scispace - formally typeset
Open AccessJournal ArticleDOI

Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer

TLDR
Panitumumab monotherapy efficacy in mCRC is confined to patients with WT KRAS tumors, and KRAS status should be considered in selecting patients withmCRC as candidates for panitumuab mon Therapy.
Abstract
Purpose Panitumumab, a fully human antibody against the epidermal growth factor receptor (EGFR), has activity in a subset of patients with metastatic colorectal cancer (mCRC). Although activating mutations in KRAS, a small G-protein downstream of EGFR, correlate with poor response to anti-EGFR antibodies in mCRC, their role as a selection marker has not been established in randomized trials. Patients and Methods KRAS mutations were detected using polymerase chain reaction on DNA from tumor sections collected in a phase III mCRC trial comparing panitumumab monotherapy to best supportive care (BSC). We tested whether the effect of panitumumab on progression-free survival (PFS) differed by KRAS status. Results KRAS status was ascertained in 427 (92%) of 463 patients (208 panitumumab, 219 BSC). KRAS mutations were found in 43% of patients. The treatment effect on PFS in the wild-type (WT) KRAS group (hazard ratio [HR], 0.45; 95% CI: 0.34 to 0.59) was significantly greater (P .0001) than in the mutant group (HR, 0.99; 95% CI, 0.73 to 1.36). Median PFS in the WT KRAS group was 12.3 weeks for panitumumab and 7.3 weeks for BSC. Response rates to panitumumab were 17% and 0%, for the WT and mutant groups, respectively. WT KRAS patients had longer overall survival (HR, 0.67; 95% CI, 0.55 to 0.82; treatment arms combined). Consistent with longer exposure, more grade III treatment-related toxicities occurred in the WT KRAS group. No significant differences in toxicity were observed between the WT KRAS group and the overall population. Conclusion Panitumumab monotherapy efficacy in mCRC is confined to patients with WT KRAS tumors. KRAS status should be considered in selecting patients with mCRC as candidates for panitumumab monotherapy.

read more

Content maybe subject to copyright    Report

Wild-Type KRAS Is Required for Panitumumab Efficacy in
Patients With Metastatic Colorectal Cancer
Rafael G. Amado, Michael Wolf, Marc Peeters, Eric Van Cutsem, Salvatore Siena, Daniel J. Freeman,
Todd Juan, Robert Sikorski, Sid Suggs, Robert Radinsky, Scott D. Patterson, and David D. Chang
From Amgen Inc, Thousand Oaks, CA;
Ghent University Hospital, Ghent,
Belgium; University Hospital Gasthuis-
berg, Leuven, Belgium; and the Ospe-
dale Niguarda Ca’ Granda, Milan, Italy.
Submitted October 1, 2007; accepted
November 20, 2007; published online
ahead of print at www.jco.org on
March 3, 2008.
Funded by Amgen Inc, Thousand
Oaks, CA.
Presented in part in oral format at
the 14th European Cancer Confer-
ence, Barcelona, Spain, September
23-27, 2007; and the American Soci-
ety of Clinical Oncology Gastrointesti-
nal Cancer Symosium, Orlando, FL,
January 25-27, 2008.
Authors’ disclosures of potential con-
flicts of interest and author contribu-
tions are found at the end of this
article.
Corresponding author: Rafael G.
Amado, MD, Amgen, Inc, One Amgen
Center Dr, MS 38-2-B, Thousand Oaks,
CA 91320-1799; e-mail: ramado@
amgen.com.
© 2008 by American Society of Clinical
Oncology
0732-183X/08/2610-1626/$20.00
DOI: 10.1200/JCO.2007.14.7116
ABSTRACT
Purpose
Panitumumab, a fully human antibody against the epidermal growth factor receptor (EGFR), has
activity in a subset of patients with metastatic colorectal cancer (mCRC). Although activating
mutations in KRAS, a small G-protein downstream of EGFR, correlate with poor response to
anti-EGFR antibodies in mCRC, their role as a selection marker has not been established in
randomized trials.
Patients and Methods
KRAS mutations were detected using polymerase chain reaction on DNA from tumor sections
collected in a phase III mCRC trial comparing panitumumab monotherapy to best supportive care
(BSC). We tested whether the effect of panitumumab on progression-free survival (PFS) differed
by KRAS status.
Results
KRAS status was ascertained in 427 (92%) of 463 patients (208 panitumumab, 219 BSC). KRAS
mutations were found in 43% of patients. The treatment effect on PFS in the wild-type (WT) KRAS
group (hazard ratio [HR], 0.45; 95% CI: 0.34 to 0.59) was significantly greater (P .0001) than in
the mutant group (HR, 0.99; 95% CI, 0.73 to 1.36). Median PFS in the WT KRAS group was 12.3
weeks for panitumumab and 7.3 weeks for BSC. Response rates to panitumumab were 17% and
0%, for the WT and mutant groups, respectively. WT KRAS patients had longer overall survival
(HR, 0.67; 95% CI, 0.55 to 0.82; treatment arms combined). Consistent with longer exposure,
more grade III treatment-related toxicities occurred in the WT KRAS group. No significant
differences in toxicity were observed between the WT KRAS group and the overall population.
Conclusion
Panitumumab monotherapy efficacy in mCRC is confined to patients with WT KRAS tumors. KRAS
status should be considered in selecting patients with mCRC as candidates for panitu-
mumab monotherapy.
J Clin Oncol 26:1626-1634. © 2008 by American Society of Clinical Oncology
INTRODUCTION
Epidermal growth factor receptor (EGFR) has been
validated as a therapeutic target in several human
tumors, including colorectal cancer (CRC).
1-4
Li-
gand occupancy of the EGFR activates the RAS/
RAF/MAPK, STAT, and PI3K/AKT signaling
pathways, which together modulate cellular prolif-
eration, adhesion, angiogenesis, migration, and
survival.
5,6
The anti-EGFR targeted antibodies
cetuximab and panitumumab administered as
monotherapy in CRC have shown response and dis-
ease stabilization rates of approximately 10% and
30%, respectively.
2,3
Although EGFR expression is
used for patient selection, clinical experience shows
that the level of EGFR expression as measured by
immunohistochemistry does not predict clini-
cal benefit.
2,7-9
KRAS, the human homolog of the Kirsten rat
sarcoma-2 virus oncogene, encodes a small GTP-
binding protein that acts as a self-inactivating signal
transducer by cycling from GDP- to GTP-bound
states in response to stimulation of a cell surface
receptor, including EGFR.
10,11
KRAS can harbor on-
cogenic mutations that yield a constitutively active
protein.
10-13
Such mutations are found in approxi-
mately 30% to 50% of CRC tumors and are com-
mon in other tumor types.
12,14-19
Several studies
have indicated that the presence of mutant KRAS in
lung and CRC tumors correlates with poor progno-
sis,
14,17,18,20
and is associated with lack of response to
EGFR inhibitors.
15,16,19,21,22
These published re-
ports investigating the role of KRAS as a selection
JOURNAL OF CLINICAL ONCOLOGY
ORIGINAL REPORT
VOLUME 26 NUMBER 10 APRIL 1 2008
1626 © 2008 by American Society of Clinical Oncology
Information downloaded from jco.ascopubs.org and provided by at Malliani on December 28, 2014 from 159.149.103.9
Copyright © 2008 American Society of Clinical Oncology. All rights reserved.

marker for EGFR inhibitor treatment were based on tumor samples
from uncontrolled studies and included patients treated with anti-
EGFR antibodies alone or in combination with irinotecan. Given the
possible prognostic role of KRAS mutational status, these uncon-
trolled studies could not isolate the relative effect of antibody treat-
ment on outcome by KRAS status from the prognostic implications of
KRAS as a marker of poor clinical outcome in CRC.
We assessed the predictive role of KRAS in a phase III, random-
ized trial comparing panitumumab monotherapy with best support-
ive care (BSC) in patients with chemotherapy-refractory metastatic
CRC.
3
The primary objective of the biomarker analyses was to deter-
mine whether the effect of panitumumab monotherapy on
progression-free survival (PFS) differed between patients whose tu-
mors contain mutant versus wild-type (WT; ie, nonmutated) KRAS.
PATIENTS AND METHODS
Trial Design and Patient Population
The design of this controlled, panitumumab monotherapy study has
been previously described.
3
Briefly, patients with metastatic CRC with EGFR
expression in 1% of tumor cells (assessed by immunohistochemistry) and
documented evidence of disease progression after failure of fluoropyrimidines
and prespecified exposure to oxaliplatin and irinotecan were randomly as-
signed to panitumumab 6 mg/kg plus BSC every 2 weeks or BSC alone. BSC
patients could receive panitumumab after disease progression. Tumor status
was assessed radiographically every 4 to 8 weeks from week 8 until disease
progression using the Response Evaluation Criteria in Solid Tumors by
blinded central review. The primary end point was PFS, defined as the interval
from random assignment to radiologic progression or death. Secondary end
points included objective response rate, overall survival (OS), and safety. All
patients, including those with unassessable or missing assessments, were in-
cluded in the response rate analysis. A best response of stable disease was
determined at or after week 8 from random assignment. At enrollment, pa-
tients provided informed consent for study procedures including research on
archived paraffin-embedded tumor samples (mostly from primary tumor
resection) for identification of predictive biomarkers. The study protocol was
approved by the ethics board at each research center.
Assay to Detect Mutant KRAS
Formalin-fixed, paraffin-embedded tumor sections were deparaffinized
and air dried, and DNA was isolated using proteinase K and a DNeasy mini-
spin column (Qiagen, Valencia, CA). Mutant KRAS was detected using a
validated KRAS mutation kit (DxS Ltd, Manchester, United Kingdom) that
identifies seven somatic mutations located in codons 12 and 13 (Gly12Asp,
Gly12Ala, Gly12Val, Gly12Ser, Gly12Arg, Gly12Cys, and Gly13Asp) using
allele-specific real-time polymerase chain reaction.
23-25
A central laboratory
(HistoGeneX, Antwerp, Belgium) validated the assay for analytic and diagnos-
tic performance, established acceptance criteria, included appropriate quality
controls for each assay, and performed the KRAS analysis in a blinded fashion.
Statistical Analysis
The primary objective of the biomarker analyses was to examine whether
the relative effect of panitumumab compared with BSC on PFS differed in
patients with tumors bearing mutant versus WT KRAS. Additional objectives
included examining whether panitumumab improved PFS, OS, and response
rate in the WT KRAS group compared with the BSC group. Safety was assessed
in both KRAS groups. Analyses were limited to patients with known KRAS
status and were categorized by randomized treatment for efficacy and safety.
Adverse events were graded per the National Cancer Institute Common Tox-
icity Criteria version 2.0 with the exception of selected skin toxicities, which
were graded using version 3.0. Statistical analyses were performed at Amgen
Inc. All analyses were prespecified in a statistical analysis plan before KRAS
mutation assessment.
A quantitative-interaction test
26
at a two-sided 5% level was used to
compare the PFS log-hazard ratio (HR; panitumumab relative to BSC) from a
Cox model with covariates for the randomization factors between the WT and
mutant KRAS groups. Based on an assessable sample size of 380 patients and
assuming 60% WT prevalence, power was estimated at more than 99% if the
HR was 1.0 in the mutant KRAS group and at 87% if the HR was 0.80 in the
mutant KRAS group, assuming an overall HR of 0.54 among all patients.
Kaplan-Meier methods were used to estimate PFS and OS. Conditional on a
significant interaction test, sequential testing at a 5% level of PFS, followed by
OS and overall response rate, were planned within the WT KRAS group
between panitumumab versus BSC. A log-rank test was used for PFS, Wil-
coxon for OS, and a generalized Cochran-Mantel-Haenszel test for response
rate, each stratified by the randomization factors.
Maximum change in tumor burden per blinded central radiology review
was summarized by treatment in each KRAS group. Propensity-score sensitiv-
ity analyses were performed to assess bias due to exclusion of patients with
unknown KRAS status.
RESULTS
Patients
Of the 463 patients originally enrolled,
3
427 (92%) were included
in the KRAS analyses (208 and 219 in the panitumumab and BSC
arms, respectively; Fig 1). KRAS status could not be determined in 18
patients because of unavailable samples and in an additional 18 pa-
tients whose samples had insufficient or poor-quality DNA. KRAS
mutations were identified in 184 (43%) of 427 patients (84 [40%] and
100 [46%] in the panitumumab and BSC arms, respectively). In the
BSC arm, 76% of patients with WT KRAS and 77% of patients with
mutant KRAS received panitumumab in a cross-over protocol, after a
median PFS time in the original study (investigator assessment) of 7.1
weeks (95% CI, 7.0 to 7.6) and 6.3 weeks (95% CI, 5.1 to 7.1) for
patients in the WT and mutant KRAS groups, respectively.
Baseline patient characteristics were balanced between the WT
and mutant KRAS groups for both panitumumab and BSC (Table 1).
The distribution of specific KRAS mutations was similar between
treatment arms (Table 2).
Efficacy
Primary end point: PFS. Similar to previously described results
in the intent-to-treat population,
3
a statistically significant improve-
ment in PFS was observed in the KRAS assessable group between
panitumumab and BSC (HR, 0.59; 95% CI, 0.48 to 0.72). Median PFS
time was 8.0 weeks for panitumumab and 7.3 weeks for BSC. The
relative effect of panitumumab versus BSC on PFS was significantly
greater among patients with WT KRAS (HR, 0.45; 95% CI, 0.34 to
0.59; median PFS of 12.3 weeks for panitumumab v7.3 weeks for BSC)
compared with patients with mutant KRAS, in whom no panitu-
mumab benefit was observed (HR, 0.99; 95% CI, 0.73 to 1.36; median
PFS of 7.4 weeks for panitumumab v 7.3 weeks for BSC; Fig 2). The
quantitative-interaction test comparing the magnitude of the relative
treatment effect on PFS between WT and mutant KRAS groups was
statistically significant (P .0001). Consistent results were obtained
with propensity-score adjusted HRs. PFS was significantly greater for
panitumumab versus BSC in the WT KRAS group (stratified log-rank
test P .0001; Fig 2). In all sensitivity analyses performed in the WT
KRAS subset, PFS favored the panitumumab arm. In particular, to
compensate for potential tumor-ascertainment bias in favor of the
BSC arm, an interval-censored sensitivity analysis was performed
KRAS Is a Selection Marker for Panitumumab Benefit in mCRC Patients
www.jco.org © 2008 by American Society of Clinical Oncology 1627
Information downloaded from jco.ascopubs.org and provided by at Malliani on December 28, 2014 from 159.149.103.9
Copyright © 2008 American Society of Clinical Oncology. All rights reserved.

whereby radiologic event times were moved to the closest assessment
time prespecified in the protocol. These analyses showed HR 0.44
(95% CI, 0.30 to 0.63) and median PFS times of 16 and 8 weeks for
panitumumab and BSC, respectively. Across all subsets examined, the
treatment effect of panitumumab on PFS in the WT KRAS group was
consistent with the primary analysis (Fig 3). Of 168 BSC patients
receiving panitumumab after progression, PFS was significantly
longer among patients with WT versus patients with mutant KRAS
(HR, 0.32; 95% CI, 0.22 to 0.45; median PFS time of 16.4 weeks for
WT and 7.9 weeks for mutant; online-only Fig A1A).
Response rate. Best overall response data were unassessable or
missing for 35 of 231 patients receiving panitumumab and for 53 of
232 BSC patients (this included 16 of 124 patients receiving panitu-
mumab with WT KRAS, 16 of 119 BSC patients with WT KRAS,15of
84 patients receiving panitumumab in the mutant KRAS group, and
32 of 100 BSC patients in the mutant KRAS group). In the KRAS
assessable group, response rate for panitumumab was 10%, stable
disease was 25%, and disease progression was 50%. For KRAS assess-
able patients in the BSC arm, 0% had a response, 10% had stable
disease, and 68% had disease progression. No responders were iden-
tified in the panitumumab mutant KRAS group (100% positive pre-
dictive value for nonresponse in the mutant group). In contrast, in the
panitumumab WT KRAS group 21 of 124 patients had a partial
response (17%; 95% CI, 11% to 25%; Fig 4). Median time to response
was 7.9 weeks (range, 7.0 to 15.6 weeks), and median duration of
response was 19.7 weeks (range, 7.9 to 88.7 weeks).
In the WT KRAS group, 42 patients receiving panitumumab
(34%) and 14 BSC patients (12%) had stable disease (Fig 4). In the
mutant KRAS group, stable disease was observed in 10 (12%) and
eight patients (8%) in the panitumumab and BSC arms, respectively.
Consistent results with PFS and response were observed when exam-
ining the magnitude of effect on target lesions for individual patients.
For the WT KRAS group, 61% of patients receiving panitumumab
with available target lesion measurements (62 of 101 in the WT group)
had a target lesion decrease, including the majority of patients with
stable disease (Fig 4). In contrast, in the mutant KRAS group, only 5%
of patients receiving panitumumab (three of 62) had minor tumor
reductions. For the BSC patients in both KRAS groups, 3% of patients
(six of 178) had some degree of tumor reduction.
Of 168 BSC patients in the KRAS assessable group that crossed
over to receive panitumumab on progression, 20 (12%) experienced a
response (including one patient with a complete response), and 55
(33%) had stable disease. All responders had WT KRAS, for a response
rate of 20 of 91 (22%; 95% CI, 14% to 32%).
OS. At the time of these analyses, a total of 391 KRAS assessable
patients (92%) had died (186 [89%] patients receiving panitumumab
and 205 [94%] BSC patients). Median follow-up time was 14.1
months for the remaining 36 patients. No statistically significant OS
difference was observed between treatment arms among all patients
(HR, 0.97; 95% CI, 0.79 to 1.18), or in either of the KRAS groups; the
HR for OS was 1.02 (95% CI, 0.75 to 1.39) and 0.99 (95% CI, 0.75 to
1.29) for the mutant and WT KRAS groups, respectively. OS was
longer overall in the WT group than in the mutant group adjusting
for stratification factors and randomized treatment (HR, 0.67; 95%
CI, 0.55 to 0.82; both arms combined; Fig 5). Multivariate analysis
showed that WT KRAS status was a predictor for OS in both the
Randomly assigned
(n = 463)
Screened for eligibility
(N = 1,040)
Excluded (did not meet
inclusion criteria, n = 577)
Randomly assigned to
BSC alone
(n = 232)
Randomly assigned to
panitumumab + BSC
(n = 231)
Excluded from
KRAS analyses
(missing or unassessable
samples, n = 36)
Identified as
mutant KRAS
(n = 84)
Identified as
WT KRAS
(n = 124)
Received panitumumab
under cross-over
protocol
(n = 77)
Received panitumumab
under cross-over
protocol
(n = 90)
Identified as
mutant KRAS
(n = 100)
Identified as
WT KRAS
(n = 119)
Fig 1. CONSORT diagram. BSC, best
supportive care; WT, wild-type.
Amado et al
1628 © 2008 by American Society of Clinical Oncology
J
OURNAL OF CLINICAL ONCOLOGY
Information downloaded from jco.ascopubs.org and provided by at Malliani on December 28, 2014 from 159.149.103.9
Copyright © 2008 American Society of Clinical Oncology. All rights reserved.

panitumumab (HR, 0.64; P .004) and BSC (HR, 0.68; P .007)
arms. Similar results for OS were observed among the 168 BSC pa-
tients receiving panitumumab after progression (HR, 0.65; 95% CI,
0.47 to 0.90; median OS time of 6.8 months for WT v 4.5 months
for mutant; online-only Fig A1B). For the 51 BSC patients who did
not cross-over to panitumumab, no difference in OS was observed
between WT and mutant KRAS groups (median OS time of 1.9 and
2 months, respectively).
Exposure and Safety
The mean number of panitumumab infusions was 10.0 (median,
8.0) and 4.9 (median, 4.0) in WT and mutant KRAS groups, respec-
tively. In the mutant KRAS group, 100% of patients receiving panitu-
mumab and 84% of BSC patients had an adverse event. In the WT
KRAS group, these numbers were 100% and 90%, respectively. By
maximum grade and by KRAS group, a higher incidence of grade 3 or
4 adverse events (44% v 28%) and treatment-related grade 3 adverse
events (25% v 12%) was observed in the panitumumab WT versus
mutant KRAS groups, respectively. In the KRAS assessable popula-
tion, 37% of patients had a grade 3 or 4 event, and 20% of patients had
a treatment-related grade 3 or 4 adverse events. The incidence of
adverse events leading to withdrawal in the panitumumab arm was
7% and 5% for the WT and mutant KRAS groups, respectively; 2% of
WT KRAS patients and 1% of mutant KRAS patients withdrew for
panitumumab-related events.
Grade 3 integument-related events occurred in 20% of all KRAS
assessable patients (in 25% of WT KRAS patients and in 13% of
mutant KRAS patients). In the mutant KRAS group, 1% of patients
Table 1. Patient Demographics and Baseline Characteristics by KRAS Status
Characteristic
Mutant Wild-Type
Panitumumab BSC Panitumumab BSC
No. % No. % No. % No. %
No. of patients 84 100 124 119
Sex
Male 47 56 64 64 83 67 76 64
Race/ethnicity
White 84 100 97 97 122 98 118 99
Baseline age, years
Median 62.0 62.0 62.5 63.0
Minimum 27 27 29 32
Maximum 79 83 82 81
Primary diagnosis
Colon cancer 53 63 65 65 86 69 82 69
Rectal cancer 31 37 35 35 38 31 37 31
ECOG performance status
0 43 51373753434034
1 28 33474756456252
2
*
13 15 16 16 15 12 17 14
Cells with EGFR membrane staining
1% to 10% 20 24 23 23 31 25 29 24
10% to 100% 63 75 77 77 93 75 89 75
Highest membrane staining intensity
3 (strong) 17 20 17 17 25 20 22 18
2 (moderate) 42 50 51 51 69 56 58 49
1 (weak) 24 29 32 32 30 24 39 33
0 1 1000000
Prior adjuvant chemotherapy
Yes 27 32 40 40 50 40 32 27
Prior lines of chemotherapy
2 54 64747479646353
3 23 27242441334941
Abbreviations: BSC, best supportive care; ECOG, Eastern Cooperative Oncology Group; EGFR, epidermal growth factor receptor.
*
Of patients treated with BSC, one patient with wild-type KRAS status and one patient with mutant KRAS status had an ECOG performance status score of 3.
Table 2. Distribution of KRAS Mutations By Treatment Arm
KRAS
Mutation
Total
(N 184)
Panitumumab
(n 84)
BSC
(n 100)
No. % No. % No. %
12Ala 15 8.2 8 9.5 7 7.0
12Asp 70
*
38.0 34 40.5 36 36.0
12Arg 3
*
1.6 0 0.0 3 3.0
12Val 40 21.7 15 17.9 25 25.0
12Cys 14 7.6 7 8.3 7 7.0
12Ser 14 7.6 5 6.0 9 9.0
13Asp 29 15.8 15 17.9 14 14.0
Abbreviations: BSC, best supportive care; Ala, alanine; Asp, aspartic acid;
Arg, arginine; Val, valine; Cys, cysteine; Ser, serine.
*
Two mutations were detected in one specimen.
KRAS Is a Selection Marker for Panitumumab Benefit in mCRC Patients
www.jco.org © 2008 by American Society of Clinical Oncology 1629
Information downloaded from jco.ascopubs.org and provided by at Malliani on December 28, 2014 from 159.149.103.9
Copyright © 2008 American Society of Clinical Oncology. All rights reserved.

had a grade 4 integument-related event; there were no grade 4 events
in the WT group. The time to any integument-related event or to an
event grade 2 or higher was similar in both KRAS groups, suggesting
that incidence differences for integument toxicity were due to differ-
ential exposure. Consistent with previous reports,
2,3
patients with the
worst grade skin toxicity in the WT KRAS group appeared to experi-
ence better PFS and OS (data not shown). In the panitumumab arm, a
higher incidence of diarrhea of any grade was observed (WT KRAS
24%; mutant KRAS 19%) but grade 3 diarrhea was comparable be-
tween groups (WT KRAS 2%; mutant KRAS 1%). The incidence of
hypomagnesemia reported as an adverse event of any grade was 3%
and 0% for WT and mutant KRAS groups, respectively. One grade 2
infusion reaction was reported as an adverse event in a patient with
mutant KRAS.
DISCUSSION
These results show that KRAS mutations predict for lack of clinical
benefit to panitumumab therapy. The presence of a control arm made
it possible to study the relative effect of panitumumab monotherapy
by KRAS mutational status independent of the potential prognostic
influence of KRAS mutations on outcomes, enabling us to conclude
that the clinical benefit observed in the KRAS unselected population
was entirely derived from the KRAS WT population. Given the cross-
over design, conclusions are limited to the effect of KRAS muta-
tional status on PFS and tumor response end points and not to OS.
Indeed, the majority of BSC patients received panitumumab on
disease progression early in the trial in both KRAS groups (median
A
0
Treatment group Events N % Median (weeks)
Panit. + BSC 76 84 90 7.4
BSC alone 95 100 95 7.3
HR = 0.99
(95% CI: 0.73 to 1.36)
Proportion Event Free (%)
Time (weeks)
70
90
100
80
60
50
40
30
20
10
2 4 6 8 10121416182022242628303234363840424446485052
0
Treatment group Events N % Median (weeks)
Panit. + BSC 115 124 93 12.3
BSC alone 114 119 96 7.3
HR = 0.45
(95% CI: 0.34 to 0.59)
Stratified log-rank P < .0001
Proportion Event Free (%)
Time (weeks)
70
90
100
80
60
50
40
30
20
10
2 4 6 8 10121416182022242628303234363840424446485052
B
No. of patients at risk
Panit + BSC 84 78 76 72 26 10 8 6 5 5 5 5 4 4 4 4 2 2 2 2 2 2 2 1 1 1
BSC alone 100 91 77 61 37 22 19 10 9 8 6 5 5 4 4 4 4 4 4 3 3 3 2 2 2 2
No. of patients at risk
Panit + BSC 124 119 112 106 80 69 63 58 50 45 44 44 33 25 21 20 17 13 13 13 10 7 7 6 5 5
BSC alone 119 109 91 81 38 20 15 15 14 11 10 9 9 6 6 6 6 5 4 3 3 2 2 2 2 1
Fig 2. Progression-free survival by treat-
ment within KRAS groups. Progression-
free survival by randomized treatment in
(A) mutant and (B) wild-type KRAS groups.
Hazard ratios (HR) are shown for panitu-
mumab (panit.) versus best supportive
care (BSC) adjusted for randomization fac-
tors (Eastern Cooperative Oncology Group
score, geographic region).
Amado et al
1630
© 2008 by American Society of Clinical Oncology
J
OURNAL OF CLINICAL ONCOLOGY
Information downloaded from jco.ascopubs.org and provided by at Malliani on December 28, 2014 from 159.149.103.9
Copyright © 2008 American Society of Clinical Oncology. All rights reserved.

Citations
More filters
Journal ArticleDOI

ESMO consensus guidelines for the management of patients with metastatic colorectal cancer

TL;DR: These ESMO consensus guidelines have been developed based on the current available evidence to provide a series of evidence-based recommendations to assist in the treatment and management of patients with mCRC in this rapidly evolving treatment setting.
Journal ArticleDOI

Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis

TL;DR: This is the largest cohort to date of patients with chemotherapy-refractory metastatic colorectal cancer treated with cetuximab plus chemotherapy in the pre-KRAS selection era and confirmed that, if KRAS is not mutated, assessing BRAF, NRAS, and PIK3CA population response rates confirmed that.
References
More filters
Journal ArticleDOI

Genetic alterations during colorectal-tumor development.

TL;DR: It is found that ras-gene mutations occurred in 58 percent of adenomas larger than 1 cm and in 47 percent of carcinomas, which are consistent with a model of colorectal tumorigenesis in which the steps required for the development of cancer often involve the mutational activation of an oncogene coupled with the loss of several genes that normally suppress tumors.
Journal Article

ras Oncogenes in Human Cancer: A Review

TL;DR: It appeared that ras gene mutations can be found in a variety of tumor types, although the incidence varies greatly and some evidence that environmental agents may be involved in the induction of the mutations.
Related Papers (5)
Frequently Asked Questions (10)
Q1. What contributions have the authors mentioned in the paper "Wild-type kras is required for panitumumab efficacy in patients with metastatic colorectal cancer" ?

Panitumumab, a fully human antibody against the epidermal growth factor receptor ( EGFR ), has activity in a subset of patients with metastatic colorectal cancer ( mCRC ) this paper. 

In addition to ascertaining resistance mechanisms, other biomarkers such as EGFR gene copy number and expression levels of EGFR ligands in tumor cells may be useful to further refine the responder population. 

while the assay employed in their study is known to detect more than 90% of known activating KRAS mutations in CRC, it would have missed additional mutations in codons 12 and 61. 

Grade 3 integument-related events occurred in 20% of all KRAS assessable patients (in 25% of WT KRAS patients and in 13% of mutant KRAS patients). 

In the KRAS assessable population, 37% of patients had a grade 3 or 4 event, and 20% of patients had a treatment-related grade 3 or 4 adverse events. 

De Roock W, Piessevaux H, De Schutter J, et al: KRAS wild-type state predicts survival and is associated to early radiological response in metastatic colorectal cancer treated with cetuximab. 

All rights reserved.time to cross-over was 7.1 weeks), and, importantly, there was demonstrated benefit of panitumumab after cross-over in patients with WT KRAS tumors. 

The presence of a control arm made it possible to study the relative effect of panitumumab monotherapy by KRAS mutational status independent of the potential prognostic influence of KRAS mutations on outcomes, enabling us to conclude that the clinical benefit observed in the KRAS unselected population was entirely derived from the KRAS WT population. 

To their knowledge, these are the first results arising from a randomized, controlled trial showing that the state of a signaling molecule downstream of a target plays a crucial role in predicting clinical benefit to a targeted therapeutic. 

Median time to response was 7.9 weeks (range, 7.0 to 15.6 weeks), and median duration of response was 19.7 weeks (range, 7.9 to 88.7 weeks).