo-naïve Age ≥18 years Adenocarcinoma histology Never or ex-light smokers* Life expectancy ≥12 weeks WHO PS 0-2 Measurable stage IIIB / IV disease Gefitinib (250 mg / day) End points Primary • Progression-free survival (non-inferiority) Secondary 1:1 randomization Carboplatin (AUC 5 or 6) / paclitaxel (200 mg / m2) 3 weekly# • • • • • Objective response rate Overall survival Quality of life Disease-related symptoms Safety and tolerability Exploratory • Biomarkers ─ EGFR mutation ─ EGFR-gene-copy number ─ EGFR protein expression *Never smokers, <100 cigarettes in lifetime; ex-light 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 WHO, World Health Organization; PS, performance status; AUC, area under the curve; EGFR, epidermal growth factor receptor Mok et al 2008 Progression-free survival Probability 1.0 of PFS N Events 0.8 Gefitinib Carboplatin / 609 453 (74.4%) paclitaxel 608 497 (81.7%) HR (95% CI) = 0.74 (0.65, 0.85) p<0.0001 0.6 5.8 74% 48% 7% Median PFS (months) 5.7 4 months progression-free 61% 6 months progression-free 48% 12 months progression-free 25% 0.4 Gefitinib demonstrated superiority relative to carboplatin/paclitaxel in terms of PFS 0.2 0.0 Patients at risk : 0 Gefitinib 609 C/P 608 4 8 12 16 20 24 Months 363 412 212 118 76 22 24 3 5 1 0 0 Primary Cox analysis with covariates; HR <1 implies a lower risk of progression on gefitinib; ITT population PFS, progression-free survival; ITT, intent-to-treat; HR, hazard ratio; CI, confidence interval; C/P, carboplatin/paclitaxel Mok et al 2008 IPASS: EGFR mutation positive status and clinical characteristics % of samples EGFR mutation positive Overall EGFR mutation positive rate = 59.7% (261 / 437) 68.5 63.0 60.0 57.1 49.0 Male 60.7 57.8 60.2 56.7 46.9 Female PS 0/1 PS 0/2 Never Light ex- Locally Metastatic Age Age smoked smoker advanced <65 yrs >65 yrs Progression-free survival in EGFR mutation positive and negative patients EGFR mutation positive EGFR mutation negative Probability of progression-free survival 1.0 0.8 0.6 0.4 Gefitinib (n=91) Carboplatin/paclitaxel (n=85) 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%) Median PFS G, 1.5 months Median PFS C/P, 5.5 months 1.0 Probability of progression-free survival Gefitinib (n=132) Carboplatin/paclitaxel (n=129) 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%) Median PFS G, 9.5 months Median PFS C/P, 6.3 months 0.8 0.6 0.4 0.2 0.2 0.0 0.0 0 Patients at risk : Gefitinib 132 C/P 129 4 8 12 16 20 24 0 4 8 Months 108 103 71 37 31 7 12 16 20 24 1 0 0 0 0 0 Months 11 2 3 1 0 0 91 85 21 58 4 14 2 1 Treatment by EGFR mutation status interaction test, p<0.0001 Cox analysis with covariates; HR <1 implies a lower risk of progression on gefitinib; ITT population Mok et al 2008 First line gefinitib vs. chemotherapy in EGFR mutated NSCLC NSCLC with sensitive EGFR mutations Stage IIIb/ IV No prior chemo. PS 0-1 age of 20-75 y.o R balanced : Institution sex stage Gefitinib n=160 Primary endpoint PFS 2ndary endpoints OS Response CBDCA+TXL n=160 Side-effects QOL • The sample size was calculated to be 320 in total (alpha=5%, power=80%) to confirm the superiority of Arm A (hazard ratio = 0.69). • An interim analysis to investigate PFS was planned 4 months after 200 pts were entered . North East Japan (NEJ) Gefitinib Study Group PFS OS Memondo, NEJM 2010 Memondo, NEJM 2010 Potential Oncogenic “Drivers” in Non-small Cell Lung Cancer (NSCLC) Adenocarcinoma K-ras EGFR B-raf Her2 Other PIK3CA ALK MET Other ALK (~5%) ALK = anaplastic lymphoma kinase; EGFR = epidermal growth factor receptor; Her2 = human epidermal growth factor receptor 2; PIK3CA = phosphoinositide-3-kinase, catalytic, alpha polypeptide Massachusetts General Hospital, data on file. [AT Shaw, personal communication] Tumor Responses to Crizotinib for Patients with ALK-positive NSCLC (N=82) 60 Maximum change in tumor size (%) Progressive disease Stable disease 40 Confirmed partial response Confirmed complete response 20 0 –20 –30% –40 –60 –80 –100 * *Partial response patients with 100% change have non-target disease present 77% of Patients with ALK-positive NSCLC Remain on Crizotinib Treatment Individual patients • Duration of treatment (median: 5.7 months) 0–3 mo 13 pts >3–6 mo 29 pts >6–9 mo 24 pts >9–12 mo 9 pts >12–18 mo 4 pts >18 mo 3 pts 0 3 6 9 12 15 Treatment duration (months) N=82; red bars represent discontinued patients 18 • Reasons for discontinuation – Related AEs 1 – Non-related AEs 1 – Unrelated death 2 – Other 2 – Progression 13 21 • Clinical Activity of Crizotinib in Patients with NSCLC Objective response rateALK-positive (ORR): 57% (95% CI: 46, 68%) – 63% including 5 as yet unconfirmed PRs – 57% (8/14) for patients with performance status 2 or 3 0 ORR % (n/N) 80 (4/5) 1 52 (14/27) 2 67 (10/15) ≥3 56 (19/34) No. prior regimens* * Unknown for 1 patient ● Response duration: 1 to 15 months ● DCR† (CR/PR/SD at 8 weeks): 87% (95% CI: 77, 93%) †Disease control rate Progression-free survival probability Median PFS has Not been Reached 70% of Patients in Follow-up for PFS 1.00 PFS probability at 6 months: 72% (95% CI: 61, 83%) 0.75 0.50 0.25 0.00 Median follow-up for PFS: 6.4 months (25–75% percentile: 3.5–10 months) 0 2.5 95% Hall–Wellner confidence bands 5.0 7.5 10.0 12.5 15.0 Progression-free survival (months) 17.5 Treatment-related Adverse Events in ALK-positive NSCLC (≥10%) Adverse event Grade 1 n (%) Grade 2 n (%) Grade 3 n (%) Grade 4 n (%) Total n (%) Nausea 43 (52) 1 (1) 0 0 44 (54) Diarrhea 38 (46) 1 (1) 0 0 39 (48) Vomiting 35 (43) 1 (1) 0 0 36 (44) Visual disturbance* 34 (42) 0 0 0 34 (42) Constipation 18 (22) 2 (2) 0 0 20 (24) Peripheral edema 13 (16) 0 0 0 13 (16) Dizziness 12 (15) 0 0 0 12 (15) Decreased appetite 11 (13) 0 0 0 11 (13) Fatigue 8 (10) 0 0 0 8 (10) *Changes in light/dark accommodation (no abnormalities on ophthalmologic exam) N=82 Conclusions: Frontline NSCLC therapy Optimal therapy is rapidly evolving Maintenance therapy is an option for highly motivated patients Clinical, histologic, and molecular biomarkers are now important considerations for therapy selection Future advances in NSCLC outcomes will likely be due to molecular selection Support for clinical trials testing this paradigm is essential