Skip to main content
Free access
ORIGINAL REPORTS
October 04, 2010

Randomized, Phase III Trial of Panitumumab With Infusional Fluorouracil, Leucovorin, and Oxaliplatin (FOLFOX4) Versus FOLFOX4 Alone As First-Line Treatment in Patients With Previously Untreated Metastatic Colorectal Cancer: The PRIME Study

Publication: Journal of Clinical Oncology
PDF

Abstract

Purpose

Panitumumab, a fully human anti–epidermal growth factor receptor (EGFR) monoclonal antibody that improves progression-free survival (PFS), is approved as monotherapy for patients with chemotherapy-refractory metastatic colorectal cancer (mCRC). The Panitumumab Randomized Trial in Combination With Chemotherapy for Metastatic Colorectal Cancer to Determine Efficacy (PRIME) was designed to evaluate the efficacy and safety of panitumumab plus infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as initial treatment for mCRC.

Patients and Methods

In this multicenter, phase III trial, patients with no prior chemotherapy for mCRC, Eastern Cooperative Oncology Group performance status of 0 to 2, and available tissue for biomarker testing were randomly assigned 1:1 to receive panitumumab-FOLFOX4 versus FOLFOX4. The primary end point was PFS; overall survival (OS) was a secondary end point. Results were prospectively analyzed on an intent-to-treat basis by tumor KRAS status.

Results

KRAS results were available for 93% of the 1,183 patients randomly assigned. In the wild-type (WT) KRAS stratum, panitumumab-FOLFOX4 significantly improved PFS compared with FOLFOX4 (median PFS, 9.6 v 8.0 months, respectively; hazard ratio [HR], 0.80; 95% CI, 0.66 to 0.97; P = .02). A nonsignificant increase in OS was also observed for panitumumab-FOLFOX4 versus FOLFOX4 (median OS, 23.9 v 19.7 months, respectively; HR, 0.83; 95% CI, 0.67 to 1.02; P = .072). In the mutant KRAS stratum, PFS was significantly reduced in the panitumumab-FOLFOX4 arm versus the FOLFOX4 arm (HR, 1.29; 95% CI, 1.04 to 1.62; P = .02), and median OS was 15.5 months versus 19.3 months, respectively (HR, 1.24; 95% CI, 0.98 to 1.57; P = .068). Adverse event rates were generally comparable across arms with the exception of toxicities known to be associated with anti-EGFR therapy.

Conclusion

This study demonstrated that panitumumab-FOLFOX4 was well tolerated and significantly improved PFS in patients with WT KRAS tumors and underscores the importance of KRAS testing for patients with mCRC.

Introduction

Colorectal cancer (CRC) is the third most common cancer among men and women in the United States, resulting in approximately 175,000 new cancers yearly.1 Worldwide, there are more than one million new cases of CRC each year.2 On the basis of its role in the pathogenesis of CRC, the epidermal growth factor receptor (EGFR) has proven to be a clinically meaningful target for monoclonal antibodies (mAbs) with efficacy established in all lines of treatment of metastatic CRC (mCRC).39 Panitumumab is a fully human mAb targeting the EGFR.
Retrospectively analyzed studies identified KRAS mutation in tumors as a negative predictive factor for panitumumab and cetuximab for improved response rate (RR), progression-free survival (PFS), and overall survival (OS).1016 In September 2007, a prospectively defined, retrospective analysis of the pivotal phase III study of panitumumab as monotherapy in the mCRC setting provided evidence that clinical benefit was specific to patients with wild-type (WT) KRAS tumors.17
The Panitumumab Randomized Trial in Combination With Chemotherapy for Metastatic Colorectal Cancer to Determine Efficacy (PRIME) is an open-label, randomized, multicenter, phase III trial prospectively investigating panitumumab plus infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment for mCRC in patients with WT KRAS tumors. Originally designed to compare the treatment effect in all randomly assigned patients, the trial was amended to focus on prospective hypothesis testing in the WT KRAS stratum.

Patients and Methods

Patients

Eligible patients were ≥ 18 years old, had previously untreated metastatic adenocarcinoma of the colon or rectum, and had an Eastern Cooperative Oncology Group (ECOG) performance status (PS) of 0 to 2. Fluorouracil-based adjuvant chemotherapy was allowed if disease recurrence occurred 6 months after completion; however, prior oxaliplatin was not allowed. At least one measurable lesion (≥ 20 mm) was required. Paraffin-embedded tumor tissue from the primary tumor or metastasis had to be available for central biomarker analyses. EGFR expression and KRAS status were not required at entry. The protocol was approved by the ethics committees at participating sites. All patients signed informed consent before any study-related procedures were performed.

Study Design and Treatment Schedule

This is an open-label, multicenter, phase III trial that compared the efficacy of panitumumab-FOLFOX4 with FOLFOX4 alone in patients with previously untreated mCRC according to tumor KRAS status. Patients were randomly assigned 1:1 to receive either panitumumab-FOLFOX4 or FOLFOX4. Random assignment was stratified by geographic region (Western Europe, Canada, and Australia v rest of the world) and ECOG PS (0 or 1 v 2).
Panitumumab was administered intravenously (IV) over 1 hour at 6 mg/kg every 2 weeks on day 1 before FOLFOX4 chemotherapy. If tolerated, subsequent infusions could be administered over 30 minutes. FOLFOX4 was administered every 2 weeks as oxaliplatin 85 mg/m2 IV infusion on day 1 and leucovorin 200 mg/m2 (or equivalent) IV infusion followed by fluorouracil 400 mg/m2 IV bolus and 600 mg/m2 22-hour continuous infusion on days 1 and 2. Treatment was administered until progression or unacceptable toxicity (Fig 1A).
Fig 1. (A) Panitumumab Randomized Trial in Combination With Chemotherapy for Metastatic Colorectal Cancer to Determine Efficacy (PRIME) study schema. (B) Treatment assignment by KRAS status. Q2W, every 2 weeks; FOLFOX4, infusional fluorouracil, leucovorin, and oxaliplatin; WT, wild type; MT, mutant.
Objective tumor response was evaluated by blinded central radiology review using modified Response Evaluation Criteria in Solid Tumors (RECIST)18 in all patients with baseline measurable disease per central review. Patients were evaluated every 8 weeks until progression. Responses were confirmed at least 4 weeks later. Resections of metastases were reported as being either complete or partial; the status of the surgical margins was not specifically captured. Patients were observed for safety 30 days after the last study drug administration and for survival every 3 months. Adverse events (AEs) were graded using the Common Terminology Criteria for Adverse Events (version 3.0) with modifications for specific skin- and nail-related toxicities. Safety results were summarized for patients who received at least one dose of protocol therapy. Patient-reported outcomes were measured and will be reported separately. An independent data monitoring committee reviewed interim analyses of safety and one descriptive interim analysis of PFS.

KRAS and Antibody Testing

KRAS testing was performed in a blinded central laboratory using allele-specific polymerase chain reaction (DxS, Manchester, United Kingdom), as previously described.17 Testing was initiated after the study population was enrolled and was completed 3 months before the primary analysis (Appendix Fig A1, online only). Serum antipanitumumab antibodies were analyzed as previously described.19

Statistical Analysis

The primary objective of this study was to assess the treatment effect of the addition of panitumumab to FOLFOX4 on PFS (blinded central radiology review) as initial therapy for mCRC in patients with WT KRAS tumors and also in patients with mutant (MT) KRAS tumors. Originally designed to test the treatment effect in all randomly assigned patients (N = 900), the study was amended to compare PFS (primary end point) and OS (secondary end point) according to KRAS status before any efficacy analyses. The sample size was increased to 1,150 to ensure adequate power to test PFS in the WT KRAS population. No multiplicity adjustments were made for end points other than PFS and OS. There was no planned interim analysis by KRAS status after the amendment, because KRAS status was not expected to be available until approximately the time of the primary analysis. A treatment comparison of OS in the WT KRAS stratum and of PFS in the MT KRAS stratum was conditional on first demonstrating a significant difference in PFS (P < .05) in the WT KRAS stratum. A treatment comparison of OS in the MT KRAS stratum was conditional on a significant difference in PFS in the same stratum. The primary OS analysis was planned when at least 50% of patients in both treatment arms had died in the WT KRAS stratum, and a significance level of P = .0499 was used to account for a planned interim OS analysis.
A log-rank test stratified by random assignment factors was used to compare PFS and OS. In the WT KRAS stratum, a hazard ratio (HR) of 0.714 was hypothesized (panitumumab-FOLFOX4 to FOLFOX4). To achieve 90% power for a two-sided, P = .05 significance level in the WT KRAS stratum, a total of 380 PFS events (radiologic progression per modified RECIST by blinded central review or death) and a sample size of 1,150 patients independent of KRAS status were required. Treatment effects on PFS and OS were estimated using stratified Cox proportional hazards models and the Kaplan-Meier method. An exact 95% CI was estimated for a stratified odds ratio for objective RR. The random assignment factors were used for analysis stratification. Descriptive analyses of treatment effects were planned in prospectively identified subsets. After protocol amendment, an interim PFS analysis after 258 events in all randomly assigned patients (because tumor KRAS results were not yet available) was retained to stop for inferior results given an estimated HR ≥ 1.28. At the time of the primary PFS analysis, a P = .001 nominal significance level was used to compare OS in an interim analysis in the WT and MT KRAS strata. The interim OS results were reported along with the primary PFS results (after the cutoff date for the primary OS analysis).20 A prespecified final analysis, which will include OS, is planned after a 2.5-year minimum follow-up period.

Results

Patients

Between August 2006 and February 2008, 1,183 patients were randomly assigned from among 133 institutions in 19 countries; 593 patients (50%) were randomly assigned to receive panitumumab-FOLFOX4, and 590 patients (50%) were randomly assigned to receive FOLFOX4 (Fig 1B). Of these patients, 1,096 patients (93%) had available tumor KRAS status results; 656 patients (60%) had WT KRAS tumors, and 440 patients (40%) had MT KRAS tumors.
Baseline demographics and disease characteristics were generally balanced within each treatment arm and KRAS stratum (Table 1), with the exception that there were more patients with three or more disease sites, elevated baseline carcinoembryonic antigen, and elevated lactate dehydrogenase in the panitumumab-FOLFOX4 arm than in the FOLFOX4 arm in the MT KRAS stratum. For the primary PFS analysis, median follow-up time from random assignment to data cutoff in the WT KRAS stratum was 13.2 months (range, 0 to 25.2 months) for patients who received panitumumab-FOLFOX4 and 12.5 months (range, 0 to 24.7 months) for patients who received FOLFOX4. Median follow-up time was 10.8 months (range, 0.7 to 22.4 months) for patients with MT KRAS status who received panitumumab-FOLFOX4 and 12 months (range, 0.2 to 23.1 months) for patients who received FOLFOX4.
Table 1. Patient Demographics and Clinical Characteristics
Demographic or Clinical Characteristic WT KRAS MT KRAS
Panitumumab- FOLFOX4(n = 325) FOLFOX4 (n = 331) Panitumumab- FOLFOX4(n = 221) FOLFOX4 (n = 219)
No. of Patients % No. of Patients % No. of Patients % No. of Patients %
Male 217 67 204 62 145 66 128 58
Age, years                
    Median 62 61 63 61
    Range 27-85 24-82 33-83 27-82
White race 296 91 307 93 196 89 196 89
ECOG performance status*                
    0-1 305 94 312 94 213 96 209 95
    ≥ 2 20 6 18 5 8 4 10 5
Region: Western Europe, Canada, Australia 194 60 187 56 119 54 122 56
Primary tumor type                
    Colon 214 66 216 65 151 68 160 73
    Rectal 111 34 115 35 70 32 59 27
Site of metastatic disease                
    Liver only 60 18 56 17 32 14 36 16
    Liver + other 223 69 227 69 156 71 159 73
    Other only 40 12 47 14 31 14 23 11
    Missing or unknown 2 1 1 < 1 2 1 1 < 1
No. of sites of disease                
    1 68 21 67 20 39 18 43 20
    2 112 34 116 35 70 32 80 37
    ≥ 3 143 44 147 44 110 50 95 43
CEA > ULN 255 78 255 77 188 85 170 78
LDH ≥ 1.5× ULN 92 28 96 29 73 33 59 27
Prior adjuvant chemotherapy 53 16 55 17 35 16 26 12
Abbreviations: WT, wild type; MT, mutant; FOLFOX4, infusional fluorouracil, leucovorin, and oxaliplatin; ECOG, Eastern Cooperative Oncology Group; CEA, carcinoembryonic antigen; ULN, upper limit of normal; LDH, lactate dehydrogenase.
*
One patient had an unknown ECOG performance status at baseline.

Efficacy

PFS.

In the WT KRAS stratum, there were 414 progression or death events—199 (61%) in the panitumumab-FOLFOX4 arm and 215 (65%) in the FOLFOX4 arm. A statistically significant improvement in PFS was observed with panitumumab-FOLFOX4 compared with FOLFOX4 (HR = 0.80; 95% CI, 0.66 to 0.97; P = .02, stratified log-rank test; Fig 2A). Median PFS was 9.6 months (95% CI, 9.2 to 11.1 months) for panitumumab-FOLFOX4 and 8.0 months (95% CI, 7.5 to 9.3 months) for FOLFOX4. From the planned subgroup analyses for PFS in the WT KRAS stratum, the HR for all subsets was less than 1 (panitumumab-FOLFOX4 v FOLFOX4), with the exception of patients ≥ 65 years old, women, and patients with ECOG PS of 2 (Fig 3A).
Fig 2. Progression-free survival in patients with (A) wild-type (WT) KRAS and (B) mutant (MT) KRAS. Overall survival in patients with (C) WT KRAS and (D) MT KRAS. FOLFOX4, infusional fluorouracil, leucovorin, and oxaliplatin; Panit., panitumumab; HR, hazard ratio.
Fig 3. Wild-type (WT) KRAS by subgroup analyses. (A) Subset analyses of progression-free survival by WT KRAS (panitumumab [Pmab] -FOLFOX4 v FOLFOX4). (B) Subset analyses of overall survival by WT KRAS (Pmab-FOLFOX4 v FOLFOX4). HR, hazard ratio; met, metastasis; ECOG, Eastern Cooperative Oncology Group.
In the MT KRAS stratum, there were 324 progression or death events—167 (76%) in the panitumumab-FOLFOX4 arm and 157 (72%) in the FOLFOX4 arm. PFS was inferior in patients receiving panitumumab-FOLFOX4 versus FOLFOX4 (HR = 1.29; 95% CI, 1.04 to 1.62; P = .02, stratified log-rank test; Fig 2B). Median PFS was 7.3 months (95% CI, 6.3 to 8.0 months) for panitumumab-FOLFOX4 and 8.8 months (95% CI, 7.7 to 9.4 months) for FOLFOX4.

OS.

In the WT KRAS stratum, at the time of the analysis, 355 deaths had occurred—165 (51%) in the panitumumab-FOLFOX4 arm and 190 (57%) in the FOLFOX4 arm. Median OS was 23.9 months (95% CI, 20.3 to 28.3 months) for panitumumab-FOLFOX4 and 19.7 months (95% CI, 17.6 to 22.6 months) for FOLFOX4 arm, resulting in an absolute difference of 4.2 months. The HR was 0.83 (95% CI, 0.67 to 1.02; P = .072) favoring the panitumumab-FOLFOX4 arm (Fig 2C). From planned subgroup analyses for OS, the HR for all subsets was less than 1, with the exception of the small subsets with no liver metastases and ECOG PS ≥ 2 (Fig 3B).
Subsequent to study treatment, 8% of patients in the panitumumab-FOLFOX4 arm and 18% of patients in the FOLFOX4 arm received anti-EGFR mAb therapy. Median time to anti-EGFR therapy from random assignment was 18 months in the panitumumab-FOLFOX4 arm and 11 months in the FOLFOX4 arm. Subsequent chemotherapy (including oxaliplatin, irinotecan, and/or fluoropyrimidine) was received by 62% of patients in the FOLFOX4 arm and 53% of patients in the panitumumab-FOLFOX4 arm and included bevacizumab in 12% and 15% of patients, respectively.
In the MT KRAS stratum, 294 deaths occurred—152 (69%) in the panitumumab-FOLFOX4 arm and 142 (65%) in the FOLFOX4 arm. Median OS was 15.5 months (95% CI, 13.1 to 17.6 months) for panitumumab-FOLFOX4 and 19.3 months (95% CI, 16.5 to 21.8 months) for FOLFOX4 (HR = 1.24; 95% CI, 0.98 to 1.57; P = .068, stratified log-rank test; Fig 2D).
Within the panitumumab-FOLFOX4 arm, the HR for OS for the WT stratum versus MT stratum was 0.57 (95% CI, 0.46 to 0.71; P < .001). In the FOLFOX4 arm, the HR for OS for the WT stratum versus MT stratum was 0.87 (95% CI, 0.70 to 1.08; P = .21).

Objective response.

In the WT KRAS stratum, the RR was 55% for panitumumab-FOLFOX4 and 48% for FOLFOX4 (stratified odds ratio, 1.35; P = .068). In the MT KRAS stratum, the RR was 40% in each arm.

Resection rate.

Metastasectomy of any site was attempted in 10.5% of patients treated with panitumumab-FOLFOX4 and 9.4% of patients treated with FOLFOX4 with WT KRAS status; complete resections were achieved in 8.3% and 7.0% of patients, respectively.

Safety

Grade 3 and 4 AEs of interest are listed in Table 2. The incidence of skin toxicity was 96% for patients receiving panitumumab-FOLFOX4 and 31% for patients receiving FOLFOX4. In the WT KRAS stratum, the rates of AEs in the panitumumab-FOLFOX4 and FOLFOX4 arms were 82% and 63%, respectively, for worst grade 3 or 4 treatment-related AEs (considered by the investigator to be related to chemotherapy and/or panitumumab treatment) and 40% and 36%, respectively, for serious AEs. Fatal AEs were reported in 16 patients (5%) in the panitumumab-FOLFOX4 arm and in 20 patients (6%) in the FOLFOX4 arm and included cases where the primary cause of death was disease progression. In each arm, there were four fatal treatment-related AEs including two that were panitumumab related, pneumonitis (no infectious etiology identified) and pneumonia.
Table 2. Grade 3 and 4 Adverse Events of Interest
Adverse Event by MedDRA Term WT KRAS (n = 649) MT KRAS (n = 435)
Panitumumab- FOLFOX4 (n = 322) FOLFOX4(n = 327) Panitumumab- FOLFOX4 (n = 217) FOLFOX4 (n = 218)
No. of Patients % No. of Patients % No. of Patients % No. of Patients %
Patients with any event 270 84 227 69 173 80 159 73
Neutropenia 136 42 134 41 81 37 103 47
Skin toxicity 116 36 7 2 66 30 3 1
Diarrhea 59 18 29 9 43 20 21 10
Neurologic toxicities 52 16 51 16 36 17 37 17
Hypokalemia 32 10 15 5 19 9 8 4
Fatigue 30 9 10 3 16 7 11 5
Mucositis* 28 9 2 < 1 12 6 6 3
Hypomagnesemia 20 6 1 < 1 13 6 1 < 1
Paronychia 11 3 0 0 4 2 0 0
Pulmonary embolism 9 3 5 2 7 3 8 4
Febrile neutropenia 8 2 7 2 7 3 7 3
Infusion-related reaction (panitumumab) 2 < 1 0 0
NOTE. All events are included, regardless of relatedness to therapy.
Abbreviations: MedDRA, Medical Dictionary for Regulatory Activities; WT, wild type; MT, mutant; FOLFOX4, infusional fluorouracil, leucovorin, and oxaliplatin.
*
Results are based on the following prespecified list of preferred terms: stomatitis, mucosal inflammation, aphthous stomatitis, mouth ulceration, mucosal dryness, and mucosal ulceration.
No grade 4 events.
In the MT KRAS stratum, the AE rates in the panitumumab-FOLFOX4 and FOLFOX4 arms were 79% and 69%, respectively, for worst grade 3 or 4 treatment-related AEs and 47% and 29%, respectively, for serious AEs. Fatal AEs were reported in 17 patients (8%) in the panitumumab-FOLFOX4 arm and in seven patients (3%) in the FOLFOX4 arm; fatal events included cases where the primary cause of death was disease progression. There were three treatment-related fatal AEs, none of which were reported as related to panitumumab (one septic shock in each arm and one febrile neutropenia in the panitumumab-FOLFOX4 arm). Grade 3 panitumumab-related infusion reactions occurred in two patients (0.3%); both patients received additional panitumumab treatment after premedication.

Treatment Exposure

The median number of cycles of panitumumab and chemotherapy and the relative dose-intensity were similar between treatment arms in both the WT and MT KRAS strata (Table 3). The most common reason for chemotherapy discontinuation was disease progression, which occurred in 51% of patients with WT KRAS tumor status and 61% of patients with MT KRAS tumor status. Other reasons for discontinuation included patient request and AEs and were balanced between treatment arms and KRAS strata.
Table 3. Treatment Exposure
Treatment Measure WT KRAS MT KRAS
Panitumumab-FOLFOX4 (n = 322) FOLFOX4 (n = 327) Panitumumab-FOLFOX4 (n = 217) FOLFOX4 (n = 218)
Median No. of cycles received        
    Panitumumab 11 10
    Oxaliplatin 11 11 11 11
    FU (bolus) 12 12 12 12
    FU (continuous infusion) 12 12 12 12
Median relative dose-intensity, %        
    Panitumumab 81 83
    Oxaliplatin 77 79 80 80
    FU (bolus) 77 81 80 81
    FU (continuous infusion) 78 81 81 81
Median cumulative total dose        
    Panitumumab, mg/kg 62 57
    Oxaliplatin, mg/m2 859 865 824 856
    FU (bolus), mg/m2 8,627 8,618 8,294 8,711
    FU (continuous infusion), mg/m2 13,483 13,229 12,878 13,109
Abbreviations: WT, wild type; MT, mutant; FOLFOX4, infusional fluorouracil, leucovorin, and oxaliplatin; FU, fluorouracil.

Antibody

Treatment-emergent binding antipanitumumab antibodies were detected in 14 (3.0%) of 470 patients who received panitumumab. Neutralizing antibodies were detected in postdose samples from two (0.4%) of 470 patients.

Discussion

PRIME is the first study, to our knowledge, to evaluate the impact of the addition of panitumumab to FOLFOX4 for the initial treatment of patients with WT KRAS mCRC. Importantly, the results were prospectively analyzed by KRAS status, and the study achieved the primary objective by demonstrating a significant improvement in the probability of being alive without disease progression when panitumumab is added to chemotherapy for patients with WT KRAS tumors. The difference in median OS for patients with WT KRAS status, although not statistically significant, was also favorable, with a 4.2-month improvement in the panitumumab-FOLFOX4 arm compared with the FOLFOX4 arm. There was a statistically significant interaction between the treatment effect and KRAS tumor status for PFS (Appendix Table A1, online only). The goal of the primary OS analysis was to obtain median estimates, and only 51% of patients had an event in the panitumumab-treated WT KRAS population at the time of this analysis. Responses were more frequent in the panitumumab-FOLFOX4 arm versus the FOLFOX4 arm (Appendix Table A2, online only); however, resection rates were similar.
A total of 87 patients were excluded from analysis who were unevaluable for KRAS, which may have introduced ascertainment bias; however, various prospective and post hoc intent-to-diagnosis sensitivity analyses provided results consistent with the primary analysis for each KRAS stratum. Details are included in Appendix Table A3, online only.
All planned subsets consistently demonstrated favorable effects of panitumumab on PFS, with the exception of patients ≥ 65 years old, women, and the small subset of patients with ECOG PS of 2 (Fig 3A). Importantly, however, a favorable OS effect was observed in women and older patients receiving panitumumab (Fig 3B). In addition, absolute differences in RR were similar between men and women (data not shown).
Safety was as expected when an anti-EGFR mAb is added to chemotherapy and included increased skin toxicity in panitumumab-treated patients. There is evidence that skin toxicity associated with EGFR inhibition can be effectively managed.21 As reported in other studies,3,5,8 an association between skin toxicity and efficacy was observed in PRIME (data not shown). Additional analyses are underway to determine the importance of skin toxicity as a predictive factor after adjusting for other prognostic characteristics. Although diarrhea and hypomagnesemia were increased, these toxicities were clinically manageable based on the infrequency with which they lead to treatment discontinuation (Amgen, data on file). Infusion reactions were rare. Two patients (0.3%) experienced a grade 3 panitumumab-related infusion reaction; both received additional panitumumab treatment.
The positive results in the WT KRAS population in PRIME are consistent with the findings of a recently reported trial in which panitumumab was added to fluorouracil, leucovorin, and irinotecan chemotherapy in previously treated patients with mCRC.22 The effect observed in PRIME is also generally consistent with that seen in two other first-line mCRC studies with cetuximab and chemotherapy, although KRAS status was analyzed retrospectively in those studies.8,9 The 4.2-month benefit in median OS observed in PRIME, although not statistically significant, is comparable to the median OS improvement of 3.5 months reported for the WT KRAS population in the phase III CRYSTAL (Cetuximab Combined With Irinotecan in First-Line Therapy for Metastatic Colorectal Cancer) study with cetuximab and fluorouracil, leucovorin, and irinotecan based on an updated retrospective efficacy analysis.23 In contrast, there was no improvement in either OS or PFS when cetuximab was added to oxaliplatin-based chemotherapy in the recently reported Medical Research Council COIN (Continuous Chemotherapy Versus Intermittent Chemotherapy) trial, which, similar to PRIME, was prospectively analyzed according to tumor KRAS status.24 Efficacy results in the WT KRAS population in PRIME also compared favorably with those reported for the subset of all patients who received FOLFOX4 chemotherapy when bevacizumab was added to treatment.25
Although data from the PRIME trial demonstrate that KRAS is not a prognostic biomarker in this treatment setting (the outcomes of patients with MT KRAS receiving FOLFOX4 were generally similar to those of patients with WT KRAS with respect to RR, PFS, and OS), this trial does confirm the importance of KRAS as a predictive biomarker of efficacy for anti-EGFR mAb therapy. As opposed to the positive effects seen in the WT KRAS strata, in the MT KRAS strata, the addition of panitumumab resulted in a significant detrimental effect on PFS. A similar detrimental effect was also observed in patients with MT KRAS tumors when cetuximab was used in combination with oxaliplatin.9 There were no appreciable differences in chemotherapy exposure or AEs to explain this finding, and the mechanism is still unknown. These results emphasize the importance of incorporating early KRAS testing into treatment algorithms for patients with mCRC.
Consistent with findings from studies of panitumumab in patients with previously treated mCRC,3,22 panitumumab, when added to FOLFOX4, significantly improved PFS for patients with previously untreated WT KRAS mCRC. Other clinically meaningful attributes of panitumumab include that severe panitumumab-related infusion reactions are rare and a standard 2-week dosing regimen allows for synchronization with chemotherapy dosing and minimizes the number of required clinic visits. In conclusion, panitumumab is well tolerated and effective when added to FOLFOX4. This combination represents a relevant and new therapeutic option for the treatment of patients with WT KRAS mCRC. Efforts are underway to identify additional predictive biomarkers for anti-EGFR therapy within the WT KRAS population.

Acknowledgment

We thank the patients, their families, and the site staff. We thank the Amgen country staff and study team: Pamela Ward, Byron Espina, Sarah Edgington, Nora Burr, Feng Xu, and Judith Dionisio. Finally, we thank Barbara Schreader of Amgen for medical writing assistance.
Presented in part at the 43rd Annual Meeting of the American Society of Clinical Oncology, June 1-5, 2007, Chicago, IL; the 2008 Gastrointestinal Cancers Symposium, January 25-27, 2008, Orlando, FL; the joint 15th Congress of the European Cancer Organisation and 34th Congress of the European Society for Medical Oncology, September 20-24, 2009, Berlin, Germany; the 6th Annual Meeting of the International Society of Gastrointestinal Oncology, October 1-3, 2009, Philadelphia, PA; the 2009 National Cancer Research Institute Cancer Conference, October 4-7, 2009, Birmingham, United Kingdom; and the 2010 Gastrointestinal Cancers Symposium, January 22-24, 2010, Orlando, FL.
See accompanying editorial on page 4668 and article on page 4706.
Clinical trial information can be found for the following: NCT00364013.
J Clin Oncol Douillard et al 28: 10.1200/JCO.2009.27.4860

Publisher's Note

The article by Douillard et al, entitled "Randomized, Phase III Trial of Panitumumab With Infusional Fluorouracil, Leucovorin, and Oxaliplatin (FOLFOX4) Versus FOLFOX4 Alone As First-Line Treatment in Patients With Previously Untreated Metastatic Colorectal Cancer: The PRIME Study" (J Clin Oncol 10.1200/JCO.2009.27.4860), was published online October 4, 2010, with errors.

In Figure 2B, erroneous data were given for the number of patients at risk receiving both panitumumab + FOLFOX4 and FOLFOX4 alone. This has been corrected as of October 25, 2010. Journal of Clinical Oncology apologizes to the authors and readers for the mistakes.

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 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: Kelly S. Oliner, Amgen (C); Michael Wolf, Amgen (C); Jennifer Gansert, Amgen (C) Consultant or Advisory Role: Jean-Yves Douillard, Amgen (C), sanofi-aventis (C), Merck Serono (C); Salvatore Siena, Amgen (C), AstraZeneca (C), Merck Serono (C), Roche (C), Celgene (C); James Cassidy, Roche (C), sanofi-aventis (C), Amgen (C), Merck Serono (C); Josep Tabernero, Amgen (C), Merck Serono (C), ImClone Systems (C); Ronald Burkes, Amgen (C) Roche (C), sanofi-aventis (C), Eli Lilly (C); Mario Barugel, Amgen (C), Roche (C); Yves Humblet, Amgen (C), Merck Serono (C), Roche (C); György Bodoky, Amgen (C), Merck Serono (C); David Cunningham, Amgen (C); Paul Ruff, Amgen (C), Novartis (C); Fernando Rivera, Amgen (C); Jean-Luc Canon, Amgen (C); Mark Rother, Amgen (C) Stock Ownership: Kelly S. Oliner, Amgen; Michael Wolf, Amgen; Jennifer Gansert, Amgen Honoraria: Jean-Yves Douillard, Amgen, sanofi-aventis; James Cassidy, Amgen, Roche, sanofi-aventis; Ronald Burkes, Amgen, Roche, sanofi-aventis, Eli Lilly; Mario Barugel, Roche; Yves Humblet, Amgen, Merck Serono; György Bodoky, Amgen, Roche; Fernando Rivera, Amgen; Paul Ruff, Amgen, Merck Serono, Pfizer; Jean-Luc Canon, Amgen; Mark Rother, Amgen Research Funding: James Cassidy, Roche, Amgen; Yves Humblet, Amgen, Merck Serono, Bayer Pharmaceuticals; David Cunningham, Amgen, Roche, Merck Serono; Fernando Rivera, Amgen; Paul Ruff, Amgen, Novartis, Roche, Merck Serono, Pfizer; Jean-Luc Canon, Amgen Expert Testimony: None Other Remuneration: None

References

1.
A Jemal, R Siegel, E Ward, etal: Cancer statistics, 2009 CA Cancer J Clin 59: 225– 249,2009
2.
DM Parkin, F Bray, J Ferlay, etal: Global cancer statistics, 2002 CA Cancer J Clin 55: 74– 108,2005
3.
E Van Cutsem, M Peeters, S Siena, etal: Open-label phase III trial of panitumumab plus best supportive care compared with best supportive care alone in patients with chemotherapy-refractory metastatic colorectal cancer J Clin Oncol 25: 1658– 1664,2007
4.
D Cunningham, Y Humblet, S Siena, etal: Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer N Engl J Med 351: 337– 345,2004
5.
DJ Jonker, CJ O'Callaghan, CS Karapetis, etal: Cetuximab for the treatment of colorectal cancer N Engl J Med 357: 2040– 2048,2007
6.
H Wilke, R Glynne-Jones, J Thaler, etal: Cetuximab plus irinotecan in heavily pretreated metastatic colorectal cancer progressing on irinotecan: MABEL Study J Clin Oncol 26: 5335– 5343,2008
7.
AF Sobrero, J Maurel, L Fehrenbacher, etal: EPIC: Phase III trial of cetuximab plus irinotecan after fluoropyrimidine and oxaliplatin failure in patients with metastatic colorectal cancer J Clin Oncol 26: 2311– 2319,2008
8.
E Van Cutsem, CH Köhne, E Hitre, etal: Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer N Engl J Med 360: 1408– 1417,2009
9.
C Bokemeyer, I Bondarenko, A Makhson, etal: Fluorouracil, leucovorin, and oxaliplatin with and without cetuximab in the first-line treatment of metastatic colorectal cancer J Clin Oncol 27: 663– 671,2009
10.
S Benvenuti, A Sartore-Bianchi, F Di Nicolantonio, etal: Oncogenic activation of the RAS/RAF signaling pathway impairs the response of metastatic colorectal cancers to anti-epidermal growth factor receptor antibody therapies Cancer Res 67: 2643– 2648,2007
11.
F Di Fiore, F Blanchard, F Charbonnier, etal: Clinical relevance of KRAS mutation detection in metastatic colorectal cancer treated by cetuximab plus chemotherapy Br J Cancer 96: 1166– 1169,2007
12.
W De Roock, H Piessevaux, J De Schutter, etal: KRAS wild-type state predicts survival and is associated to early radiological response in metastatic colorectal cancer treated with cetuximab Ann Oncol 19: 508– 515,2008
13.
DJ Freeman, T Juan, M Reiner, etal: Association of K-ras mutational status and clinical outcomes in patients with metastatic colorectal cancer receiving panitumumab alone Clin Colorectal Cancer 7: 184– 190,2008
14.
A Lièvre, JB Bachet, V Boige, etal: KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab J Clin Oncol 26: 374– 379,2008
15.
CS Karapetis, S Khambata-Ford, DJ Jonker, etal: K-ras mutations and benefit from cetuximab in advanced colorectal cancer N Engl J Med 359: 1757– 1765,2008
16.
A Jimeno, WA Messersmith, FR Hirsch, etal: KRAS mutations and sensitivity to epidermal growth factor receptor inhibitors in colorectal cancer: Practical application of patient selection J Clin Oncol 27: 1130– 1136,2009
17.
RG Amado, M Wolf, M Peeters, etal: Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer J Clin Oncol 26: 1626– 1634,2008
18.
P Therasse, SG Arbuck, EA Eisenhauer, etal: New guidelines to evaluate the response to treatment in solid tumors: European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada J Natl Cancer Inst 92: 205– 216,2000
19.
JA Lofgren, S Dhandapani, JJ Pennucci, etal: Comparing ELISA and surface plasmon resonance for assessing clinical immunogenicity of panitumumab J Immunol 178: 7467– 7472,2007
20.
J Douillard, S Siena, J Cassidy, etal: Randomized phase 3 study of panitumumab with FOLFOX4 compared to FOLFOX4 alone as 1st-line treatment (tx) for metastatic colorectal cancer (mCRC): the PRIME trial Eur J Cancer Supplements 7: 6,2009 abstr 10LBA
21.
ME Lacouture, EP Mitchell, B Piperdi, etal: Skin Toxicity Evaluation Protocol With Panitumumab (STEPP), a phase II, open-label, randomized trial evaluating the impact of a pre-emptive skin treatment regimen on skin toxicities and quality of life in patients with metastatic colorectal cancer J Clin Oncol 28: 1351– 1357,2010
22.
M Peeters, TJ Price, A Cervantes, etal: Randomized phase III study of panitumumab with fluorouracil, leucovorin, and irinotecan (FOLFIRI) compared with FOLFIRI alone as second-line treatment in patients with metastatic colorectal cancer J Clin Oncol 28: 4706– 4713,2010
23.
E Van Cutsem, P Rougier, C Köhne, etal: A meta-analysis of the CRYSTAL and OPUS studies combining cetuximab with chemotherapy (CT) as 1st-line treatment for patients (pts) with metastatic colorectal cancer (mCRC): Results according to KRAS and BRAF mutation status Eur J Cancer Supplements 7: 345,2009 abstr 6007
24.
T Maughan, RA Adams, CG Smith, etal: Addition of cetuximab to oxaliplatin-based combination chemotherapy (CT) in patients with KRAS wildtype advanced colorectal cancer (ACRC): A randomised superiority trial (MRC COIN) Eur J Cancer Supplements 7: 4,2009 abstr 6LBA
25.
LB Saltz, S Clarke, E Díaz-Rubio, etal: Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: A randomized phase III study J Clin Oncol 26: 2013– 2019,2008

Appendix

Investigators of the Panitumumab Randomized Trial in Combination With Chemotherapy for Metastatic Colorectal Cancer to Determine Efficacy (PRIME) are listed in Table A4.
Fig A1. Panitumumab Randomized Trial in Combination With Chemotherapy for Metastatic Colorectal Cancer to Determine Efficacy (PRIME) study timeline. (*) Original enrollment goal was achieved at the time of the amendment. PFS, progression-free survival; ITT, intent to treat; DMC, data monitoring committee; CRC, colorectal cancer; BSC, best supportive care; OS, overall survival.
Table A1. Predictive Value of KRAS Status Using Quantitative Interaction Test for Treatment Effect on PFS and OS
End Point Value
PFS  
    WT ITD KRAS patients  
        No. of patients 738
        No. of events 474
        HR (panitumumab-FOLFOX4 v FOLFOX4) 0.853
            95% CI 0.710 to 1.023
    MT KRAS patients  
        No. of patients 436
        No. of events 321
        HR (panitumumab-FOLFOX4 v FOLFOX4) 1.261
            95% CI 1.006 to 1.581
    P for quantitative interaction test .0083
OS  
    WT ITD KRAS patients  
        No. of patients 738
        No. of events 268
        HR (panitumumab-FOLFOX4 v FOLFOX4) 0.801
            95% CI 0.628 to 1.022
    MT KRAS patients  
        No. of patients 436
        No. of events 193
        HR (panitumumab-FOLFOX4 v FOLFOX4) 1.609
            95% CI 1.199 to 2.161
    P for quantitative interaction test < .001
NOTE. For PFS, an analysis was planned to test for a treatment by KRAS interaction at a two-sided 5% significance level. OS was analyzed descriptively. A quantitative interaction test (Gail M, Simon R: Biometrics 41:361-372, 1985) was used to compare the relative treatment effects between the WT ITD and MT KRAS analysis sets with KRAS unevaluable patients included in the WT ITD set. For both PFS and OS, the relative treatment effect was estimated by the log HR (panitumumab-FOLFOX4 v FOLFOX4 alone) from a Cox model adjusted by the random assignment factors (Eastern Cooperative Oncology Group performance status and geographic region) and stratified by quintiles of propensity scores estimated from a logistic regression model with prespecified covariates for the probability of being in the panitumumab treatment group. The analysis excluded patients with covariate missing values. The analyses of PFS were based on data from the blinded central radiology review.
Abbreviations: PFS, progression-free survival; OS, overall survival; WT, wild type; ITD, intent to diagnose; HR, hazard ratio; FOLFOX4, infusional fluorouracil, leucovorin, and oxaliplatin; MT, mutant.
Table A2. Efficacy by KRAS Stratum and Treatment Arm
Efficacy WT KRAS MT KRAS
Panitumumab-FOLFOX4 FOLFOX4 P Panitumumab-FOLFOX4 FOLFOX4 P
Survival            
    No. of patients 325 331   221 219  
    Progression-free survival*     .02     .02
        Progression event            
            No. of patients 199 215   167 157  
            % 61 65   76 72  
        Median progression-free survival, months 9.6 8.0   7.3 8.8  
            95% CI 9.2 to 11.1 7.5 to 9.3   6.3 to 8.0 7.7 to 9.4  
    Overall survival     .07     .07
        Deaths            
            No. of patients 165 190   152 142  
            % 51 57   69 65  
        Median overall survival, months 23.9 19.7   15.5 19.3  
            95% CI 20.3 to 28.3 17.6 to 22.6   13.1 to 17.6 16.5 to 21.8  
Response*            
    No. of patients 317 323   215 211  
    Objective response rate     .07     .98
        No. of patients 175 154   85 85  
        % 55 48   40 40  
        95% CI 50 to 61 42 to 53   33 to 46 34 to 47  
    Complete response            
        No. of patients 0 1   0 0  
        % 0 < 1   0 0  
    Partial response            
        No. of patients 175 153   85 85  
        % 55 47   40 40  
    Stable disease            
        No. of patients 95 117   81 91  
        % 30 36   38 43  
Abbreviations: WT, wild type; MT, mutant; FOLFOX4, infusional fluorouracil, leucovorin, and oxaliplatin.
*
By central radiology review.
Included only patients With baseline measurable disease per central review.
Table A3. Intent-to-Diagnose Sensitivity Analysis for PFS and OS With KRAS Status Imputed for Unevaluable Patients Based on Observed Prevalence
End Point* WT KRAS Evaluable (n = 656) WT KRAS Evaluable + Imputed (n = 708) MT KRAS Evaluable (n = 440) MT KRAS Evaluable + Imputed (n = 475)
PFS        
    HR 0.80 0.81 1.29 1.26
        95% CI 0.66 to 0.97 0.79 to 0.83 1.04 to 1.62 1.21 to 1.32
    P .024 .027 .023 .036
        95% CI NA .012 to .049 NA .011 to .081
OS        
    HR 0.83 0.84 1.24 1.22
        95% CI 0.67 to 1.02 0.81 to 0.87 0.98 to 1.57 1.16 to 1.27
    P .073 .090 .069 .093
        95% CI NA .042 to .157 NA .035 to .183
Abbreviations: PFS, progression-free survival; OS, overall survival; WT, wild type; MT, mutant; HR, hazard ratio; NA, not available.
*
HR is from proportional hazards model stratified by random assignment factors; P value is for the same HR; columns for evaluable + imputed display the mean and 95% CI of parameter estimates from 5,000 simulated trials.
Prospective primary analysis limited to patients evaluable for KRAS status.
Table A4. PRIME Study Investigators
Country Investigators
Argentina Pedro Eugenia Alvarez, Mario Barugel, Cesar Blajman, María Andrea Cuevas, Mauricio Kotliar, Marcelo Tatángelo, María Viniegra, Raul Cesar Wainstein
Australia Poh See Choo, Stephen Clarke, Andrew Haydon
Belgium Jean-Luc Canon, Frédéric Forget, Geert D Haens, Yves Humblet, Bart Neyns, Marc Peeters, Eric Van Cutsem, Jean-Luc Van Laethem
Brazil Carlos Barrios, Carlos Beato, Adilson Faccio, Rochelle Farias, Paulo Hoff, Yeni Nascimento, Francisco Paulo Rodolfo Radke, Ronaldo Ribeiro, Christian Sutmöller, Brigitte Van Eyll
Canada Natalie Aucoin, Ronald Burkes, Haji Chalchal, Felix Couture, Richard Dalfen, Catherine Doyle, John Gapski, Rakesh Goel, Kamal-Uddin Haider, Cynthia Luk, Barbara Melosky, Karen Mulder, Jonathan Noble, Bryn Pressnail, Stephen Reingold, Mark Rother, Richard Shao, Silvana Spadafora, David Walde, Rafal Wierzbicki, Jonathon Wilson, Ralph Wong, Labib Zibdawi
Chile Monica Campos, Hans Harbst, Waldo Ortuzar, Marcelo Adan Garrido Salvo, Eduardo Yañez
Costa Rica Raul Fernando Falla, Rene Valladares
Czech Republic Ilona Kocakova, Milan Kohoutek, Jana Prausová, Martin Šmakal
Estonia Krista Leppik
France Jean-Yves Douillard, Faress Husseini
Hungary Gyorgy Bodoky, Jozsef Cseh, Zsuzsanna Papai, Béla Piko, Agnes Ruzsa
Italy Oscar Alabiso, Carlo Barone, Stefano Cascinu, Luigi Cavanna, Silvana Chiara, Libero Ciuffreda, Pier Franco Conte, Francesco Di Costanzo, Luigi Dogliotti, Giampietro Gasparini, Vittorio Gebbia, Andrea Martoni, Rodolfo Mattioli, Salvatore Siena, Alberto Sobrero, Claudio Verusio, Eugenio Villa, Alberto Zaniboni
Latvia Iveta Kudaba, Gunta Purkalne
Mexico Germán Calderillo, Daniel Capdeville, Jesus Cardenas, Hector Gonzalez, Jesus Lopez, Jhony Alberto De la Cruz Vargas
Poland Maria Basiska-Morawiec, Andrzej Deptata, Jacek Jassem, Andrzej Kaiser, Piotr Koralewski, Andrzej Mruk, Marek Nowacki, Joanna Pikiel, Wojciech Rogowski, Piotr Tomczak, Marek Wojtukiewicz, Iwona Zander
South Africa Graham Cohen, Louis Goedhals, Diana Pillay, Johannes Raats, Bernado Rapoport, Barbara Robertson, Paul Ruff
Spain José Enrique Alés, Andrés Cervantes, Mauel Constenla, Pilar García Alfonso, Cristina Grávalos, Monica Guillot, Joan Maurel, Fernando Rivera, Javier Sastre, Josep Tabernero
Switzerland Markus Borner, Daniel Rauch, Arnaud Roth
United Kingdom James Cassidy, David Cunningham, Mark Harrison, Paul Ross, Anne Thomas
Abbreviation: PRIME, Panitumumab Randomized Trial in Combination With Chemotherapy for Metastatic Colorectal Cancer to Determine Efficacy.

Information & Authors

Information

Published In

Journal of Clinical Oncology
Pages: 4697 - 4705
PubMed: 20921465

History

Published online: October 04, 2010
Published in print: November 01, 2010

Permissions

Request permissions for this article.

Authors

Affiliations

Jean-Yves Douillard [email protected]
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Salvatore Siena
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
James Cassidy
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Josep Tabernero
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Ronald Burkes
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Mario Barugel
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Yves Humblet
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
György Bodoky
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
David Cunningham
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Jacek Jassem
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Fernando Rivera
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Ilona Kocákova
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Paul Ruff
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Maria Błasińska-Morawiec
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Martin Šmakal
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Jean-Luc Canon
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Mark Rother
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Kelly S. Oliner
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Michael Wolf
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.
Jennifer Gansert
From the Centre René Gauducheau, Nantes, France; Ospedale Niguarda Ca' Granda, Milan, Italy; The Beatson West of Scotland Cancer Centre, Glasgow; The Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Vall d'Hebron University Hospital, Barcelona; Hospital Universitario Marqués de Valdecilla, Santander, Spain; Mount Sinai Hospital, Toronto; The Credit Valley Hospital, Mississauga, Ontario, Canada; Hospital de Gastroenterología, Buenos Aires, Argentina; Université Catholique de Louvain, Brussels; Grand Hôpital de Charleroi, Charleroi, Belgium; Szent Laszlo Hospital, Budapest, Hungary; Medical University of Gdansk, Gdansk; Wojewodzki Szpital Specjalistyczny, im. M. Kopernika, Lodz, Poland; Masarykuv Onkologicky Ustav, Brno; Institut Onkologie a Rehabilitace na Plesi s.r.o., Nová Ves pod Pleší, Czech Republic; University of the Witwatersrand, Johannesburg, South Africa; and Amgen, Thousand Oaks, CA.

Notes

Corresponding author: Jean-Yves Douillard, MD, PhD, Centre René Gauducheau, Bd J Monod, 44805 St-Herblain, France; e-mail: [email protected].

Author Contributions

Conception and design: Jean-Yves Douillard, Salvatore Siena, Josep Tabernero, Martin Šmakal, Michael Wolf, Jennifer Gansert
Administrative support: Martin Šmakal
Provision of study materials or patients: Jean-Yves Douillard, Salvatore Siena, James Cassidy, Josep Tabernero, Ronald Burkes, Mario Barugel, Yves Humblet, David Cunningham, Jacek Jassem, Fernando Rivera, Ilona Kocákova, Maria Błasińska-Morawiec, Martin Šmakal, Mark Rother
Collection and assembly of data: Salvatore Siena, Josep Tabernero, Ronald Burkes, Ilona Kocákova, Paul Ruff, Martin Šmakal, Mark Rother, Kelly S. Oliner, Michael Wolf, Jennifer Gansert
Data analysis and interpretation: Jean-Yves Douillard, Salvatore Siena, Josep Tabernero, Yves Humblet, György Bodoky, David Cunningham, Martin Šmakal, Michael Wolf, Jennifer Gansert
Manuscript writing: Jean-Yves Douillard, Salvatore Siena, Josep Tabernero, Ronald Burkes, Jacek Jassem, Paul Ruff, Michael Wolf, Jennifer Gansert
Final approval of manuscript: Jean-Yves Douillard, Salvatore Siena, James Cassidy, Josep Tabernero, Ronald Burkes, Mario Barugel, Yves Humblet, György Bodoky, David Cunningham, Jacek Jassem, Fernando Rivera, Ilona Kocákova, Paul Ruff, Maria Błasińska-Morawiec, Martin Šmakal, Jean-Luc Canon, Mark Rother, Kelly S. Oliner, Michael Wolf, Jennifer Gansert

Disclosures

Authors' disclosures of potential conflicts of interest and author contributions are found at the end of this article.

Funding Information

Supported by Amgen, Thousand Oaks, CA.

Metrics & Citations

Metrics

Altmetric

Citations

Article Citation

Download Citation

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click Download.

For more information or tips please see 'Downloading to a citation manager' in the Help menu.

Format





Download article citation data for:
Jean-Yves Douillard, Salvatore Siena, James Cassidy, Josep Tabernero, Ronald Burkes, Mario Barugel, Yves Humblet, György Bodoky, David Cunningham, Jacek Jassem, Fernando Rivera, Ilona Kocákova, Paul Ruff, Maria Błasińska-Morawiec, Martin Šmakal, Jean-Luc Canon, Mark Rother, Kelly S. Oliner, Michael Wolf, Jennifer Gansert
Journal of Clinical Oncology 2010 28:31, 4697-4705

View Options

View options

PDF

View PDF

Get Access

Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login

Purchase Options

Purchase this article to get full access to it.

Purchase this Article

Subscribe

Subscribe to this Journal
Renew Your Subscription
Become a Member

Media

Figures

Other

Tables

Share

Share

Share article link

Share