throbber
VOLUME 29 䡠 NUMBER 26 䡠 SEPTEMBER 10 2011
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`O R I G I N A L R E P O R T
`
`Fred R. Hirsch, Rafal Dziadziuszko,
`Wilbur A. Franklin, Marileila Varella-
`Garcia, Paul A. Bunn Jr, D. Ross Camidge,
`University of Colorado Cancer Center,
`Aurora, CO; Fairooz Kabbinavar, Univer-
`sity of California, Los Angeles, David
`Geffen School of Medicine, Los Ange-
`les, CA; Tim Eisen, Cambridge Biomedi-
`cal Research Center, Cambridge,
`United Kingdom; Renato Martins, Fred
`Hutchinson Cancer Research Center,
`Seattle, WA; Frederick M. Schnell,
`Central Georgia Cancer Center, Macon,
`GA; Katherine Richardson, Frank Rich-
`ardson, Bret Wacker, David W. Stern-
`berg, Jason Rusk, OSI Pharmaceuticals,
`Melville, NY.
`
`Submitted January 11, 2011; accepted
`May 26, 2011; published online ahead
`of print at www.jco.org on August 8,
`2011.
`
`Supported by an unrestricted grant
`from OSI Pharmaceuticals and from
`National Cancer Institute Lung SPORE
`Award No. P50 CA058187.
`
`Authors’ disclosures of potential con-
`flicts of interest and author contribu-
`tions are found at the end of this
`article.
`
`Clinical Trials repository link available on
`JCO.org.
`
`Corresponding author: Paul A. Bunn Jr,
`MD, University of Colorado Cancer Center,
`12801 E 17th Ave, Aurora, CO 80045;
`e-mail: Paul.Bunn@ucdenver.edu.
`
`© 2011 by American Society of Clinical
`Oncology
`
`0732-183X/11/2926-3567/$20.00
`
`DOI: 10.1200/JCO.2010.34.4929
`
`A Randomized, Phase II, Biomarker-Selected Study
`Comparing Erlotinib to Erlotinib Intercalated With
`Chemotherapy in First-Line Therapy for Advanced
`Non–Small-Cell Lung Cancer
`Fred R. Hirsch, Fairooz Kabbinavar, Tim Eisen, Renato Martins, Fredrick M. Schnell, Rafal Dziadziuszko,
`Katherine Richardson, Frank Richardson, Bret Wacker, David W. Sternberg, Jason Rusk, Wilbur A. Franklin,
`Marileila Varella-Garcia, Paul A. Bunn Jr, and D. Ross Camidge
`
`A
`
`B
`
`S
`
`T
`
`R
`
`A
`
`C
`
`T
`
`Purpose
`Erlotinib prolongs survival in patients with advanced non–small-cell lung cancer (NSCLC). We report the
`results of a randomized, phase II study of erlotinib alone or intercalated with chemotherapy (CT ⫹
`erlotinib) in chemotherapy-naïve patients with advanced NSCLC who were positive for epidermal
`growth factor receptor (EGFR) protein expression and/or with high EGFR gene copy number.
`Patients and Methods
`A total of 143 patients were randomly assigned to either erlotinib 150 mg daily orally until disease
`progression (PD) occurred or to chemotherapy with paclitaxel 200 mg/m2 intravenously (IV) and
`carboplatin dosed by creatinine clearance (AUC 6) IV on day 1 intercalated with erlotinib 150 mg orally
`on days 2 through 15 every 3 weeks for four cycles followed by erlotinib 150 mg orally until PD
`occurred (CT ⫹ erlotinib). The primary end point was 6-month progression-free survival (PFS);
`secondary end points included response rate, PFS, and survival. EGFR, KRAS mutation, EGFR
`fluorescent in situ hybridization and immunohistochemistry, and E-cadherin and vimentin protein levels
`were also assessed.
`Results
`Six-month PFS rates were 26% and 31% for the two arms (CT ⫹ erlotinib and erlotinib alone,
`respectively). Both were less than the historical control of 45% (P ⫽ .001 and P ⫽ .011,
`respectively). Median PFS times were 4.57 and 2.69 months, respectively. Patients with tumors
`harboring EGFR activating mutations fared better on erlotinib alone (median PFS, 18.2 months v
`4.9 months for CT ⫹ erlotinib).
`Conclusion
`The feasibility of a multicenter biomarker-driven study was demonstrated, but neither treatment
`arms exceeded historical controls. This study does not support combined chemotherapy and
`erlotinib in first-line treatment of EGFR-selected advanced NSCLC, and the patients with tumors
`harboring EGFR mutations had a better outcome on erlotinib alone.
`
`J Clin Oncol 29:3567-3573. © 2011 by American Society of Clinical Oncology
`
`INTRODUCTION
`
`Erlotinib, an epidermal growth factor receptor
`(EGFR) –directed tyrosine kinase inhibitor (TKI),
`prolongs progression-free survival (PFS) and overall
`survival (OS) in unselected patients with non–
`small-cell lung cancer (NSCLC) in the first-line,
`second/third-line and first-line maintenance
`therapies.1-3 Randomized studies of chemotherapy
`in combination with erlotinib demonstrated no ad-
`vantage and possible antagonism among these
`therapies in an unselected population.4,5 Preclinical
`studies suggested that G1 cell cycle arrest induced by
`
`erlotinib could interfere with the G2/M cytotoxicity
`of taxanes and suggested that appropriate schedul-
`ing of erlotinib with taxanes produce additive or
`synergistic growth inhibition.6 We previously dem-
`onstrated that patients with advanced NSCLC who
`were negative for EGFR by both fluorescent in situ
`hybridization (FISH) and immunohistochemistry
`(IHC) had no benefit from gefitinib therapy in the
`second/third-line setting.7
`These studies led to the current randomized,
`phase II study evaluating erlotinib versus chemo-
`therapy intercalated with erlotinib in chemotherapy-
`naive patients with advanced NSCLC who were
`
`© 2011 by American Society of Clinical Oncology
`
`3567
`
`APOTEX EX. 1032-001
`
`

`

`Hirsch et al
`
`positive for EGFR protein expression and/or high EGFR gene copy
`number. This study was initiated before the results from the Iressa
`Pan-Asia Study (IPASS), which identified the significance of EGFR
`mutation testing before first-line therapy, was available.8 Other goals
`were to determine the feasibility of a prospective biomarker multi-
`center study and to select a treatment arm for a randomized, phase
`III trial.
`
`PATIENTS AND METHODS
`
`Study Design
`This was an international, randomized, phase II study of erlotinib as
`single-agent treatment or of carboplatin/paclitaxel chemotherapy intercalated
`with erlotinib in newly diagnosed patient with NSCLC who had EGFR-
`positive tumors assessed by IHC or FISH. Thirty-seven centers in the United
`States and five in the United Kingdom participated. The primary end point was
`the percentage of patients alive and without tumor progression at 6 months (ie,
`6-month PFS). Secondary end points included tumor response rate (RR), PFS,
`and OS as well as the exploration of the correlation between clinical outcome
`and biomarkers of interest. Key inclusion criteria were sufficient tumor tissue
`sample for EGFR testing; histologically or cytologically advanced (ie, stages
`IIIB or IV) NSCLC; radiologically measurable or evaluable disease; and ade-
`quate organ function. Patients who received any prior or concurrent antican-
`cer therapy for advanced NSCLC and patients who had uncontrolled brain
`metastases were excluded.
`Web-based, centralized random assignment was performed by IDDI
`(Brussels, Belgium) by using an adaptive random assignment method by
`Pocock and Simon.9 Patients were stratified by the number of positive tests
`for EGFR expression (by IHC, FISH: 1 or 2) smoking status (current,
`former, or never), ECOG performance status (0/1 or 2), and extent of
`disease (stage IIIB or IV).
`The study was approved by each institution’s institutional review board/
`ethics committee. Written informed consent was obtained from all patients for
`participation, including for tissue analyses and banking.
`Treatment
`Patients were randomly assigned (1:1) to receive erlotinib 150 mg daily
`orally until disease progression (PD) occurred or to receive chemotherapy
`(paclitaxel 200 mg/m2 intravenously [IV] and carboplatin dosed by creatinine
`clearance [AUC 6] according to local practice IV on day 1) alternating with
`erlotinib 150 mg orally on days 2 through 15 every 3 weeks for four cycles,
`followed by erlotinib 150 mg orally daily until PD occurred. Patients were
`evaluated every 6 weeks by chest x-ray or computed tomography (CT) scan for
`PD. After PD, patients were treated at physician’s discretion (Fig 1). Ongoing
`patient follow-up was conducted every 3 months.
`Biomarkers
`The University of Colorado Cancer Center (UCCC, Aurora, CO) re-
`ceived tumor samples from sites to assess EGFR IHC and FISH. UCCC per-
`formed quality-control assessments before the analyses to ensure sufficient
`tumor tissue. With consent, the remnant tissue was used for EGFR mutation
`testing by Genzyme Genetics (Westborough, MA) and KRAS mutation anal-
`ysis by OSI Pharmaceuticals (Boulder, CO). IHC was assessed for E-cadherin
`and vimentin by OSI Pharmaceuticals.
`EGFR IHC
`Protein expression for EGFR by IHC was assayed with the Dako (Car-
`pentaria, CA) EGFR PharmDX kit. For the purpose of eligibility, positive
`EGFR IHC was defined by greater than 10% positive cells assessed by two
`independent reviewers.10 In cases of discrepancies, the final score was
`based on a consensus meeting.
`EGFR FISH
`FISH analysis was performed according to previously published meth-
`ods.11,12 Samples identified with EGFR high polysomy (⬎ four copies of the
`EGFR gene present in 40% to 100% cells) or with EGFR gene amplification
`
`Tumor tissue
`analyzed at
`UCCC
`(N = 214)
`
`IHC positive and/or FISH positive
`
`Screen Failure:
`IHC negative AND FISH negative
`(n = 12)
`UNK/UNK
`(n = 24)
`UNK/Negative
`(n = 3)
`
`Randomly
`assigned
`(n = 143)
`
`Arm A
`erlotinib 150
`mg/day
`(n = 72)
`
`Arm B
`CBDP (AUC6) +
`paclitaxel (200
`mg/m2) d1, x 4
`cycles;
`erlotinib 150 mg/d,
`d2-15
`(n = 71)
`
`Safety/efficacy
`analysis
`(n = 69)
`Discontinued
` PD
` AE
` Patient request
` Death
` Other reason
`
` (n = 50)
`(n = 7)
`(n = 6)
`(n = 4)
`(n = 2)
`
`Safety/efficacy analysis
`(n = 68)
`Discontinued
` PD
` AE
` Patient request
` Other reason
`
` (n = 49)
`(n = 14)
`(n = 3)
`(n = 4)
`
`Fig 1. CONSORT diagram. AUC, area under the curve; AE, adverse event;
`CBDP, carboplatin; FISH, fluorescent in situ hybridization; IHC, immunohisto-
`chemistry; PD, progressive disease; UCCC, University of Colorado Cancer
`Center; UNK, unknown.
`
`(gene/chromosome ratio ⬎ two or ⱖ 15 gene copies in ⱖ 10% cells) were
`considered positive for copy number gain (FISH positive). All other samples
`were considered FISH negative. The FISH assessment was performed by two
`independent reviewers, and discrepant assessments were solved by consen-
`sus discussion.
`EGFR Mutation
`EGFR exons 18 through 21 were amplified by polymerase chain reaction
`(PCR) at Genzyme Genetics according to their standard procedure for EGFR
`mutational analysis. The resultant PCR fragments were sequenced by using
`BigDye version 1 and 3130 Genetic Analyzer (Applied Biosystems, Foster City,
`CA). EGFR activating mutations were noted by deletions on exon 19 or L858R
`mutations on exon 21. Patients with other mutations or deletions were classi-
`fied wild type (WT) for analyses.
`KRAS Mutation, E- Cadherin, and Vimentin
`The DNA isolated for EGFR mutational analysis was used for KRAS
`mutational analysis in codons 12 and 13. The protein expression of E-cadherin
`was assessed by IHC with antibody H-108 (Santa Cruz Biotechnology No.
`7870, Santa Cruz, CA). The assessment was considered high when at least 40%
`of the cells stained with intensity 2 or 3. The vimentin status was determined by
`IHC with antibody V9 (Dako No. M0725). The results were considered high
`when there was at least 10% staining of any intensity.
`Statistical Analysis
`This was a pick-the-winner, phase II design that was not adequately
`powered to test for treatment differences, as proposed by Simon et al13 Both
`
`3568
`
`© 2011 by American Society of Clinical Oncology
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`APOTEX EX. 1032-002
`
`

`

`EGFR-Selected Therapy in Advanced NSCLC
`
`treatment arms were considered experimental, and the treatment arm with the
`numerically superior PFS was to be considered for testing in future studies.
`The sample size was based on the ability to detect, with a one-sided ␣of
`.05, an improvement in the 6-month PFS rate from an historical 45% with
`standard first-line platinum-based therapy to a hypothesized 60%, which
`would be a clinically meaningful improvement.4,5 PFS was defined as the time
`from random assignment until occurrence of documented radiologic and/or
`symptomatic PD according to RECIST (Response Evaluation Criteria in Solid
`Tumors), version 1.0, or until death in the absence of progression.14 Patients
`who did not experience progression were censored on the last day known to be
`free of progression by objective tumor measurements. Patients who received
`other therapy before documented PD were censored on the day subsequent
`therapy started. Survival was defined as time from random assignment until
`documented death. Patients who were still alive were censored on the last day
`known to be alive.
`PFS and OS analyses included patients who received any study therapy.
`The 6-month PFS rates with 90% CIs were calculated for each treatment arm,
`and Kaplan-Meier estimates of PFS and OS were constructed for each treat-
`ment arm. In each arm, the 6-month PFS rate was compared with the historical
`control of 45%. Analyses of RR included patients who received any study
`therapy and had measurable disease.
`Kaplan-Meier estimates of PFS were calculated for each biomarker level
`(positive v negative or mutation v WT) within each treatment arm. Log-rank
`analyses were performed to test for significant difference between biomarker
`levels. All P values presented are for exploratory purposes. RR and disease
`control rates (DCRs) were compared between the two groups with two-sided
`Fisher’s exact tests. A P value ⱕ .05 was considered statistically significant.
`
`Table 1. Biomarker Results On the Basis of Treatment Arm and Key
`Patient Characteristics
`
`Biomarker Result
`
`IHC result
`Positive
`Negative
`FISH result
`Positive
`Negative
`EGFR mutation result
`Mutation
`Activating mutation
`Exon 19 deletion
`Exon 21 L858R mutation
`Other mutation
`No mutation
`KRAS mutation result
`Mutation
`No mutation
`E-cadherin
`High
`Low
`Vimentin
`High
`Low
`
`% of Total Patients by Treatment Arm
`
`Erlotinib
`(n ⫽ 72)
`
`CP ⫹ Intercalated
`Erlotinib (n ⫽ 71)
`
`Total
`(N ⫽ 143)
`
`93
`4
`
`54
`43
`
`18
`12
`11
`1
`6
`67
`
`18
`75
`
`36
`33
`
`29
`40
`
`92
`8
`
`54
`46
`
`17
`10
`4
`6
`7
`65
`
`23
`73
`
`30
`38
`
`17
`48
`
`92
`6
`
`54
`45
`
`17
`11
`8
`3
`6
`66
`
`20
`74
`
`33
`36
`
`23
`44
`
`RESULTS
`
`Abbreviations: CP, carboplatin/paclitaxel; IHC, immunohistochemistry; FISH,
`fluorescent in situ hybridization.
`
`Patient Characteristics and Tumor Samples
`Key patient characteristics and demographics were balanced be-
`tween arms (Appendix Table A1, online only). Two-hundred forty
`patients with advanced NSCLC were screened, and formalin-fixed,
`paraffin-embedded biopsies were obtained in 214 patients (Fig 1).
`EGFR IHC and/or FISH results were obtained for 190 samples (89%);
`24 (11%) failed the quality control analysis (eg, insufficient tissue for
`analysis) and were not evaluated. At least one of the two EGFR tests
`was positive in 175 samples (92%); 12 (6%) were negative for both
`assays; and three had combinations of negative and unknown results.
`Between March 2007 and December 2008, 143 patients were eligible
`and randomly assigned; 92% were positive by IHC, and 54% were
`positive by FISH (Table 1); 45% were positive by both IHC and FISH.
`Seventy-two patients were randomly assigned to erlotinib, and 71
`patients were randomly assigned to chemotherapy plus erlotinib; 137
`patients were included in the efficacy and safety analyses. Six patients
`did not receive study drug; three were in the erlotinib arm, and three
`were in the CT plus erlotinib arm.
`The 214 tumor tissue samples consisted of primary lung lesions
`(n ⫽ 145 [67%][), metastatic sites (n ⫽ 55 [26%]), and tumor from an
`unknown location (n ⫽ 14 [7%]). Biomarker results are listed in Table
`1. The average time from receipt of tissue at the central lab to bio-
`marker results being provided to the treatment site was 4 working days
`(range, 1 to 9 days).
`EGFR mutation results were obtained from 119 patients (83%),
`and activating EGFR mutations were found in 16 patients (11%;
`n ⫽ 11, exon-19 deletions; n ⫽ 5, exon-21 L858R). No difference in
`distribution between the treatment arms was seen. Two patients had
`concurrent L858R activating mutation and T790M-acquired resis-
`tance mutation. EGFR activating mutations were higher among
`women (16% v 6% in men), adenocarcinoma histology (15% v 0% in
`
`others), Asian ethnicity (38% v 8% in non-Asians), and never smokers
`(28% v 5% in former smokers and 4% in current smokers).
`KRAS mutation analysis was performed in 135 patients, and 29
`(20%) had mutations. No patient had both EGFR and KRAS muta-
`tion. KRAS mutation rates were highest in current smokers (40% v
`22% in former and 8% in never smokers).
`EGFR FISH was performed in 141 patients and was positive in 77
`patients (54%). No difference in the distribution of EGFR FISH posi-
`tivity was seen regarding sex, histology or smoking status. EGFR IHC
`was positive in 132 (92%) of 141 patients; no difference was associated
`with sex, histology or smoking status. E-cadherin expression was high
`in 47 (48%) of 98 patients, and vimentin was high in 33 (24%) of
`96 patients.
`The associations among EGFR mutation, KRAS mutations, and
`EGFR FISH are shown in Figure 2 for the 119 patients evaluable for
`FISH, EGFR mutation, and KRAS mutation. Of the 66 EGFR FISH-
`positive tumors, 10 had KRAS mutations. Among 16 tumors with
`EGFR activating mutations, 13 were EGFR FISH positive.
`
`Treatment Administration
`Patients in the erlotinib arm received a median of 10.3 weeks of
`treatment (range, 1.1 to 125.7 weeks). Patients in the chemotherapy
`plus erlotinib arm received a median of 9.8 weeks (range, 0.1 to
`95.6 weeks).
`
`Efficacy
`PFS. Kaplan- Meiers curves of PFS are shown in Figure 3. For
`the overall population, the curves favored the chemotherapy plus
`
`www.jco.org
`
`© 2011 by American Society of Clinical Oncology
`
`3569
`
`APOTEX EX. 1032-003
`
`

`

`CP + intercalated erlotinib (n = 68)
`Median: 4.57 months
`6-month rate: 26.4%
`
`Erlotinib (n = 69)
`Median: 2.69 months
`6-month rate: 30.7%
`
`12
`15
`18
`Time (months)
`
`21
`
`24
`
`27
`
`30
`
`CP + intercalated erlotinib (n = 50)
`Median: 5.29 months
`6-month rate: 28.9%
`
`Erlotinib (n = 49)
`Median: 2.10 months
`6-month rate: 24.6%
`
`Hirsch et al
`
`1.00
`
`0.75
`
`0.50
`
`0.25
`
`A
`
`Survival (probability)
`Progression-Free
`
`KRAS
`mutation
`positive
`
`0
`
`3
`
`6
`
`9
`
`EGFR by FISH
`
`EGFR-activating
`mutation
`positive
`
`3
`
`13
`
`0
`
`0
`
`43
`
`10
`
`1.00
`
`0.75
`
`0.50
`
`0.25
`
`B
`
`Survival (probability)
`Progression-Free
`
`0
`
`3
`
`6
`
`9
`
`12
`15
`18
`Time (months)
`
`21
`
`24
`
`27
`
`30
`
`CP + intercalated erlotinib (n = 6)
`Median: 4.90 months
`6-month rate: 41.7%
`
`Erlotinib (n = 9)
`Median: 18.20 months
`6-month rate: 88.9%
`
`1.00
`
`0.75
`
`0.50
`
`0.25
`
`C
`
`Survival (probability)
`Progression-Free
`
`0
`
`3
`
`6
`
`9
`
`12
`15
`18
`Time (months)
`
`21
`
`24
`
`27
`
`30
`
`Fig 3. Kaplan-Meier plots of progression-free survival. (A) All patients; (B) EGFR
`wild-type patients; (C) EGFR mutant patients. CP, carboplatin/paclitaxel.
`
`therapy plus erlotinib arm (P⫽ 1.0). EGFR FISH-positive patients had
`a numerically higher RR (18.9% in the erlotinib arm and 26% in the
`chemotherapy plus erlotinib arm) compared with the EGFR FISH-
`negative patients (3% in the erlotinib arm and 19% in the chemother-
`apy plus erlotinib arm; Table 3). KRAS mutations had a trend toward
`a negative effect on RR and DCR when these patients were com-
`pared with patients who were KRAS WT (erlotinib: RR 0% v 16%
`[P ⫽ .1908]; DCR 31% v 53% [P ⫽ .2168]; and chemotherapy plus
`
`Fig 2. Thirty-six patients were fluorescent in situ hybridization negative, KRAS
`wild type (WT), and EGFR WT.
`
`erlotinib arm during the first 6 months and then crossed to favor the
`erlotinib arm. The 6-month PFS rate was 31% (90% CI, 22% to 40%)
`in the erlotinib arm, and it was 26% (90% CI, 17% to 36%) with
`chemotherapy plus erlotinib. The 6-month PFS rate in each arm was
`less than the historical control of 45% (erlotinib arm, P ⫽ .011;
`chemotherapy plus erlotinib arm, P ⫽ .001). The median PFS times
`were 2.69 months and 4.57 months within the two groups, respectively
`(Table 2). The 6-month PFS rate for patients with EGFR activation
`mutations was considerably better in the erlotinib arm than in the
`chemotherapy plus erlotinib arm (89% v 42%, respectively), as was the
`median PFS (18.2 months v 4.9 months, respectively).
`Within the erlotinib arm, patients with EGFR activating muta-
`tions had a 6-month PFS rate of 89% compared with 24% for the
`EGFR WT patients (P ⬍ .001). In the chemotherapy plus erlotinib
`arm, patients with EGFR mutations had a 6-month PFS rate of 42%
`compared with 28% for the EGFR WT patients (P ⫽ .502).
`For the EGFR FISH-positive patients, the 6-month PFS rate was
`39% in the erlotinib arm, and it was 23% in the chemotherapy plus
`erlotinib arm; the PFS rates were and 22% and 30%, respectively, for
`the FISH-negative group. In the erlotinib arm, among the EGFR WT
`patients, the 6-month PFS rate for the FISH-positive group was 27%,
`and it was 21% for the FISH-negative group (P ⫽ .520).
`EGFR IHC did not confer any difference in the 6-month PFS rate
`(Table 2). KRAS mutation appeared to have a negative effect on
`6-month PFS rate for patients in both arms, although the results were
`not statistically significant (Table 2). For E-cadherin or vimentin ex-
`pression, no differential association was seen (Table 2).
`
`Tumor Response
`The overall response rate (RR; ie, CR ⫹ PR) was 11.6% in the
`erlotinib arm, and it was 22.4% in the chemotherapy plus erlotinib
`arm (Table 3). For patients with activating EGFR mutation, the RR
`was 67% in the erlotinib arm and it was 33% in the chemotherapy plus
`erlotinib arm. For EGFR WT patients, the RRs were 0% and 23% in
`the two arms, respectively. The DCR was 100% in patients who were
`EGFR activating mutation positive, and it was 36% in EGFR WT
`patients in the erlotinib arm (P ⫽ .0004) compared with 67% for
`mutation-positive patients and 68% for WT patients in the chemo-
`
`3570
`
`© 2011 by American Society of Clinical Oncology
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`APOTEX EX. 1032-004
`
`

`

`EGFR-Selected Therapy in Advanced NSCLC
`
`Table 2. Analyses of PFS and OS for Biomarker Subsets
`
`Table 3. Response by Biomarker Status
`
`All Patients
`
`EGFR WT Only
`
`% of Patients by Response and Treatment Arm
`
`Biomarker Subset
`
`Erlotinib
`
`CP ⫹
`Erlotinib Erlotinib
`
`CP ⫹
`Erlotinib
`
`EGFR FISH positive
`No.
`Median PFS
`6-month PFS rate
`12-month OS rate
`EGFR FISH negative
`No.
`Median PFS
`6-month PFS rate
`12-month OS rate
`P for PFS of positive v negativeⴱ
`KRAS mutation
`No.
`Median PFS
`6-month PFS rate
`12-month OS rate
`KRAS WT
`No.
`Median PFS
`6-month PFS rate
`12-month OS rate
`P for PFS of mutated v WTⴱ
`E-cadherin positive: high
`No.
`Median PFS
`6-month PFS rate
`12-month OS rate
`E-cadherin negative: low
`No.
`Median PFS
`6-month PFS rate
`12-month OS rate
`P for PFS of high v lowⴱ
`Vimentin positive: high
`No.
`Median PFS
`6-month PFS rate
`12-month OS rate
`Vimentin negative: low
`No.
`Median PFS
`6-month PFS rate
`12-month OS rate
`P for PFS of high v lowⴱ
`
`37
`2.76
`39.2
`62.2
`
`30
`2.27
`22.2
`58.3
`.075
`
`13
`2.23
`11.5
`40.4
`
`51
`3.15
`38.2
`63.1
`.078
`
`25
`2.76
`28.0
`58.8
`
`22
`1.54
`33.2
`75.6
`.794
`
`20
`1.48
`27.1
`58.2
`
`28
`2.50
`27.5
`66.5
`.757
`
`35
`5.06
`23.4
`54.7
`
`33
`4.17
`29.8
`38.4
`.778
`
`15
`2.96
`8.6
`53.3
`
`51
`4.90
`31.7
`44.3
`.078
`
`21
`4.90
`21.4
`53.1
`
`26
`5.06
`31.0
`31.4
`.725
`
`12
`5.78
`41.6
`50.3
`
`33
`4.90
`22.0
`38.7
`.163
`
`23
`2.10
`27.6
`57.3
`
`26
`1.91
`22.0
`55.5
`.492
`
`11
`2.23
`NC
`47.7
`
`38
`1.97
`27.3
`57.9
`.550
`
`19
`2.69
`21.1
`55.9
`
`18
`1.45
`24.2
`69.6
`.495
`
`15
`1.41
`7.5
`50.3
`
`22
`2.27
`26.0
`66.1
`.287
`
`27
`5.29
`21.7
`60.1
`
`23
`4.24
`38.1
`49.5
`.652
`
`12
`2.30
`11.4
`58.3
`
`38
`5.36
`33.9
`53.5
`.092
`
`15
`5.52
`31.4
`65.2
`
`21
`5.06
`27.3
`34.4
`.836
`
`8
`6.01
`53.6
`75.0
`
`27
`5.29
`23.1
`41.6
`.058
`
`Abbreviations: PFS, progression-free survival; OS, overall survival; WT,
`wild type; CP, carboplatin/paclitaxel; FISH, fluorescent in situ hybridization;
`NC, not calculated.
`ⴱP values are from the log-rank test comparing the erlotinib and CP ⫹
`erlotinib curves.
`
`erlotinib: RR 20% v 24% [P⫽ 1.0]; DCR, 53% v 78% [P⫽ .0977]). For
`EGFR IHC, E-cadherin status, and vimentin status, no statistically
`significant differences were seen for RR.
`
`OS
`
`Among 137 patients assessed for survival, the median survival
`time was 16.7 months in the erlotinib arm, and it was 11.43 months in
`the chemotherapy plus erlotinib arm. The 12-month survival rates
`
`CR ⫹ PR
`
`CR ⫹ PR ⫹ SD
`
`CP ⫹
`Intercalated
`Erlotinib
`(n ⫽ 67)ⴱ
`
`Erlotinib
`(n ⫽ 69)
`
`CP ⫹
`Intercalated
`Erlotinib
`(n ⫽ 67)ⴱ
`
`Erlotinib
`(n ⫽ 69)
`
`11.6
`
`9.4
`33.3
`
`18.9
`3.3
`
`53.8
`66.7
`25.0
`0
`
`0
`15.7
`
`8.0
`18.2
`
`15.0
`10.7
`
`22.4
`
`21.3
`33.3
`
`25.7
`18.8
`
`36.4
`33.3
`40.0
`22.7
`
`20.0
`24.0
`
`28.6
`16.0
`
`27.3
`21.2
`
`46.4
`
`45.3
`66.7
`
`54.1
`36.7
`
`84.6
`100.0
`50.0
`35.6
`
`30.8
`52.9
`
`52.0
`36.4
`
`40.0
`42.9
`
`71.6
`
`72.1
`66.7
`
`74.3
`68.8
`
`81.8
`66.7
`100.0
`68.2
`
`53.3
`78.0
`
`76.2
`56.0
`
`100.0
`57.6
`
`Factor
`
`Overall
`EGFR by IHC status
`Positive
`Negative
`EGFR by FISH status
`Positive
`Negative
`EGFR mutation status
`Mutation
`Activating mutation
`Other mutation
`No mutation
`KRAS mutation status
`Mutation
`No mutation
`E-cadherin
`High
`Low
`Vimentin
`High
`Low
`
`Abbreviations: CR, complete response; PR, partial response; SD, stable
`disease; CP, carboplatin/paclitaxel; IHC, immunohistochemistry; FISH, fluores-
`cent in situ hybridization.
`ⴱOne patient on the CP ⫹ intercalated erlotinib arm had no measurable
`disease at baseline and was nonevaluable for response.
`
`were 59% and 46%, respectively (Table 2; Fig 4). For patients with
`activating EGFR mutations, the 12-month OS rate was 100% in the
`erlotinib arm, and it was 41.7% in the chemotherapy plus erlotinib
`arm. However, in the EGFR WT patients, survival curves were nearly
`overlapping, and the median survival times were 15.6 months and 13.3
`months in the erlotinib and the chemotherapy plus erlotinib arms,
`respectively (Fig 4).
`For the EGFR FISH-positive patients no statistically difference
`was seen between the two treatment arms (12-month OS rates of 62%
`for erlotinib and 55% for chemotherapy plus erlotinib). In the FISH-
`negative group, the 12-months OS rates were 56.5% with erlotinib and
`38.4% with chemotherapy plus erlotinib (Table 2). For the EGFR
`IHC-positive patients, the 12-month OS rates were 56.8% in the
`erlotinib arm and 40.1% in the chemotherapy plus erlotinib arm. No
`difference in survival was seen among patients within the same treat-
`ment arm for KRAS mutation versus WT, E-cadherin high versus low
`expression, or vimentin high versus low expression (Table 2).
`Toxicity. The most common adverse event was skin rash (81%
`[grades 3 to 4, 9%] in erlotinib arm and 76% [grades 3 to 4, 4%] in the
`chemotherapy plus erlotinib arm; Table A2). In the chemotherapy
`plus erlotinib arm, 10 patients (15%) had chemotherapy adjustments
`as a result of hematologic toxicity, and 29 patients (43%) had them as
`a result of nonhematologic toxicity. There was at least one dose
`interruption of erlotinib in 17 patients (25%) in the erlotinib arm and
`in 23 patients (34%) in the chemotherapy plus erlotinib arm.
`
`www.jco.org
`
`© 2011 by American Society of Clinical Oncology
`
`3571
`
`APOTEX EX. 1032-005
`
`

`

`Hirsch et al
`
`control evaluation. Thus, molecular results were available on 80% of
`the patients, demonstrating that molecular phenotyping can be done
`in the majority of patients in a reasonable time frame to select therapy.
`The primary goal was to evaluate treatment outcomes from interca-
`lating erlotinib with chemotherapy and erlotinib alone. In this EGFR-
`selected population, the intercalated therapy provided similar
`outcomes to erlotinib alone on the basis of the primary end point of
`6-month PFS. Neither treatment arm exceeded the historical 6-month
`PFS rate of 45%.4,5
`An exploratory goal was to determine whether erlotinib alone
`could be superior to intercalated therapy in any of the biomarker-
`selected patients. Patients with activating EGFR mutations treated
`with erlotinib alone had superior RRs, superior PFS, and superior OS
`compared with the intercalated therapy arm. The favorable response
`rates and outcome for patients with advanced NSCLC who had EGFR
`mutations and who were treated with EGFR TKIs alone are consis-
`tent with studies evaluating gefitinib in untreated patients with
`advanced-stage NSCLC7,15,16 and with the OPTIMAL study, which
`demonstrated superior PFS with erlotinib alone compared with
`chemotherapy in patients with EGFR mutations.16 In patients with
`EGFR mutations, the lower RR and shorter PFS with intercalated
`therapy suggest antagonism between the treatments.3,8 In patients
`with EGFR WT, the results with intercalated therapy were similar to
`those reported with chemotherapy alone.4,5
`In studies of erlotinib in later lines of therapy, patients with EGFR
`WT treated with erlotinib had a superior survival compared with
`placebo.1,2 Thus, a remaining question is whether a single biomarker
`or combination of biomarkers will help to select patients with EGFR
`WT tumors for EGFR TKI therapy in the first line and beyond. This
`trial does not support use of erlotinib or chemotherapy plus erlotinib
`in the first-line setting in EGFR WT patients who could be selected
`by any other analyzed biomarker, including EGFR FISH or KRAS
`mutations. When the EGFR-mutated patients are excluded from
`the FISH-positive analysis and from the KRAS WT analysis, there
`remains no striking difference in outcome between those treated
`with erlotinib or intercalated therapy on the basis of FISH or KRAS
`status. Our finding is supported by the results from Cancer and
`Leukemia Group B study CALGB 30406, in which no difference
`between erlotinib alone or chemotherapy plus erlotinib was found
`in never smokers or light smokers who had lung adenocarcinomas
`with high response and by the positive outcome in patients with
`EGFR mutations treated with erlotinib alone, which support the
`use of an EGFR TKI alone as first-line therapy in patients with
`NSCLC who have EGFR mutations.17
`In summary, this study could not demonstrate any benefit of
`combining chemotherapy and intercalated EGFR TKI in patients with
`advanced NSCLC. The study demonstrated the feasibility of a pro-
`spective, multi-institutional biomarker study in advanced NSCLC
`and supports the importance of determining the EGFR mutation
`status of patients with advanced NSCLC before initial therapy.
`
`CP + intercalated erlotinib (n = 68)
`Median: 11.43 months
`12-month rate: 46.4%
`
`Erlotinib (n = 69)
`Median: 16.72 months
`12-month rate: 58.6%
`
`1.00
`
`0.75
`
`0.50
`
`0.25
`
`A
`
`Overall Survival
`
`(probability)
`
`0
`
`3
`
`6
`
`9
`
`18
`15
`12
`Time (months)
`
`21
`
`24
`
`27
`
`30
`
`CP + intercalated erlotinib (n = 50)
`Median: 13.34 months
`12-month rate: 55.1%
`
`Erlotinib (n = 49)
`Median: 15.61 months
`12-month rate: 56.4%
`
`1.00
`
`0.75
`
`0.50
`
`0.25
`
`B
`
`Overall Survival
`
`(probability)
`
`0
`
`3
`
`6
`
`9
`
`18
`15
`12
`Time (months)
`
`21
`
`24
`
`27
`
`30
`
`CP + intercalated erlotinib (n = 6)
`Median: 11.43 months
`12-month rate: 41.7%
`Erlotinib (n = 9)
`Median: . months
`12-month rate: 100.0%
`
`1.00
`
`0.75
`
`0.50
`
`0.25
`
`C
`
`Overall Survival
`
`(probability)
`
`0
`
`3
`
`6
`
`9
`
`18
`15
`12
`Time (months)
`
`21
`
`24
`
`27
`
`30
`
`Fig 4. Kaplan-Meier plots of overall survival. (A) All patients; (B) EGFR wild-type
`patients; (C) EGFR mutant patients. CP, carboplatin/paclitaxel.
`
`DISCUSSION
`
`Personalized medicine requires molecular analyses of tumor tissue
`obtained before therapy to select the best treatment. One goal of this
`study was to determine whether molecular tests could be performed
`on lung cancer samples from untreated patients with advanced-stage
`NSCLC in a clinically relevant time frame (defined in this study as ⬍ 5
`working days). We obtained tissue from 215 (89.6%) of 240 screened
`patients without additional rebiopsy. Of these, 11% failed quality
`
`AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS
`OF INTEREST
`
`Although all authors completed the disclosure declaration, the following
`author(s) indicated a financial or other interest that is relevant to the subject
`matter under consideration in this article. Certain relationships marked
`with a “U” are those for which no compensation was received; those
`
`3572
`
`© 2011 by American Society of Clinical Oncology
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`APOTEX EX. 1032-006
`
`

`

`EGFR-Selected Therapy in Advanced NSCLC
`
`relationships marked with a “C” were compensated. For a detailed
`description of the disclosure categories, or for more information about
`ASCO’s conflict of interest policy, please refer to the Author Disclosure
`Declaration and the Disclosures of Potential Conflicts of Interest section in
`Information for Contributors.
`Employment or Leadership Position: Frank Richardson, OSI
`Pharmaceuticals (C); Bret Wacker, OSI Pharmaceuticals (C); David W.
`Sternberg, OSI Pharmaceuticals (C) Consultant or Advisory Role: Fred
`R. Hirsch, Genentech (C), Celgene (C), GlaxoSmithKline (C), Eli Lilly
`(C), OSI Pharmaceuticals (C), Pfizer (C), Boehringer Ingelheim (C),
`Ventana (C); Tim Eisen, Roche (C), AstraZeneca (C); Rafal
`Dziadziuszko, AstraZeneca (C); Katherine Richardson, OSI
`Pharmaceuticals (C); Jason Rusk, OSI Pharmaceuticals (C); Paul A.
`Bunn Jr, AstraZeneca (C), Bayer Pharmaceuticals (C), Boehringer
`Ingelheim (C), Bristol-Myers Squibb (C), Eli Lilly (C), GlaxoSmithKline
`(C), Merck (C), Novartis (C), OSI Pharmaceuticals (C), Genentech (C)
`Stock Ownership: Tim Eisen, AstraZeneca; David W. Sternberg,

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket