ArticlePDF Available

The impact of panitumumab treatment on survival and quality of life in patients with RAS wild-type metastatic colorectal cancer

Taylor & Francis
Cancer Management and Research
Authors:

Abstract and Figures

Panitumumab is a fully human monoclonal antibody targeting the epidermal growth factor receptor (EGFR). It is currently approved for the treatment of RAS wild-type (WT) metastatic colorectal cancer (mCRC) in combination with chemotherapy in first- and second-line and as monotherapy in chemorefractory patients. This review will provide an overview of main efficacy data on panitumumab from its early development up to latest evidences, including novel perspectives on predictive biomarkers of anti-EGFRs efficacy and mechanisms of secondary resistance. Quality of life (QoL) related issues and panitumumab safety profile will be addressed as well.
This content is subject to copyright. Terms and conditions apply.
REVIEW
The impact of panitumumab treatment on survival
and quality of life in patients with RAS wild-type
metastatic colorectal cancer
This article was published in the following Dove Press journal:
Cancer Management and Research
Francesca Battaglin
1
Alberto Puccini
2
Selma Ahcene Djaballah
3
Heinz-Josef Lenz
1
1
Division of Medical Oncology, Norris
Comprehensive Cancer Center, Keck
School of Medicine, University of
Southern California, Los Angeles, CA
90033, USA;
2
Medical Oncology Unit 1,
IRCCS Ospedale Policlinico San Martino,
Genova, Italy;
3
Medical Oncology Unit 1,
Clinical and Experimental Oncology
Department, Veneto Institute of
Oncology IOV - IRCCS, Padua 35128,
Italy
Abstract: Panitumumab is a fully human monoclonal antibody targeting the epidermal
growth factor receptor (EGFR). It is currently approved for the treatment of RAS wild-type
(WT) metastatic colorectal cancer (mCRC) in combination with chemotherapy in rst- and
second-line and as monotherapy in chemorefractory patients. This review will provide an
overview of main efcacy data on panitumumab from its early development up to latest
evidences, including novel perspectives on predictive biomarkers of anti-EGFRs efcacy and
mechanisms of secondary resistance. Quality of life (QoL) related issues and panitumumab
safety prole will be addressed as well.
Keywords: panitumumab, colorectal cancer, EGFR, RAS, biomarker, quality of life
Introduction
Colorectal cancer (CRC) is the third most frequently diagnosed malignancy both in
men and in women and represents one of the leading causes of cancer-related
mortality worldwide.
1
In recent years, an extended molecular characterization of
CRC has led to a deeper understanding of the mechanisms of development and
heterogeneity of this disease. Novel targeted agents including vascular endothelial
growth factors (VEGF)-, epidermal growth factor receptor (EGFR)- and more
recently immune checkpoints-inhibitors have become available for the treatment
of mCRC, adding to standard chemotherapy with 5-uorouracil, oxaliplatin and
irinotecan.
2
The improvement in medical treatments, together with enhanced locor-
egional and surgical approaches, has translated into a longer median overall survi-
val (OS) of patients with mCRC which has surpassed 30 months in modern day
practice.
3
The EGFR signaling pathway plays a critical role in CRC development and
EGFR inhibitors are well established therapeutic agents in mCRC treatment.
Panitumumab is a fully human monoclonal antibody (mAb) which targets with
high afnity the extracellular domain of EGFR, competitively inhibiting the binding
of other ligands and thus preventing the activation of the EGFR downstream
signaling cascade (Figure 1).
4
In malignant cells the activation of EGFR promotes
cell proliferation through the KRAS/RAF/MAPK and the PI3K/AKT/mTOR axes.
5
EGFR blockade by panitumumab results in inhibition of cell growth, induction of
apoptosis, decreased of proinammatory cytokines and VEGF production, and
EGFR downregulation through receptor internalization.
6,7
Over time, the clinical
Correspondence: Heinz-Josef Lenz
Division of Medical Oncology, Norris
Comprehensive Cancer Center, Keck
School of Medicine, University of
Southern California, 1441 Eastlake
Avenue, Suite 3456, Los Angeles, CA
90033, USA
Tel +1 323 865 3967
Email lenz@med.usc.edu
Cancer Management and Research Dovepress
open access to scientic and medical research
Open Access Full Text Article
submit your manuscript | www.dovepress.com Cancer Management and Research 2019:11 59115924 5911
DovePress © 2019 Battaglin et al. This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.
php and incorporate the Creative Commons Attribution Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the
work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For
permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).
http://doi.org/10.2147/CMAR.S186042
efcacy of panitumumab in mCRC has been proven by
several randomized trials across different treatment lines,
however since early studies it was clear that not all
patients benet from this treatment. Hence, the identica-
tion of predictive biomarkers has been paramount in pani-
tumumab studies, paving the way to the discovery of rat
sarcoma (RAS) mutations as negative predictive biomar-
kers for anti-EGFRs activity.
8
In this review, we will provide an overview of panitu-
mumab activity across different treatment scenarios and
treatment lines in mCRC. We will also address the impact
of panitumumab treatment on patientsquality of life
(QoL) and discuss novel perspectives on patient selection
and primary and secondary resistance mechanisms to anti-
EGFR agents.
Regulatory approval and molecular
patient selection
Panitumumab is currently approved by the Food and Drug
Administration (FDA) and European Medicines Agency
(EMA) for the treatment of RAS wild-type (WT) mCRC in
combination with FOLFOX (5-uorouracil, leucovorin
and oxaliplatin) or FOLFIRI (5-uorouracil, leucovorin
and irinotecan) in the rst-line setting; in combination
with FOLFIRI in the second-line setting; and as mono-
therapy following disease progression after prior che-
motherapy treatment (uoropyrimidine-, oxaliplatin- and
irinotecan-containing regimens).
4
Regulatory Agencies have also provided recommenda-
tions on validated laboratory techniques and accreditation
criteria for RAS mutation testing, which should be performed
only in highly qualied and certied laboratories. In 2017,
the American Society of Clinical Oncology (ASCO) in col-
laboration with the Association for Molecular Pathology, the
College of American Pathologists, and the American Society
for Clinical Pathology, published a set of dedicated guide-
lines on the evaluation of molecular biomarkers in CRC.
9
According to the current standard of practice every patient
being considered for anti-EGFR treatment must receive RAS
mutational testing and the analysis should include KRAS and
NRAS codons 12, 13 of exon 2; 59, 61 of exon 3; and 117 and
146 of exon 4.
8
More recently, several other tumor molecular features
and mutations in genes involved in EGFR-related path-
ways have been shown to play a role in anti-EGFRs
resistance mechanisms. The V-Raf murine sarcoma viral
oncogene homolog B1 (BRAF) V600E mutation is one of
these, and growing evidence supports the use of BRAF as a
negative predictive biomarker in clinical practice. An
overview of novel biomarkers of primary and acquired
resistance mechanisms is provided in the next sections.
Clinical efcacy
Panitumumab monotherapy
The open label phase III 408 trial was the rst study to
demonstrate a progression-free survival (PFS) benet,
although small, with single agent panitumumab compared
to best supportive care (BSC) in unselected pre-treated
mCRC (8 versus 7.3 weeks, hazard ratio (HR) 0.54; 95%
condence interval (CI), 0.440.66; P<0.0001).
10
Later on,
a retrospective biomarker analysis from this study shed
light on the predictive role of KRAS exon 2 mutation on
panitumumab efcacy, demonstrating a clear improvement
in PFS for patients with WT tumors (12.3 versus 7.3 weeks,
HR 0.45; 95% CI, 0.340.59; P<0.0001), while no benet
was observed in patients with mutated tumors (PFS 7.4
versus 7.3 weeks for panitumumab versus BSC, HR 0.99;
95% CI, 0.731.36).
11
These ndings opened a new era for
biomarker discovery and molecular patient selection, lead-
ing the restriction of the use of anti-EGFR agents to KRAS
exon 2 (codon 12 and 13) WT tumors in 2008.
PI3K
PTEN
AKT
mTOR
RAS
RAF
MEK
ERK
EGFR
Panitumumab
Cell survival, proliferation, angiogenesis,
metastatic spread
Figure 1 Panitumumab, a fully humanized monoclonal IgG2 antibody, inhibits the
EGFR pathway.
Abbreviations: AKT, AKT8 virus oncogene cellular homolog; EGFR, epidermal
growth factor receptor; ERK, extracellular signalregulated kinase; MEK, mitogen-
activated protein kinase kinase; mTOR, mammalian target of rapamycin; PI3K,
phosphatidyilinositol 3-kinase; PTEN, phosphatase and tensin homolog; RAF, v-Raf
murine sarcoma viral oncogene homolog; RAS, rat sarcoma viral oncogene
homolog.
Battaglin et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
Cancer Management and Research 2019:11
5912
The activity of panitumumab monotherapy has been
compared to that of cetuximab, the rst approved anti-
EGFR agent, in an open-label randomized phase III trial
in patients with chemotherapy-refractory KRAS exon
2 WT mCRC.
12
Panitumumab was non-inferior to cetux-
imab in terms of OS, PFS, and objective response rate
(ORR), with reported OS of 10.4 months and 10 months,
respectively (HR 0.97, 95% CI 0.841.11, P=0.0007).
Combination therapy
Shortly after the 408 study, several trials evaluated the
efcacy of panitumumab in association with chemotherapy
doublets, showing signicant benet from the addition of
panitumumab compared to chemotherapy alone in KRAS
exon 2 WT patients, both in rst- and in second-line
settings (efcacy data of main trials are summarized in
Table 1).
The phase III randomized 181 trial compared second-
line treatment with panitumumab-FOLFIRI to FOLFIRI
alone.
13
The study was later amended to prospectively
evaluate KRAS exon 2 status as a predictor of panitumu-
mab efcacy. In the KRAS WT population, a signicant
improvement in PFS was observed when panitumumab
was added to chemotherapy (median PFS 5.9 versus
3.9 months, HR 0.73; 95% CI, 0.590.90; P=0.004);
response rate was also improved to 35% versus 10% by
the addition of panitumumab. A non-signicant trend
toward increased OS was observed for the panitumumab
arm: median OS 14.5 versus 12.5 months, HR 0.85, 95%
CI, 0.701.04; P=0.12. Conversely, no benet was
observed in patients whose tumors harbored a KRAS
mutation.
14
In the rst-line setting, the phase III randomized
PRIME study demonstrated the benet of combining pani-
tumumab with FOLFOX-4 compared to FOLFOX-4 alone
in KRAS exon 2 WT mCRC.
15,16
Further efcacy analysis
of this study, based on a more extensive patient molecular
selection after the emerging evidence on the role of rare
RAS activating mutations (KRAS exon 3 and 4, NRAS exon
2, 3 and 4) and BRAF mutations in anti-EGFRs
resistance,
17
proved for the rst time a striking advantage
from panitumumab treatment in the extended RAS WT
population and lack of efcacy in RAS-mutated tumors.
Notably, in 446 RAS/BRAF WT patients, panitumumab
was shown to confer a greater magnitude of OS benet
compared to KRAS exon 2 WT, with an impressive
7.4 months improvement over chemotherapy alone (28.3
versus 20.9 months, HR 0.74; 95% CI, 0.570.96;
P=0.02).
18
The presence of a BRAF mutation was also
conrmed as an independent negative prognostic factor
both for PFS and OS, irrespective of treatment.
Similar results were obtained from updated molecular
analyses of randomized rst-,
19
second-
14
and third-line
20
trials. A meta-analysis also conrmed the presence of
extended RAS mutations as negative predictive biomarkers
for anti-EGFRs activity in mCRC, with no difference
between KRAS exon 2 mutations and other KRAS or
NRAS mutations.
21
These data led to the FDA restriction
for the use of panitumumab to extended KRAS and NRAS
WT mCRC. More recently, evidence on the role of
BRAFV600E mutation as a biomarker of resistance to
anti-EGFR agents has been conrmed by large meta-ana-
lyses showing a lack of treatment benet from anti-EGFR
mAbs, both in terms of PFS and OS, for BRAF-mutated
mCRC.
2224
Both anti-EGFRs and anti-VEGF agents are approved
for the rst-line treatment of RAS WT mCRC in associa-
tion with chemotherapy and have recently been compared
in different head-to-head randomized trials. The phase II
PEAK study investigated the addition of panitumumab
versus bevacizumab to FOLFOX chemotherapy in the
rst-line setting.
25
Although not designed to prove the
superiority of one treatment over the other, this study
showed a signicant improvement in PFS (13.1 versus
10.1 months, HR 0.61; 95% CI, 0.420.88; P=0.0075),
and a trend towards OS (41.3 versus 28.9 months, HR
0.70; 95% CI, 0.481.04; P=0.08) from panitumumab
versus bevacizumab in the extended RAS/BRAF WT popu-
lation, suggesting a survival benet from rst-line use of
panitumumab in association to chemotherapy in these
patients.
26
A recent exploratory pooled analysis evaluating
the effect of sequence of biological therapies on OS in
patients with RAS or RAS/BRAF WT mCRC treated with
panitumumab across the PRIME, PEAK and 181 trials,
conrmed a trend towards improved OS for rst-line pani-
tumumab plus chemotherapy followed by second-line
VEGF inhibitors, compared with rst-line bevacizumab
followed by second-line anti-EGFRs.
27
Large prospective
randomized trials are warranted to further evaluate the
optimal rst-/second-line targeted treatment sequence in
RAS WT mCRC. Of interest, the ongoing CR-
SEQUENCE trial from the Spanish Cooperative Group
for the Treatment of Digestive Tumors (TTD), evaluating
the efcacy of FOLFOX plus panitumumab followed by
FOLFIRI plus bevacizumab (Sequence 1) versus FOLFOX
plus bevacizumab followed by FOLFIRI plus
Dovepress Battaglin et al
Cancer Management and Research 2019:11 submit your manuscript | www.dovepress.com
DovePress 5913
Table 1 Efcacy results from main panitumumab trials
Trial (phase)
Ref
Treatment
Arms (n.)
Treatment
Line
Primary
Endpoint
ORR (%) PFS OS
KRAS ex
2WT
RAS WT KRAS ex
2WT
RAS WT KRAS ex
2WT
RAS WT
408 (III)
NCT00113763
10,11,20
Panitumumab
(n.231)
BSC (n.232)
3rd/+ PFS 17%
0%
17%
0%
2.87 m
(n.124)
1.7 m
(n.119)
[HR 0.45;
P<0.0001]
HR 0.39;
95% CI,
0.270.56;
P<0.001
8.1 m
(n.124)
7.6 m
(n.119)
[HR 0.99;
95% CI,
0.751.29]
Not
reported
0007 (III)
NCT01412957
80
Panitumumab
(n.189)
BSC (n.188)
3rd/+ OS 27%
1.6%
[P<0.0001]
31%
2.3%
[P<0.0001]
3.6 m
(n.189)
1.7 m
(n.188)
[HR 0.51;
P<0.0001]
5.2 m
(n.142)
1.7 m
(n.128)
[HR 0.46;
P<0.0001]
10 m
(n.189)
7.4 m
(n.188)
[HR 0.73;
P=0.0096]
10 m
(n.142)
6.9 m
(n.128)
[HR 0.70;
P=0.0135]
ASPECCT (III)
NCT01001377
12
Panitumumab
(n.499)
Cetuximab
(n.500)
3rd/+ OS 22%
20%
NE 4.1 m
(n.499)
4.4 m
(n.500)
[HR 1.00;
95% CI,
0.88
1.14]
NE 10.4 m
(n.499)
10 m
(n.500)
[HR 0.97;
Z-score
3.19;
P=0.0007]
NE
PRIME (III)
NCT00364013
15,16
FOLFOX +
Panitumumab
(n.593)
FOLFOX
(n.590)
1st PFS 57%
48%
[P=0.02]
Not
reported
10.0 m
(n.325)
8.6 m
(n.331)
[HR 0.80;
P=0.01]
10.1 m
(n.259)
7.9 m
(n.253)
[HR 0.72;
P=0.004]
23.8 m
(n.325)
19.4 m
(n.331)
[HR 0.83;
P=0.03]
25.8 m
(n.259)
20.2 m
(n.253)
[HR 0.77;
P=0.009]
314 (II)
NCT00508404
19,47
FOLFIRI +
Panitumumab
(n.154)
1st ORR 56% 59% 8.9 m
(n.86)
11.2 m
(n.68)
Not
Reported
Not
reported
PEAK (II)
NCT00819780
25,26
FOLFOX +
Panitumumab
(n.142)
FOLFOX +
Bevacizumab
(n.143)
1st PFS 57.8%
53.5%
65%
60%
10.9 m
(n.142)
10.1 m
(n.143)
[HR 0.87;
P=0.35]
12.8 m
(n.88)
10.1 m
(n.82)
[HR 0.68;
P=0.029]
34.2 m
(n.142)
24.3 m
(n.143)
[HR 0.62;
P=0.009]
36.9 m
(n.88)
28.9 m
(n.82)
[HR 0.76;
P=0.15]
PLANET-TTD
(II)
NCT00885885
28
FOLFOX +
Panitumumab
(n.38)
FOLFIRI +
Panitumumab
(n.39)
1st ORR 74%
67%
78%
73%
13 m
(n.38)
14 m
(n.39)
[HR 0.90;
P=0.728]
13 m (n.27)
15 m (n.26)
[HR 0.70;
P=0.307]
37 m (n.38)
41 m (n.39)
[HR 1.0;
P=0.966]
39 m
(n.27)
49 m
(n.26)
[HR 0.9;
P=0.824]
(Continued)
Battaglin et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
Cancer Management and Research 2019:11
5914
panitumumab (Sequence 2) in untreated patients with
unresectable RAS WT left-sided mCRC (NCT03635021).
Of note, panitumumab treatment was consistently asso-
ciated with higher early tumor shrinkage (ETS) rates and
greater depth of response (DpR) in a large retrospective
analysis of patients with RAS WT mCRC from the rando-
mized rst-line PRIME, PEAK and PLANET
28
trials.
Irrespective of treatment, ETS and DpR were associated
with improved PFS, OS and resection rates in this analy-
sis, suggesting that achieving these endpoints during rst-
line treatment is linked with favorable outcomes.
29
In the third-line setting, regorafenib and triuridine/tipir-
acil are recommended after progression to standard cytotoxic
and targeted treatments. However, in RAS WT patients not
previously treated with anti-EGFR antibodies, cetuximab in
combination with irinotecan or panitumumab monotherapy
may be considered as a third-line. Of interest, in the context
of the continuum of care of mCRC patients, several studies
and case reports have reported data about different treatment
strategies in second- or third-line, including the reintroduc-
tion or re-challenge with cetuximab or panitumumab in
patients who have been previously treated with anti-EGFR
drugs as a rst-line.
30,31
Despite promising results, further
perspective trials are warranted to establish the role of this
strategy in the third-line setting in RAS WT mCRC patients
(see paragraph 6).
32
Combination with intensied
chemotherapy
Panitumumab has also been tested in combination with the
triple chemotherapy regimen FOLFOXIRI in several small
studies.
In 2013, a single arm phase II trial enrolled 37
patients with quadruple WT (KRAS, NRAS, HRAS,
BRAF) initially unresectable mCRC to receive treatment
with panitumumab in association to a modied
FOLFOXIRI regimen.
33
Median PFS was 11.3 months.
The ORR, primary endpoint of the study, was 89% with
one complete response, allowing 16 metastases resection,
13 of which (35%) R0. Another single arm phase II trial
assessing the efcacy of FOLFOXIRI plus panitumumab
in RAS WT tumors was published in 2016.
34
ORR was
59% (no complete responses) and 10 patients (66%)
underwent surgery and secondary R0 resection. Median
PFS was 13.3 months.
Table 1 (Continued).
Trial (phase)
Ref
Treatment
Arms (n.)
Treatment
Line
Primary
Endpoint
ORR (%) PFS OS
KRAS ex
2WT
RAS WT KRAS ex
2WT
RAS WT KRAS ex
2WT
RAS WT
VOLFI (II)
NCT01328171
35
mFOLFOXIRI
+
Panitumumab
(n.63)
mFOLFOXIRI
(n.33)
1st ORR - 87.3%
60.6%;
[P=0.0041]
- 9.7 m
(n.63)
10.1 m
(n.33)
[HR 0.92;
P=0.72]
Not
Reported
Not
Reported
181 (III)
NCT00339183
13,14,81
FOLFIRI +
Panitumumab
(n.591)
FOLFIRI
(n.595)
2nd PFS, OS 35%
10%
[P<0.0001]
41%
10%
5.9 m
(n.303)
3.9 m
(n.294)
[HR 0.73;
P=0.004]
6.4 m
(n.208)
4.6 m
(n.213)
[HR 0.70;
P=0.007]
14.5 m
(n.303)
12.5 m
(n.294)
[HR 0.85;
P=0.12]
16.2 m
(n.303)
13.9 m
(n.294)
[HR 0.81;
P=0.08]
SPIRITT (II)
NCT00418938
82
FOLFIRI +
Panitumumab
(n.91)
FOLFIRI +
Bevacizumab
(n.91)
2nd PFS 32%
19%
NE 7.7 m
(n.91)
9.2 m
(n.91)
[HR 1.01;
P=0.97]
NE 18 m (n.91)
21.4 m
(n.91)
[HR 1.06;
P=0.75]
NE
Abbreviations: BSC, best supportive care; CI, condence interval; ex, exon; HR, hazard ratio; m, months; n, number of patients; NE, not evaluated; PFS, progression free
survival; ORR, objective response rate; OS, overall survival; Ref, reference; WT, wild-type.
Dovepress Battaglin et al
Cancer Management and Research 2019:11 submit your manuscript | www.dovepress.com
DovePress 5915
More recently, promising results were presented from
the randomized phase II VOLFI trial, which enrolled 96
patients with unresectable RAS WT mCRC to receive either
mFOLFOXIRI plus panitumumab or FOLFOXIRI alone.
35
First-line treatment with mFOLFOXIRI plus panitumumab
resulted in signicantly higher ORR compared to che-
motherapy alone (87.3% versus 60.6%; OR: 4.47; 95%
CI, 1.6112.38; P=0.0041), and higher disease control rate
(DCR) (97% versus 79%, P=0.0071). Secondary resection
rates were 33.3% in the anti-EGFR arm (61.9% R0) versus
12.1% in the chemotherapy-only arm in the overall popula-
tion, and 75% versus 36.4% in the potentially resectable
cohort. Median PFS was not signicantly different between
treatment arms in the overall population.
To clarify whether the intensication of chemotherapy
treatment in combination with panitumumab may be bene-
cial, two trials are currently ongoing. The phase III
TRIPLETE trial is testing the efcacy of FOLFOXIRI
plus panitumumab versus mFOLFOX6 plus panitumumab
in previously untreated RAS/BRAF WT mCRC
(NCT03231722). The phase II PANIRINOX trial is asses-
sing treatment with FOLFIRINOX plus panitumumab ver-
sus mFOLFOX6 plus panitumumab (NCT02980510).
Results of these trials are warranted to further evaluate the
efcacy and safety of this intensied treatment strategy.
Maintenance treatment
Maintenance treatment with the anti-VEGF bevacizumab
in combination with a uoropyrimidine after a period of
induction therapy in patients with a good response to the
initial treatment has become a standard of care for mCRC
and is included in main international guidelines. On the
other hand, there is less evidence on maintenance strate-
gies involving anti-EGFR mAbs.
The role of continuing panitumumab as a maintenance
therapy after rst-line treatment was rstly evaluated in a
retrospective analysis of patients from the PRIME and PEAK
trials receiving maintenance therapy with panitumumab plus
5-uorouracil/leucovorin (5-FU/LV).
36
Overall, the median
duration of panitumumab maintenance was 21 weeks (inter-
quartile range: 1141). The analysis showed an OS and PFS
benet in continuing the administration of panitumumab in
addition to 5-FU/LV versus chemotherapy ± bevacizumab,
with PRIME patients having a median OS of 40.2 versus
24.1 months and PEAK patients a median OS of 39.1 versus
28.9 months, respectively.
More recently, the phase II VALENTINO study investi-
gated the efcacy of a maintenance treatment with 5FU/LV
plus panitumumab versus single-agent panitumumab follow-
ing rst-line FOLFOX plus panitumumab in patients with RAS
WT mCRC. This study showed that maintenance with pani-
tumumab alone following induction with FOLFOX plus pani-
tumumab achieves inferior PFS than the 5FU/LV plus
panitumumab combination: 10-months PFS 52.8% versus
62.8%, median PFS 10.2 versus 13 months, respectively
(P=0.011).
37
Data from the Japanese phase II SAPPHIRE trial,
where patients not progressing after 6 cycles of
FOLFOX plus panitumumab were randomized to receive
5-FU/LV and panitumumab as maintenance therapy or to
continue induction treatment, showed similar 9-months
PFS in the two arms, thus supporting the use of panitu-
mumab plus 5-FU/LV as a maintenance treatment in order
to delay disease progression while preventing the occur-
rence of oxaliplatin-induced neuropathy.
38
Tumor sidedness in panitumumab trials
Over the past few years, several studies highlighted the
prognostic value of primary tumor location (left colon
versus right colon) and data have focused on location as
a potential predictive biomarker for anti-EGFRs activity,
especially in the rst-line setting. In particular, left-sided
primary tumors have been shown to have better prognosis
and improved treatment outcomes from the use of EGFR
inhibitors in addition to combination chemotherapy.
Data from 927 patients with extended RAS WT mCRC
enrolled in three randomized trials on panitumumab
(PRIME, PEAK and 181) showed that the overall prog-
nosis was worse for right-sided tumors than for left-sided
ones, regardless of treatment. The addition of panitumu-
mab to chemotherapy led to striking PFS and OS out-
comes in left-sided tumors; conversely, patients with RAS
WT right-sided primary tumors derived no benet from
the addition of anti-EGFRs to chemotherapy. A higher
proportion of patients with right-sided tumors harbored
BRAF mutations, thus contributing to the worse prognosis
of this group, nevertheless, similar efcacy data were also
obtained in the RAS/BRAF WT population.
39
Similar
results were found consistently across several different
trials of panitumumab in second- and later-lines of
treatment,
40
and trials investigating cetuximab-based
treatments.
41
A more recent retrospective analysis of
patients with RAS WT mCRC from the PRIME and
PEAK trials further evaluated the effects of primary
tumor location on ETS, DpR, and long-term survival.
First-line panitumumab was associated with improved
Battaglin et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
Cancer Management and Research 2019:11
5916
ETS (PRIME: 62% versus 36%; PEAK: 58% versus 41%)
and DpR (PRIME: 59% versus 49%; PEAK: 70% versus
48%) in patients with left-sided mCRC, and panitumumab
treatment consistently predicted long-term survival.
Notably, in the pooled analyses of the studies, more
patients with right-sided disease achieved ETS if treated
with panitumumab than comparator (39% versus 29%),
thus ETS may identify a subgroup of patients with right-
sided disease who might respond to panitumumab.
42
Large meta-analyses of rst-line trials comparing che-
motherapy plus bevacizumab to chemotherapy plus anti-
EGFRs have shown a signicant benet in ORR, PFS and
OS in patients with left-sided primary tumors treated with
anti-EGFR mAbs compared to bevacizumab, whereas
right-sided tumors have been shown to be a negative
prognostic indicator for OS for all treatments and to ben-
et more from bevacizumab treatment.
43,44
Several hypotheses have been proposed to explain
these ndings, involving the role of different embryogenic
origin, the association of right-sided tumors with specic
molecular phenotypes (particularly, CMS1-immune and
CMS3-metabolic), different methylation signatures and
the distinct microbiota in right versus left colon, support-
ing the role of tumor sidedness as a surrogate for tumor
biology.
45
A limitation of these data is the unplanned retrospec-
tive nature of the abovementioned analyses, however, in
light of their consistency across a number of different
randomized trials, NCCN guidelines have recently incor-
porated into their recommendation to exclude anti-EGFR
antibodies in the rst-line treatment of right-sided RAS
WT mCRC.
2
It has to be noted, however, that when selecting the
optimal treatment strategy for a RAS WT mCRC patient a
comprehensive evaluation of the clinical scenario, treat-
ment goals, expected toxicities and patientscharacteris-
tics and preferences must be taken into account, leading to
a personalized approach that may favor, for instance, an
anti-VEGF therapy as rst-line for a left-sided RAS WT
mCRC, saving anti-EGFR agents for a later treatment line.
Quality of life, safety and tolerability
Anti-EGFR therapy frequently results in skin-related toxi-
cities (eg acneiform rash, xerosis, paronychia). These side
effects can negatively affect treatment compliance and
patientsquality of life (QoL)
46
and it is important to
evaluate how the impact of such adverse events weigh
against the benets of panitumumab in mCRC patients.
Therefore, maintenance of QoL is an important objective
in clinical trials and patient-reported outcomes (PROs) are
a useful way of measuring the impact of treatment
on QoL.
Study 314 was a single-arm, multicenter, phase II study
evaluating the efcacy and safety of panitumumab plus
FOLFIRI as rst-line treatment for patients with mCRC.
47
In this trial, QoL was measured using the EuroQoL 5-
domain (EQ-5D) and the EORTC QoL Questionnaires
(QLQ-C30) as an exploratory endpoint. Notably, panitu-
mumab plus FOLFIRI had minimal impact on patients
QoL, as EQ-5D and QLQ-C30 scores remained stable
throughout the study despite the high incidence of skin-
related toxicity.
48
In the PRIME trial,
15
QoL was assessed as a prespeci-
ed tertiary endpoint, using the EQ-5D health state index
(HSI) and overall health rating (OHR) measures. There
were no statistically signicant differences between the
panitumumab plus FOLFOX4 and FOLFOX4 arms in
HSI or OHR scores from baseline to progression or to
discontinuation.
49
Of interest, in this study the authors
assessed whether skin toxicities and early tumor shrinkage
(ETS) may have had impact on QoL. However, no sig-
nicant differences in QoL outcomes were observed
between patients with grade (G) 02 skin toxicity and
those with G3+ skin toxicity, as well as no difference in
QoL for those with ETS versus those without ETS.
Nonetheless, patients with tumor-related symptoms at
baseline who experienced ETS showed a statistically
meaningful improvement in QoL compared with those
who did not have ETS.
The evaluation of changes in health-related QoL
(HRQoL) using the EQ-5D was a tertiary objective also
in the second-line phase III 181 trial.
13
A total of 530
patients (263 treated with panitumumab plus FOLFIRI
and 267 with FOLFIRI) were included in the HRQoL
analysis, representing 88.8% of the overall KRAS WT
population. There were no statistically signicant or clini-
cally meaningful overall differences in the change in
HRQoL when comparing treatment arms. In addition,
regardless of the severity of skin toxicity, patients treated
with panitumumab maintained a similar HRQoL.
50
Panitumumab has also been reported to provide better
control of symptoms and maintenance of HRQoL com-
pared with BSC alone in patients with chemorefractory
KRAS WT mCRC.
51
Taken together, these data suggest that the addition of
panitumumab to chemotherapy regimens as a rst-,
Dovepress Battaglin et al
Cancer Management and Research 2019:11 submit your manuscript | www.dovepress.com
DovePress 5917
second- or later-line treatment of patients with RAS WT
mCRC provides improvements in survival outcomes with-
out compromising HRQoL.
Safety and tolerability data are available from clinical
trials evaluating panitumumab as a monotherapy or in
combination with chemotherapy in mCRC. Based on a
pooled analysis of patients enrolled in panitumumab trials
(n=2,224), the most commonly reported adverse reactions
(AE) are skin reactions occurring in approximately 94% of
patients, including rash (47%), dermatitis acneiform
(39%), pruritus (36%), erythema (33%), dry skin (21%),
and paronychia (20%). Other very commonly reported AE
occurring in 20% of patients are diarrhea (46%), nausea
(39%), vomiting (26%), constipation (23%), abdominal
pain (23%), fatigue (35%), pyrexia (21%), and decreased
appetite (30%).
52
In phase II trials, the most frequent panitumumab-
related AE involved skin (9296%), nails (2830%),
eyes (817%), hair (8%).
53
EGFR is expressed in normal
skin cells; therefore, dermatologic AE are directly linked
to EGFR blockade. Acneiform rash usually appears after
the rst treatment administration, while paronychia and
desquamation usually appear by the fourth week of
treatment.
54
In a pooled analysis of 920 patients treated
with panitumumab monotherapy included in ten phase I-
III clinical trials most patients experienced G12 skin
toxicities that rarely resulted in treatment discontinuation.
Importantly, the development of skin toxicities G2 has
been associated with improved PFS and OS;
55
therefore, it
is considered a strong predictive biomarker of clinical
benet in patients treated with EGFR inhibitors.
Since these toxicities can result in treatment disconti-
nuation and can potentially affect the patients QoL,
increase patient risk for additional infections, and lead to
suboptimal anti-EGFR schedules -all of which may affect
clinical outcomes- their management should be an impor-
tant focus when administering these agents.
56
Hence,
novel strategies to reduce the incidence and the severity
of skin toxicity have been developed based on the
STEPP
54
(Skin Toxicity Evaluation Protocol with
Panitumumab) and J-STEPP
57
randomized studies. The
randomized phase II STEPP study evaluated the impact
of a pre-emptive strategy (primary prophylaxis) including
skin moisturizers, sunscreen, topical steroids, and doxycy-
cline for the duration of anti-EGFR therapy, versus a
reactive treatment after toxicity occurrence.
54
The pre-
emptive strategy signicantly reduced the incidence of
G2 skin toxicity at 6 weeks compared to standard care
(29% versus 62%, respectively). Similarly, the Japanese
open-label, multicenter, randomized J-STEPP study
showed that the cumulative incidence of G2 skin toxi-
cities in 6 weeks was 21.3% in the pre-emptive group
compared with 62.5% in the reactive group (RR=0.34;
95% CI, 0.190.62; P<0.001).
57
Panitumumab administration should be withheld at the
rst occurrence of G3 skin toxicities. Re-introduction of
panitumumab at the original dose is recommended once
toxicity has subside, while dose reduction is recommended
upon subsequent occurrence of G3 toxicities (80% of the
original dose at the second occurrence and 60% at the third
occurrence).
4
Discontinuation of treatment is implemented
at the forth occurrence or if G3 skin toxicities do not
recover after 12 withheld doses.
When panitumumab is administered as monotherapy
severe diarrhea is uncommon, however its incidence
increases when panitumumab is associated with che-
motherapy. In fact, G34 diarrhea occurred in up to 28%
of patients in trials combining an EGFR inhibitor with
chemotherapy.
58
Another common AE that may occur during panitumu-
mab treatment is hypomagnesemia, due to the effects of
EGFR inhibition in the ascending loop of Henle and in the
distal convoluted renal tubule. Incidence of hypomagnese-
mia can be up to 2836% and was found to be associated
with treatment duration.
59
In most cases panitumumab-
induced hypomagnesemia is asymptomatic, however for
patients who experience a symptomatic G2 hypomagne-
semia, oral or intravenous replacement should be consid-
ered. Of interest, early onset of hypomagnesemia during
anti-EGFR treatment has been associated with treatment
efcacy.
60
Panitumumab is a fully human mAb, hence incidence
of infusion-related reactions is very low (13%). The use
of routine premedication before the administration of pani-
tumumab is recommended if a previous infusion reaction
has occurred.
4
Panitumumab in the elderly
population
Despite the high prevalence of CRC in the elderly popula-
tion, these patients have been underrepresented in clinical
trials and their optimal treatment is yet to be determined,
with only few data available on anti-EGFR treatment in
combination with chemotherapy. In the daily practice,
treatment of older cancer patients is challenging and a
Battaglin et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
Cancer Management and Research 2019:11
5918
careful assessment of patientsperformance status, comor-
bidities, age-related organ function, life expectancy, poten-
tial treatment-related toxicity and QoL issues should be
implemented in the decision making to select those
patients who could benet from treatment.
The use of panitumumab as monotherapy in the rst-
line setting in elderly and frail patients was investigated by
Sastre and colleagues, who treated 33 KRAS WT patients
over 70 years of age with an ECOG functional status up to
3 in a single arm phase II trial. Treatment with panitumu-
mab was demonstrated to be an active and safe option in
this group of patients. ORR was 9.1% with a 6-months
PFS rate of 53.3% and median OS of 12.3 months in the
extended RAS WT patients.
61
Encouraging data were also
reported in another study investigating panitumumab
monotherapy in molecularly selected RAS and BRAF WT
frail elderly patients deemed unt for chemotherapy.
62
However, data on the adoption of chemotherapy plus
anti-EGFRs in elderly mCRC patients are scarce. In the
subgroup analysis of RAS WT patients from the PRIME
study the combination of FOLFOX-4 and panitumumab
showed a benet over FOLFOX-4 in the subset of patients
aged more than 65 years (n=188), in terms of OS (26.6
versus 17.4 months; HR 0.78; 95% CI, 0.581.09), PFS
(9.7 versus 9.2 months; HR 0.88; 95% CI, 0.651.19) and
ORR (49% versus 42%), without raising any safety
concern.
63
Positive results in terms of tolerability and
efcacy were also recently reported in a retrospective
study of 100 patients aged over 70 years (95.4% ECOG
performance status 01) treated with doublet chemother-
apy plus panitumumab, with a median PFS of 9.4 months
(95% CI, 7.811.0) and a median OS of 23.0 months (95%
CI, 20.625.3).
64
To clarify the safety and efcacy of panitumumab in
association with chemotherapy in the elderly population a
dedicated trial, the phase II PANDA study (NCT02904031),
is currently ongoing, enrolling patients over 70 years of age
with an ECOG performance status 1 or 2 if aged 70 to
75 years and an ECOG performance status 0 or 1 if aged
>75 years. In this study elderly patients with a diagnosis of
RAS and BRAF WT mCRC are randomized to a rst-line
treatment with panitumumab in combination with FOLFOX
or 5FU/LV. Of note, secondary endpoints of the study
include the evaluation of the prognostic role of geriatric
assessment tools and toxicity risk scores to aid patient
selection in the elderly population. Safety and efcacy
results of this trial are warranted to inform targeted treat-
ment choices in elderly patients.
Novel mechanisms of resistance and
future perspectives
Several additional mechanisms of primary resistance to
anti-EGFRs have been identied in RAS WT mCRC so
far, based on preclinical data and retrospective evaluations.
However, the routine use of these biomarkers in clinical
practice is not recommended at present, and further pro-
spective validation is warranted. These include HER2
amplication, PIK3CA mutations (exon 9 and 20), MET
amplication, FGFR1 and PDGFRA mutations and loss of
PTEN function.
65
HER2 amplication, in particular, has
recently gained attention as a promising druggable target
in mCRC. Based on a strong pre-clinical rationale,
66
the
proof-of-concept phase II HERACLES trial has shown
promising activity of a combined HER2 blockade with
trastuzumab and lapatinib in treatment-refractory HER2-
positive mCRC.
67
Notably, all patients enrolled in the trial
received previous EGFR inhibitors and none of those
evaluable for response achieved an objective response to
either panitumumab or cetuximab, supporting the role of
HER2 amplication as a resistance mechanism to anti-
EGFRs. Several trials are currently investigating HER2
blockade strategies in HER2-amplied mCRC, opening
new perspectives for this subset of patients. Other novel
treatment strategies combining EGFR inhibitors with dif-
ferent targeted agents (ie panitumumab plus the mTOR
inhibitor everolimus;
68
or panitumumab plus BRAF and
MEK inhibition in BRAFV600E-mutant tumors
69
), aiming
to overcome primary resistance to anti-EGFR agents, are
also under investigation.
More recently, a panel of multiple combined genomic
alterations comprising activating mutations of the
MAPKs or PI3K/AKT axis, NTRK/ROS1/ALK/RET rear-
rangements, HER2 amplication or mutations, and MET
amplication (the PRESSING panel), has been shown to
be able to predict primary resistance to anti-EGFRs in
RAS/BRAF WT mCRCs.
70
Additionally, a right-sided pri-
mary tumor location was found to be associated with
resistance to anti-EGFRs, conrming previous literature
evidence. Overall, the combined evaluation of the
PRESSING panel and primary tumor location demon-
strated the best predictive accuracy. These results open
novel perspectives on the clinical application of a more
comprehensive molecular characterization of RAS/BRAF
WT mCRCs to further improve and rene patient selec-
tion for anti-EGFR treatment and possibly tailor persona-
lized targeted approaches.
Dovepress Battaglin et al
Cancer Management and Research 2019:11 submit your manuscript | www.dovepress.com
DovePress 5919
Clonal selection induced by treatment pressure is often
responsible for the development of secondary resistance to
EGFR inhibitors, and emerging mutations in the RAS/
RAF/MAPK pathway can be identied in tumor samples
at progression in patients previously diagnosed with a RAS
WT tumors.
71
Several trials are investigating different
approaches to multiple target inhibition based on the emer-
gence of different resistance drivers. In this setting, the use
of liquid biopsies and the analysis of circulating tumor
DNA (ctDNA) are being evaluated as a less invasive and
more comprehensive approach to pharmacogenomic pro-
ling and biomarkers monitoring in mCRC patients.
72
These techniques might play, in the near future, a pivotal
role in improving patient selection and targeted treatment
strategies by implementing early detection of the emer-
gence of treatment resistance and allowing a dynamic
molecular proling.
73
Indeed, repeated ctDNA analyses
have been able to capture the emergence of resistant clones
during treatment with panitumumab or cetuximab in RAS
WT patients, showing that this phenomenon is closely
related to treatment exposure, with a dynamic increase
during anti-EGFRs administration followed by a rapid
decline at withdrawal.
74,75
In a recent biomarker analysis
from a second-line phase II trial of panitumumab in asso-
ciation with irinotecan in KRAS WT mCRC, plasma test-
ing of cell-free DNA revealed a mutant RAS emergence
rate of 36.7% (n=39), and rst detected emergence of RAS
mutations preceded progression by a median of 3.6 months
(range, 0.37.5).
76
However, patients who had emergent
RAS mutations at progression had similar median PFS to
those who remained WT and a mutant RAS allele fre-
quency threshold that could predict near-term outcomes
was not identied, thus calling for further evaluation of the
clinical value of this approach. Interestingly, recently pub-
lished results from retrospective analyses evaluating emer-
gent mutations in circulating cell-free DNA in patients
treated with panitumumab in the ASPECCT study showed
that patients with a higher RAS mutant allele frequency at
baseline had worse clinical outcomes than those with a
lower frequency (P<0.001). However, extended RAS muta-
tion, by itself, did not preclude clinical responses to pani-
tumumab in this setting and emergent ctDNA RAS
mutations were not associated with less favorable patient
outcomes in panitumumab-treated patients.
77,78
Further
research is needed to identify a clinically relevant thresh-
old for baseline and emergent ctDNA RAS mutations.
Of note, focusing on the issue of analytical sensitivity in
evaluating predictive biomarkers to anti-EGFR treatments,
the phase II ULTRA trial investigated a high-sensitivity
tumor tissue genotyping technique of KRAS, NRAS, BRAF
and PIK3CA to ultra-select irinotecan-resistant mCRC
patients for panitumumab plus FOLFIRI treatment.
Results from this study identify the optimal RAS/BRAF
mutational threshold for outcome prediction to be 5%,
suggesting that the biological and clinical implications of
mutation frequencies below this cut-off still warrant further
investigations.
79
Finally, re-challenge strategies after treatment breaks in
patients with RAS WT tumors that demonstrated a pre-
vious response to anti-EGFR agents are currently under
study. The phase II CHRONOS study (NCT03227926)
aims to investigate a re-challenge strategy with panitumu-
mab as third-line treatment after a rst-line treatment with
anti-EGFRs in RAS/BRAF WT mCRC, with a molecular
follow-up based on ctDNA. In this study liquid biopsies
for ctDNA testing are prospectively collected during the
rst-line and the re-challenge phases to test the correlation
between circulating ctDNA biomarkers and treatment
response. Interestingly, the possibility of continuing pani-
tumumab beyond progression is also being investigated in
a multicenter single-arm phase II Japanese clinical trial of
second-line FOLFIRI plus panitumumab after rst-line
treatment with FOLFOX plus panitumumab in initial
RAS WT mCRC (UMIN000026817). Mutational status
using ctDNA will be prospectively assessed at multiple
time-points during this study as one of the planned sec-
ondary endpoints.
Conclusions
Panitumumab in association with chemotherapy is a valu-
able rst- or second-line treatment option in patients with
RAS WT mCRC, as well as a monotherapy option in
advanced lines for chemorefractory patients. The toxicity
prole of panitumumab is manageable and this agent has a
favorable impact on patients QoL, showing positive
results also in the population of frail and elderly mCRC
patients. Novel treatment scenarios are opening for pani-
tumumab including combinations with intensied che-
motherapy regimens to implement conversion to
resectability in initially unresectable patients and mainte-
nance treatment strategies. The development of panitumu-
mab has signicantly added to the treatment options for
RAS WT mCRC, and has contributed to expanding the
horizons of mCRC molecular proling.
Current efforts are directed to dissect the mechanisms
of primary resistance beyond RAS status and the
Battaglin et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
Cancer Management and Research 2019:11
5920
mechanisms of acquired resistance to panitumumab (and
more generally anti-EGFRs), which entails a more com-
prehensive molecular characterization of RAS WT tumors,
the assessment of additional mutational and clinico-patho-
logical features, ie BRAF status and tumor sidedness, and
the development of novel technologies to capture the
dynamic heterogeneity of the genomic landscape displayed
by mCRC under targeted treatment pressure. Recent
advancements in this eld warrant a prospective validation
of new predictive biomarkers in RAS WT mCRC, in order
to further rene patient selection and develop novel mole-
cularly-tailored treatment strategies to optimize outcomes
and patients benet.
Acknowledgments
This manuscript was partly supported by the National Cancer
Institute (grant number P30CA014089), the Gloria Borges
WunderGlo Foundation-The Wunder Project, the Dhont
Family Foundation, the San Pedro Peninsula Cancer Guild,
the Daniel Butler Research Fund, the Call to Cure Research
Fund, and the Fong Research Project. The content is solely
the responsibility of the authors and does not necessarily
represent the ofcial views of the National Cancer Institute
or the National Institutes of Health. Francesca Battaglin and
Alberto Puccini are co-rst authors for this study.
Author contributions
All authors contributed to data analysis, drafting and revis-
ing the article, gave nal approval of the version to be
published, and agree to be accountable for all aspects of
the work.
Disclosure
FB has received travel/accommodations from Bayer and
Amgen Inc. HJL has received clinical trial nancial support
from Merck Serono and Roche, honoraria for advisory
board membership and lectures from Bayer, Boehringer
Ingelheim, Genentech, Pzer, Merck Serono and Roche,
and travel/accommodations from Bayer, Merck Serono
and Roche. The authors report no other conicts of interest
in this work.
References
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J
Clin.2018;68(1):730.
2. National Comprehensive Cancer Network. Clinical practice guidelines in
oncology. Colon Cancer. Version 1.2019. Available from: https://www.nccn.
org/professionals/physician_gls/pdf/colon.pdf Accessed April 01, 2019.
3. Cremolini C, Schirripa M, Antoniotti C, et al. First-line chemother-
apy for mCRC-a review and evidence-based algorithm. Nat Rev Clin
Oncol.2015;12(10):607619. doi:10.1038/nrclinonc.2015.129
4. Vectibix® (Panitumumab). Thousand Oaks, CA: Amgen Inc.; 2017.
Available from: https://www.accessdata.fda.gov/drugsatfda_docs/
label/2017/125147s207lbl.pdf. Accessed January 16, 2019.
5. Citri A, Yarden Y. EGF-ERBB signalling: towards the systems level.
Nat Rev Molecular Cell Biol.2006;7(7):505516. doi:10.1038/nrm1962
6. Yang BB, Lum P, Chen A, et al. Pharmacokinetic and pharmacody-
namic perspectives on the clinical drug development of panitumu-
mab. Clin Pharmacokinet.2010;49(11):729740. doi:10.2165/
11535970-000000000-00000
7. Giannopoulou E, Antonacopoulou A, Matsouka P, Kalofonos HP.
Autophagy: novel action of panitumumab in colon cancer.
Anticancer Res.2009;29(12):50775082.
8. Allegra CJ, Rumble RB, Hamilton SR, et al. Extended RAS gene
mutation testing in metastatic colorectal carcinoma to predict
response to anti-epidermal growth factor receptor monoclonal anti-
body therapy: american society of clinical oncology provisional clin-
ical opinion update 2015. J Clin Oncol.2016;34(2):179185.
doi:10.1200/JCO.2015.63.9674
9. Sepulveda AR, Hamilton SR, Allegra CJ, et al. Molecular biomarkers for
the evaluation of colorectal cancer: guideline from the American society
for clinical pathology, college of American pathologists, association for
molecular pathology, and American society of clinical oncology. JMol
Diagn.2017;19(2):187225. doi:10.1016/j.jmoldx.2016.11.001
10. Van Cutsem E, Peeters M, Siena S, et al. Open-label phase III trial of
panitumumab plus best supportive care compared with best suppor-
tive care alone in patients with chemotherapy-refractory metastatic
colorectal cancer. J Clin Oncol.2007;25(13):16581664.
doi:10.1200/JCO.2006.08.1620
11. Amado RG, Wolf M, Peeters M, et al. Wild-type KRAS is required for
panitumumab efcacy in patients with metastatic colorectal cancer. J
Clin Oncol.2008;26(10):16261634. doi:10.1200/JCO.2007.14.7116
12. Price TJ, Peeters M, Kim TW, et al. Panitumumab versus cetuximab
in patients with chemotherapy-refractory wild-type KRAS exon 2
metastatic colorectal cancer (ASPECCT): a randomised, multicentre,
open-label, non-inferiority phase 3 study. Lancet Oncol.2014;15
(6):569579. doi:10.1016/S1470-2045(14)70118-4
13. Peeters M, Price TJ, Cervantes A, et al. Randomized phase III study
of panitumumab with uorouracil, leucovorin, and irinotecan
(FOLFIRI) compared with FOLFIRI alone as second-line treatment
in patients with metastatic colorectal cancer. J Clin Oncol.2010;28
(31):47064713. doi:10.1200/JCO.2009.27.6055
14. Peeters M, Oliner KS, Price TJ, et al. Analysis of KRAS/NRAS
mutations in a phase III study of panitumumab with FOLFIRI com-
pared with FOLFIRI alone as second-line treatment for metastatic
colorectal cancer. Clin Cancer Res.2015;21(24):54695479.
doi:10.1158/1078-0432.CCR-15-0526
15. Douillard JY, Siena S, Cassidy J, et al. Randomized, phase III trial of
panitumumab with infusional uorouracil, leucovorin, and oxaliplatin
(FOLFOX4) versus FOLFOX4 alone as rst-line treatment in patients
with previously untreated metastatic colorectal cancer: the PRIME study.
J Clin Oncol.2010;28(31):46974705. doi:10.1200/JCO.2009.27.4860
16. Douillard JY, Siena S, Cassidy J, et al. Final results from PRIME:
randomized phase III study of panitumumab with FOLFOX4 for rst-
line treatment of metastatic colorectal cancer. Ann Oncol.2014;25
(7):13461355. doi:10.1093/annonc/mdu141
17. Therkildsen C, Bergmann TK, Henrichsen-Schnack T, Ladelund S,
Nilbert M. The predictive value of KRAS, NRAS, BRAF, PIK3CA
and PTEN for anti-EGFR treatment in metastatic colorectal cancer: a
systematic review and meta-analysis. Acta Oncol.2014;53(7):852
864. doi:10.3109/0284186X.2014.895036
18. Douillard JY, Oliner KS, Siena S, et al. Panitumumab-FOLFOX4
treatment and RAS mutations in colorectal cancer. N Engl J Med.
2013;369(11):10231034. doi:10.1056/NEJMoa1305275
Dovepress Battaglin et al
Cancer Management and Research 2019:11 submit your manuscript | www.dovepress.com
DovePress 5921
19. Karthaus M, Hofheinz RD, Mineur L, et al. Impact of tumour RAS/
BRAF status in a rst-line study of panitumumab + FOLFIRI in
patients with metastatic colorectal cancer. Br J Cancer.2016;115
(10):12151222. doi:10.1038/bjc.2016.343
20. Peeters M, Oliner KS, Parker A, et al. Massively parallel tumor
multigene sequencing to evaluate response to panitumumab in a
randomized phase III study of metastatic colorectal cancer. Clin
Cancer Res.2013;19(7):19021912. doi:10.1158/1078-0432.CCR-
12-1913
21. Sorich MJ, Wiese MD, Rowland A, Kichenadasse G, McKinnon RA,
Karapetis CS. Extended RAS mutations and anti-EGFR monoclonal
antibody survival benet in metastatic colorectal cancer: a meta-
analysis of randomized, controlled trials. Ann Oncol.2015;26
(1):1321. doi:10.1093/annonc/mdu378
22. Rowland A, Dias MM, Wiese MD, et al. Meta-analysis of BRAF
mutation as a predictive biomarker of benet from anti-EGFR mono-
clonal antibody therapy for RAS wild-type metastatic colorectal can-
cer. Br J Cancer.2015;112(12):18881894. doi:10.1038/bjc.2015.173
23. Pietrantonio F, Petrelli F, Coinu A, et al. Predictive role of BRAF
mutations in patients with advanced colorectal cancer receiving
cetuximab and panitumumab: a meta-analysis. Eur J Cancer.
2015;51(5):587594. doi:10.1016/j.ejca.2015.01.054
24. van Brummelen EMJ, de Boer A, Beijnen JH, Schellens JHM. BRAF
mutations as predictive biomarker for response to anti-EGFR mono-
clonal antibodies. Oncologist.2017. doi:10.1634/theoncologist.2017-
0031
25. Schwartzberg LS, Rivera F, Karthaus M, et al. PEAK: a randomized,
multicenter phase II study of panitumumab plus modied uorour-
acil, leucovorin, and oxaliplatin (mFOLFOX6) or bevacizumab plus
mFOLFOX6 in patients with previously untreated, unresectable,
wild-type KRAS exon 2 metastatic colorectal cancer. J Clin Oncol.
2014;32(21):22402247. doi:10.1200/JCO.2013.53.2473
26. Rivera F, Karthaus M, Hecht JR, et al. Final analysis of the rando-
mised PEAK trial: overall survival and tumour responses during rst-
line treatment with mFOLFOX6 plus either panitumumab or bevaci-
zumab in patients with metastatic colorectal carcinoma. Int J
Colorectal Dis.2017;32(8):11791190. doi:10.1007/s00384-017-
2800-1
27. Peeters M, Forget F, Karthaus M, et al. Exploratory pooled analysis
evaluating the effect of sequence of biological therapies on overall
survival in patients with RAS wild-type metastatic colorectal carci-
noma. ESMO Open.2018;3(2):e000297. doi:10.1136/esmoopen-
2017-000297
28. Carrato A, Abad A, Massuti B, et al. First-line panitumumab plus
FOLFOX4 or FOLFIRI in colorectal cancer with multiple or
unresectable liver metastases: a randomised, phase II trial
(PLANET-TTD). Eur J Cancer.2017;81:191202. doi:10.1016/j.
ejca.2017.04.024
29. Taieb J, Rivera F, Siena S, et al. Exploratory analyses assessing the
impact of early tumour shrinkage and depth of response on survival
outcomes in patients with RAS wild-type metastatic colorectal cancer
receiving treatment in three randomised panitumumab trials. J
Cancer Res Clin Oncol.2018;144(2):321335. doi:10.1007/s00432-
017-2534-z
30. Goldberg RM, Montagut C, Wainberg ZA, et al. Optimising the use
of cetuximab in the continuum of care for patients with metastatic
colorectal cancer. ESMO Open.2018;3(4):e000353. doi:10.1136/
esmoopen-2018-000353
31. Cremolini C, Rossini D, DellAquila E, et al. Rechallenge for patients
with RAS and BRAF wild-type metastatic colorectal cancer with
acquired resistance to rst-line cetuximab and irinotecan: a phase 2
single-arm clinical trial. JAMA Oncol. Epub 2018 Nov 21.
32. Mauri G, Pizzutilo EG, Amatu A, et al. Retreatment with anti-EGFR
monoclonal antibodies in metastatic colorectal cancer: systematic
review of different strategies. Cancer Treat Rev.2019;73:4153.
doi:10.1016/j.ctrv.2018.12.006
33. Fornaro L, Lonardi S, Masi G, et al. FOLFOXIRI in combination
with panitumumab as rst-line treatment in quadruple wild-type
(KRAS, NRAS, HRAS, BRAF) metastatic colorectal cancer patients:
a phase II trial by the Gruppo Oncologico Nord Ovest (GONO). Ann
Oncol.2013;24(8):20622067. doi:10.1093/annonc/mdt165
34. Bendell JC, Zakari A, Peyton JD, et al. A phase II study of
FOLFOXIRI plus panitumumab followed by evaluation for resection
in patients with metastatic KRAS wild-type colorectal cancer with
liver metastases only. Oncologist.2016;21(3):279280. doi:10.1634/
theoncologist.2015-0439
35. Geissler M, Klingler T, Martens UM, et al. 453PD1st-line
mFOLFOXIRI + panitumumab vs FOLFOXIRI treatment of RAS
wt mCRC: a randomized phase II VOLFI trial of the AIO (KRK-
0109). Ann Oncol.2018;29(suppl_8). doi:10.1093/annonc/mdx807
36. Modest DP, Rivera F, Bachet JB, et al. Panitumumab-based main-
tenance after oxaliplatin discontinuation in metastatic colorectal can-
cer: a retrospective analysis of two randomised trials. Int J Cancer.
2019. doi:10.1002/ijc.32110
37. Pietrantonio F, Morano F, Corallo S, et al. First-line FOLFOX plus
panitumumab (Pan) followed by 5FU/LV plus pan or single-agent pan
as maintenance therapy in patients with RAS wild-type metastatic
colorectal cancer (mCRC): the VALENTINO study. J Clin Oncol.
2018;36(15_suppl):3505. doi:10.1200/JCO.2018.36.15_suppl.3505
38. Nakamura M, Munemoto Y, Takahashi M, et al. SAPPHIRE: a
randomized phase II study of mFOLFOX6+ panitumumab versus 5-
FU/LV + panitumumab after 6 cycles of frontline mFOLFOX6+
panitumumab in patients with colorectal cancer. J Clin Oncol.
2018;36(4_suppl):729. doi:10.1200/JCO.2018.36.4_suppl.729
39. Peeters M. Outcome according to Left Vs. Right Side in the
Panitumumab Studies. Presented At: ESMO 2016 Congress, Special
Session. Copenhagen, Denmark; October 711, 2016.
40. Boeckx N, Koukakis R, de Beeck KO, et al. Effect of primary tumor
location on second- or later-line treatment outcomes in patients with
RAS wild-type metastatic colorectal cancer and all treatment lines in
patients with RAS mutations in four randomized panitumumab stu-
dies. Clin Colorectal Cancer.2018;17(3):170178.e173. doi:10.1016/
j.clcc.2018.03.005
41. Tejpar S, Stintzing S, Ciardiello F, et al. Prognostic and predictive
relevance of primary tumor location in patients with RAS wild-type
metastatic colorectal cancer: retrospective analyses of the CRYSTAL
and FIRE-3 trials. JAMA Oncol.2017;3(2):194201. doi:10.1001/
jamaoncol.2016.3797
42. Peeters M, Price T, Taieb J, et al. Relationships between tumour
response and primary tumour location, and predictors of long-term
survival, in patients with RAS wild-type metastatic colorectal cancer
receiving rst-line panitumumab therapy: retrospective analyses of
the PRIME and PEAK clinical trials. Br J Cancer.2018;119(3):303
312. doi:10.1038/s41416-018-0165-z
43. Holch JW, Ricard I, Stintzing S, Modest DP, Heinemann V. The
relevance of primary tumour location in patients with metastatic
colorectal cancer: a meta-analysis of rst-line clinical trials. Eur J
Cancer.2017;70:8798. doi:10.1016/j.ejca.2016.10.007
44. Arnold D, Lueza B, Douillard JY, et al. Prognostic and predictive
value of primary tumour side in patients with RAS wild-type meta-
static colorectal cancer treated with chemotherapy and EGFR direc-
ted antibodies in six randomised trials. Ann Oncol.2017.
doi:10.1093/annonc/mdx175
45. Stintzing S, Tejpar S, Gibbs P, Thiebach L, Lenz HJ. Understanding
the role of primary tumour localisation in colorectal cancer treatment
and outcomes. Eur J Cancer.2017;84:6980. doi:10.1016/j.
ejca.2017.07.016
46. Wagner LI, Lacouture ME. Dermatologic toxicities associated with
EGFR inhibitors: the clinical psychologists perspective. Impact on
health-related quality of life and implications for clinical manage-
ment of psychological sequelae. Oncology (Williston Park).2007;21
(11 Suppl 5):3436.
Battaglin et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
Cancer Management and Research 2019:11
5922
47. Kohne CH, Hofheinz R, Mineur L, et al. First-line panitumumab plus
irinotecan/5-uorouracil/leucovorin treatment in patients with meta-
static colorectal cancer. J Cancer Res Clin Oncol.2012;138(1):65
72. doi:10.1007/s00432-011-1061-6
48. Thaler J, Karthaus M, Mineur L, et al. Skin toxicity and quality of
life in patients with metastatic colorectal cancer during rst-line
panitumumab plus FOLFIRI treatment in a single-arm phase II
study. BMC Cancer.2012;12:438. doi:10.1186/1471-2407-12-438
49. Siena S, Tabernero J, Bodoky G, et al. Quality of life during rst-line
FOLFOX4±panitumumab in RAS wild-type metastatic colorectal
carcinoma: results from a randomised controlled trial. ESMO Open.
2016;1(2):e000041. doi:10.1136/esmoopen-2016-000041
50. Bennett L, Zhao Z, Barber B, et al. Health-related quality of life in
patients with metastatic colorectal cancer treated with panitumumab
in rst- or second-line treatment. Br J Cancer.2011;105(10):1495
1502. doi:10.1038/bjc.2011.409
51. Odom D, Barber B, Bennett L, et al. Health-related quality of life and
colorectal cancer-specic symptoms in patients with chemotherapy-
refractory metastatic disease treated with panitumumab. Int J
Colorectal Dis.2011;26(2):173181. doi:10.1007/s00384-010-1112-5
52. Vectibix. INN-panitumumab. Available from: https://www.ema.
europa.eu/en/documents/product-information/vectibix-epar-product-
information_en.pdf Accessed March 31, 2019.
53. Mitchell EP, Piperdi B, Lacouture ME, et al. The efcacy and safety
of panitumumab administered concomitantly with FOLFIRI or
Irinotecan in second-line therapy for metastatic colorectal cancer:
the secondary analysis from STEPP (Skin Toxicity Evaluation
Protocol With Panitumumab) by KRAS status. Clin Colorectal
Cancer.2011;10(4):333339. doi:10.1016/j.clcc.2011.06.004
54. Lacouture ME, Mitchell EP, Piperdi B, et al. Skin toxicity evaluation
protocol with panitumumab (STEPP), a phase II, open-label, rando-
mized trial evaluating the impact of a pre-emptive skin treatment
regimen on skin toxicities and quality of life in patients with meta-
static colorectal cancer. J Clin Oncol.2010;28(8):13511357.
doi:10.1200/JCO.2008.21.7828
55. Peeters M, Siena S, Van Cutsem E, et al. Association of progression-
free survival, overall survival, and patient-reported outcomes by skin
toxicity and KRAS status in patients receiving panitumumab mono-
therapy. Cancer.2009;115(7):15441554. doi:10.1002/cncr.24088
56. Lacouture ME, Anadkat M, Jatoi A, Garawin T, Bohac C, Mitchell E.
Dermatologic toxicity occurring during anti-EGFR monoclonal inhi-
bitor therapy in patients with metastatic colorectal cancer: a systema-
tic review. Clin Colorectal Cancer.2018;17(2):8596. doi:10.1016/j.
clcc.2017.12.004
57. Kobayashi Y, Komatsu Y, Yuki S, et al. Randomized controlled trial
on the skin toxicity of panitumumab in Japanese patients with meta-
static colorectal cancer: HGCSG1001 study; J-STEPP. Future Oncol.
2015;11(4):617627. doi:10.2217/fon.14.251
58. Fakih M, Vincent M. Adverse events associated with anti-EGFR
therapies for the treatment of metastatic colorectal cancer. Curr
Oncol.2010;17(Suppl 1):S18S30. doi:10.3747/co.v17is1.615
59. Costa A, Tejpar S, Prenen H, Van Cutsem E. Hypomagnesaemia
and targeted anti-epidermal growth factor receptor (EGFR)
agents. Target Oncol.2011;6(4):227233. doi:10.1007/s11523-
011-0200-y
60. Vincenzi B, Galluzzo S, Santini D, et al. Early magnesium mod-
ications as a surrogate marker of efcacy of cetuximab-based
anticancer treatment in KRAS wild-type advanced colorectal cancer
patients. Ann Oncol.2011;22(5):11411146. doi:10.1093/annonc/
mdq550
61. Sastre J, Massuti B, Pulido G, et al. First-line single-agent panitu-
mumab in frail elderly patients with wild-type KRAS metastatic
colorectal cancer and poor prognostic factors: a phase II study of
the Spanish cooperative group for the treatment of digestive
tumours. Eur J Cancer.2015;51(11):13711380. doi:10.1016/j.
ejca.2015.04.013
62. Pietrantonio F, Cremolini C, Aprile G, et al. Single-agent panitu-
mumab in frail elderly patients with advanced RAS and BRAF
wild-type colorectal cancer: challenging drug label to light up new
hope. Oncologist.2015;20(11):12611265. doi:10.1634/theoncolo-
gist.2015-0171
63. Douillard J, Siena S, Peeters M, Koukakis R, Terwey J, Tabernero J.
547pimpact of baseline age on efcacy and safety of rst-line pani-
tumumab (PMAB) + folfox4 vs folfox4 treatment. Ann Oncol.
2014;25(suppl_4):iv187iv187. doi:10.1093/annonc/mdu333.49
64. Asimakopoulou N, Souglakos J, Kentepozidis N, et al. Efcacy of
panitumumab in older patients with metastatic colorectal cancer: a
retrospective analysis using the database of the Hellenic Oncology
Research Group (HORG). J Geriatr Oncol.2019;10(1):143148.
doi:10.1016/j.jgo.2018.08.002
65. Bertotti A, Papp E, Jones S, et al. The genomic landscape of response
to EGFR blockade in colorectal cancer. Nature.2015;526(7572):263
267. doi:10.1038/nature14969
66. Bertotti A, Migliardi G, Galimi F, et al. A molecularly annotated
platform of patient-derived xenografts (xenopatients)identies
HER2 as an effective therapeutic target in cetuximab-resistant color-
ectal cancer. Cancer Discov.2011;1(6):508523. doi:10.1158/2159-
8290.CD-11-0109
67. Sartore-Bianchi A, Trusolino L, Martino C, et al. Dual-targeted
therapy with trastuzumab and lapatinib in treatment-refractory,
KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal
cancer (HERACLES): a proof-of-concept, multicentre, open-label,
phase 2 trial. Lancet Oncol.2016;17(6):738746. doi:10.1016/
S1470-2045(16)00150-9
68. Townsend A, Tebbutt N, Karapetis C, et al. Phase IB/II study of
second-line therapy with Panitumumab, Irinotecan, and Everolimus
(PIE) in KRAS Wild-Type Metastatic Colorectal Cancer. Clin Cancer
Res.2018;24(16):38383844. doi:10.1158/1078-0432.CCR-17-3590
69. Corcoran RB, Andre T, Atreya CE, et al. Combined BRAF, EGFR,
and MEK inhibition in patients with BRAF(V600E)-mutant color-
ectal cancer. Cancer Discov.2018;8(4):428443. doi:10.1158/2159-
8290.CD-17-1226
70. Cremolini C, Morano F, Moretto M, et al. Dissecting primary resis-
tance to anti-EGFRs in RAS and BRAF wt metastatic colorectal
cancer (mCRC): a case-control study. J Clin Oncol.2017;35(suppl;
abstr 11508). doi:10.1200/JCO.2017.35.15_suppl.11508
71. Misale S, Di Nicolantonio F, Sartore-Bianchi A, Siena S, Bardelli A.
Resistance to anti-EGFR therapy in colorectal cancer: from hetero-
geneity to convergent evolution. Cancer Discov.2014;4(11):1269
1280. doi:10.1158/2159-8290.CD-14-0462
72. Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating
tumor DNA in early- and late-stage human malignancies. Sci Transl
Med.2014;6(224):224ra224. doi:10.1126/scitranslmed.3007094
73. Siravegna G, Mussolin B, Buscarino M, et al. Clonal evolution and
resistance to EGFR blockade in the blood of colorectal cancer
patients. Nat Med.2015;21(7):795801. doi:10.1038/nm.3870
74. Morelli MP, Overman MJ, Dasari A, et al. Characterizing the patterns
of clonal selection in circulating tumor DNA from patients with
colorectal cancer refractory to anti-EGFR treatment. Ann Oncol.
2015;26(4):731736. doi:10.1093/annonc/mdv005
75. Parseghian CM, Loree JM, Morris VK, et al. Anti-EGFR-resistant clones
decay exponentially after progression: implications for anti-EGFR re-
challenge. Ann Oncol.2019;30(2):243249. doi:10.1093/annonc/mdy509
76. Sartore-Bianchi A, Siena S, Bardelli A, et al. Dynamic molecular
analysis and clinical correlates of tumor evolution within a phase II
trial of panitumumab-based therapy in metastatic colorectal cancer.
Ann Oncol.2017;29(1):119126.
77. Peeters M, Price T, Boedigheimer M, et al. Evaluation of emergent
mutations in circulating cell-free DNA and clinical outcomes in
patients with metastatic colorectal cancer treated with panitumumab
in the ASPECCT study. Clin Cancer Res.2019;25(4):12161225.
doi:10.1158/1078-0432.CCR-18-2072
Dovepress Battaglin et al
Cancer Management and Research 2019:11 submit your manuscript | www.dovepress.com
DovePress 5923
78. Kim TW, Peeters M, Thomas A, et al. Impact of emergent circulating
tumor DNA RAS mutation in panitumumab-treated chemoresistant
metastatic colorectal cancer. Clin Cancer Res.2018;24(22):5602
5609. doi:10.1158/1078-0432.CCR-17-3377
79. Santos C, Azuara D, Vieitez JM, et al. Phase II study of high-
sensitivity genotyping of KRAS, NRAS, BRAF and PIK3CA to
ultra-select metastatic colorectal cancer patients for panitumumab
plus FOLFIRI: the ULTRA trial. Ann Oncol.2019;30(5):796803.
doi:10.1093/annonc/mdz082
80. Kim TW, Elme A, Kusic Z, et al. A phase 3 trial evaluating
panitumumab plus best supportive care vs best supportive care in
chemorefractory wild-type KRAS or RAS metastatic colorectal
cancer. Br J Cancer.2016;115(10):12061214. doi:10.1038/
bjc.2016.309
81. Peeters M, Price TJ, Cervantes A, et al. Final results from a rando-
mized phase 3 study of FOLFIRI {±} panitumumab for second-line
treatment of metastatic colorectal cancer. Ann Oncol.2014;25
(1):107116. doi:10.1093/annonc/mdt523
82. Hecht JR, Cohn A, Dakhil S, et al. SPIRITT: a randomized,
multicenter, phase II study of panitumumab with FOLFIRI and
bevacizumab with FOLFIRI as second-line treatment in patients
with unresectable wild type KRAS metastatic colorectal cancer.
Clin Colorectal Cancer.2015;14(2):7280. doi:10.1016/j.
clcc.2014.12.009
Cancer Management and Research Dovepress
Publish your work in this journal
Cancer Management and Research is an international, peer-reviewed
open access journal focusing on cancer research and the optimal use of
preventative and integrated treatment interventions to achieve improved
outcomes, enhanced survival and quality of life for the cancer patient.
The manuscript management system is completely online and includes
a very quick and fair peer-review system, which is all easy to use.
Visit http://www.dovepress.com/testimonials.php to read real quotes
from published authors.
Submit your manuscript here: https://www.dovepress.com/cancer-management-and-research-journal
Battaglin et al Dovepress
submit your manuscript | www.dovepress.com
DovePress
Cancer Management and Research 2019:11
5924
... Cetuximab and panitumumab were equally effective in the phase III ASPECCT study for first-line therapy of CRC. However, anti-EGFR drugs are not a top priority for second-line therapy, as they have shown modest efficacy in multiple studies [14]. ...
... DN-30, an antibody that binds to the MET's IPT (immunoglobulin-like plexins transcription factor domain), shows potential to inhibit the spread of MET-positive metastatic melanoma and gastric cancer. ABT-700, a humanized antibody, has completed phase I clinical trials for several solid tumors and has demonstrated tumor regression in preclinical cancer models with MET amplification [14]. ...
Article
Full-text available
Colon cancer is one of the most common cancers in the United States of America. In addition to conventional treatment approaches such as surgery, chemotherapy, and radiation for colorectal cancer, immunotherapy has gained recognition over the past few years. However, its effectiveness in colorectal cancer treatment is controversial. Our study investigates the survival and progression-free rates of immunotherapy for different types of colorectal cancer over the last 10 years. We conducted literature reviews from various clinical trials and research studies to evaluate immunotherapy's role in colorectal cancer treatment. We also investigated how it affects clinical outcomes. We discovered a range of effective immunotherapy approaches targeting various growth factors and signaling pathways. These modalities include monoclonal antibodies aimed at growth factors such as epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), human epidermal growth factor receptor 2 (HER2), and downstream signaling pathways such as mitogen-activated protein kinase (MAPK), kirsten rat sarcoma viral oncogene (KRAS), B-raf proto-oncogene, serine/threonine kinase (BRAF), and phosphatase and tensin homolog (PTEN). Additionally, we identified immune checkpoint inhibitors, such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) inhibitors and programmed cell death ligand 1 (PD-L1) inhibitors, as well as target therapy and adoptive cell therapy as promising immunotherapeutic options. Nevertheless, the application of immunotherapy remains highly limited due to various factors influencing survival and progression-free rates, including tumor microenvironment, microsatellite instability, immune checkpoint expression, and gut microbiome. Additionally, its effectiveness is restricted to a small subgroup of patients, accompanied by side effects and the development of drug resistance mechanisms. To unlock its full potential, further clinical trials and research on molecular pathways in colorectal cancer are imperative. This will ultimately enhance drug discovery success and lead to more effective clinical management approaches.
... Previous clinical reports on targeted agents in patients with mCRC have shown that treatment can provide survival benefits without diminishing QoL. [41][42][43][44][45][46][47][48] This includes studies Figure 4. Median time to definitive deterioration a in FACT-C subscales for encorafenib plus cetuximab with or without binimetinib compared with control: (A) Functional well-being; (B) physical well-being; (C) social/family well-being; (D) emotional well-being. Probability (%) of median time to definitive 10% deterioration in FACT-C colorectal cancer subscales (A) functional well-being, (B) physical well-being, (C) social/family well-being, and (D) emotional well-being for patients treated with encorafenib plus cetuximab with or without binimetinib (triplet and doublet, respectively) compared with the control. ...
... [41][42][43] Similar findings have been reported for bevacizumab, panitumumab, aflibercept, and regorafenib. [44][45][46][47][48] In addition to improving OS as reported previously, encorafenib plus cetuximab with or without binimetinib delays QoL decline in previously treated patients with BRAF V600E-mutant mCRC. ...
Article
Full-text available
Background In the BEACON CRC study (NCT02928224), encorafenib plus cetuximab with binimetinib {9.3 versus 5.9 months; hazard ratio (HR) [95% confidence interval (CI)]: 0.60 [0.47-0.75]} or without binimetinib [9.3 versus 5.9 months; HR (95% CI): 0.61 (0.48-0.77)] significantly improved overall survival (OS) compared with the previous standard of care (control) in patients with BRAF V600E metastatic colorectal cancer (mCRC). Quality of life (QoL) was a secondary endpoint, assessed using validated instruments. Patients and methods BEACON CRC was a randomized, open-label, phase III study comparing encorafenib plus cetuximab with or without binimetinib and the investigator’s choice of irinotecan plus cetuximab or FOLFIRI plus cetuximab (chemotherapy control) in patients with previously treated BRAF V600E mCRC. Patient-reported QoL assessments included the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC) and Functional Assessment of Cancer Therapy—Colorectal (FACT-C). The primary outcome for these tools was time to definitive 10% deterioration. Results Encorafenib plus cetuximab, both with and without binimetinib, was associated with longer median times to definitive 10% deterioration versus the control group in the EORTC Global Health Status scale [HR (95% CI): 0.65 (0.52-0.80) versus 0.61 (0.49-0.75), respectively] and the FACT-C functional well-being subscale [HR (95% CI): 0.62 (0.50-0.76) versus 0.58 (0.47-0.72), respectively]. Consistent results were observed across all subscales of the EORTC and FACT-C instruments. QoL was generally maintained during treatment for the global EORTC and FACT-C scales. Conclusions In addition to improving OS, encorafenib plus cetuximab with or without binimetinib delays QoL decline in previously treated patients with BRAF V600E-mutant mCRC.
... The review by Battaglin et al. [46] highlighted the efficacy of panitumumab, either in combination with chemotherapy as a first-or second-line treatment or as a monotherapy in advanced lines for chemorefractory patients with RAS wild-type metastatic CRC, according to the analyzed trials [47][48][49]; however, quality of life was not directly assessed in these trials. Among other conclusions Battaglin et al., underscored the manageable toxicity profile of panitumumab and its favorable impact on the QoL for patients, including those who are frail and elderly. ...
Article
Full-text available
This systematic review critically evaluates the impact of systemic treatments on outcomes and quality of life (QoL) in patients with RAS-positive stage IV colorectal cancer, with studies published up to December 2023 across PubMed, Scopus, and Web of Science. From an initial pool of 1345 articles, 11 relevant studies were selected for inclusion, encompassing a diverse range of systemic treatments, including panitumumab combined with FOLFOX4 and FOLFIRI, irinotecan paired with panitumumab, regorafenib followed by cetuximab ± irinotecan and vice versa, and panitumumab as a maintenance therapy post-induction. Patient demographics predominantly included middle-aged to elderly individuals, with a slight male predominance. Racial composition, where reported, showed a majority of Caucasian participants, highlighting the need for broader demographic inclusivity in future research. Key findings revealed that the addition of panitumumab to chemotherapy (FOLFOX4 or FOLFIRI) did not significantly compromise QoL while notably improving disease-free survival, with baseline EQ-5D HSI mean scores ranging from 0.76 to 0.78 and VAS mean scores from 70.1 to 74.1. Improvements in FACT-C scores and EQ-5D Index scores particularly favored panitumumab plus best supportive care in KRAS wild-type mCRC, with early dropout rates of 38–42% for panitumumab + BSC. Notably, cetuximab + FOLFIRI was associated with a median survival of 25.7 months versus 16.4 months for FOLFIRI alone, emphasizing the potential benefits of integrating targeted therapies with chemotherapy. In conclusion, the review underscores the significant impact of systemic treatments, particularly targeted therapies and their combinations with chemotherapy, on survival outcomes and QoL in patients with RAS-positive stage IV colorectal cancer, and the need for personalized treatment.
... In my opinion, the pragmatic design of the NORDIC9-study properly models the real-world setting where physicians often avoid the use of an EGFRi in combination with chemotherapy as first-line option in vulnerable older patients (even with left-sided primary tumor), mainly due to the need for injection port, skin toxicities, and the related supportive care issues [296,297]. ...
Thesis
Full-text available
Colorectal cancer is accountable for the second highest mortality among all cancer types and predominantly affects adults aged 70 years or older. Owing to the aging populations worldwide, the number of older adults with cancer is steeply increasing including those with colorectal cancer. Despite the majority of patients with metastatic colorectal cancer (mCRC) is older than 70 years, the optimal treatment strategy is less clear due to the underrepresentation of older adults in randomized controlled trials and very few clinical trials dedicated to this large group of patients. The investigator-initiated randomized phase II study, the NORDIC9, prospectively tested an approach among older patients with mCRC who were not considered being candidates for full-dose combination treatment; reduced-dose combination vs full-dose single agent chemotherapy. The NORDIC9-study included 160 patients from four Nordic countries between March 2015 and October 2017 and established that reduced-dose combination chemotherapy resulted in significant prolonged progression-free survival with a trend towards prolonged overall survival, fewer toxicities, and hospital admissions compared to full-dose monotherapy. These encouraging results added important knowledge to the literature and raised several questions regarding the patients' quality of life and the potential prognostic and predictive factors in the NORDIC9 cohort. Therefore, we conducted four sub-studies further investigating quality of life, the prognostic value of systemic inflammation explored through plasma biomarkers, the prognostic value of physical functioning, and the RAS/BRAF mutation status. Finally, we also explored the predictive value of treatment allocation according to RAS/BRAF status. We found that patients receiving reduced-dose combination treatment maintained their quality of life and physical performance and had lower symptom burden compared to the full-dose monotherapy arm. The results of the biomarker analysis clearly demonstrated that patients who had elevated plasma C-reactive protein (CRP) at inclusion had significant shorter overall survival and progression-free survival than those with normal level of CRP, regardless of treatment. When analyzing survival outcomes according to physical performance, we found that Eastern Cooperative Oncology Group performance status and Vulnerable Elderly Survey-13 were proper tools to foresee shorter survival. These tools evaluate activities of daily living and instrumental activities of daily living in different ways, although, patients with higher scores on these scales had significantly higher risk for shorter survival. RAS/BRAF mutation status is a cornerstone of the therapeutic decision-making in young and fit adults with mCRC; however, its real clinical impact on daily clinical practice is less known and its potential role determining the use of targeted therapies is less established in older adults with mCRC. Our results demonstrate that patients whose mCRC was driven by BRAFV600E mutation had significantly shorter overall survival and progression-free survival compared to patients presented with other molecular sub-types. In this particular group of patients with BRAFV600E mutation, the use of reduced-dose combination chemotherapy resulted in a remarkable overall survival advantage compared to the full-dose monotherapy arm. I conclude that reduced-dose combination chemotherapy is the treatment of choice in older adults with mCRC who are ineligible for intensive chemotherapy. Its use resulted in prolonged survival, less toxicities, fewer hospital admissions, and preserved quality of life and physical performance. Using biomarkers, measurements of physical functioning, and RAS/BRAF mutation status provide important prognostic information and have therapeutic implication, thus, contribute to proper patient selection. These factors may influence survival and enhance communication in the shared decision-making process.
Article
Full-text available
Members of the EGFR family of tyrosine kinase receptors are major regulators of cellular proliferation, differentiation, and survival. In humans, abnormal activation of EGFR is associated with the development and progression of many cancer types, which makes it an attractive target for molecular-guided therapy. Two classes of EGFR-targeted cancer therapeutics include monoclonal antibodies (mAbs), which bind to the extracellular domain of EGFR, and tyrosine kinase inhibitors (TKIs), which mostly target the intracellular part of EGFR and inhibit its activity in molecular signaling. While EGFR-specific mAbs and three generations of TKIs have demonstrated clinical efficacy in various settings, molecular evolution of tumors leads to apparent and sometimes inevitable resistance to current therapeutics, which highlights the need for deeper research in this field. Here, we tried to provide a comprehensive and systematic overview of the rationale, molecular mechanisms, and clinical significance of the current EGFR-targeting drugs, highlighting potential candidate molecules in development. We summarized the underlying mechanisms of resistance and available personalized predictive approaches that may lead to improved efficacy of EGFR-targeted therapies. We also discuss recent developments and the use of specific therapeutic strategies, such as multi-targeting agents and combination therapies, for overcoming cancer resistance to EGFR-specific drugs.
Article
Fibroblast growth factor 2 (FGF-2) is not only an angiogenic factor, but also a mitogen for epidermal keratinocytes. FGF-2 has been shown to be positively immunoreactive in the basal layer of psoriatic lesions. In previous work, we used the Escherichia coli (E. coli) expression system to biosynthesize a biologically active anti-FGF-2 nanobody (Nb) screened by phage display technology, but the low yield limited its clinical application. In this study, we aimed to increase the yield of anti-FGF-2 Nb, and evaluate its therapeutic potential for psoriasis by inhibiting FGF-2-mediated mitogenic signaling in psoriatic epidermal keratinocytes. We demonstrated a 16-fold improvement in the yield of anti-FGF-2 Nb produced in the Pichia pastoris (P. pastoris) compared to the E. coli expression system. In vitro, the FGF-2-induced HaCaT cell model (FHCM) was established to mimic the key feature of keratinocyte overproliferation in psoriasis. Anti-FGF-2 Nb was able to effectively inhibit the proliferation and migration of FHCM. In vivo, anti-FGF-2 Nb attenuated the severity of imiquimod (IMQ)-induced psoriatic lesions in mice, and also improved the inflammatory microenvironment by inhibiting the secretion of inflammatory cytokines (IL-1β, IL-6, IL-23, and TNF-α), chemokines (CXCL1 and CCL20), and neutrophil infiltration in skin lesions. These were mainly related to the suppression of FGF-2-mediated mitogenic signaling in psoriatic keratinocytes. In conclusion, we have improved the production of anti-FGF-2 Nb and demonstrated the modality of attenuating the abnormal proliferative behavior of psoriatic keratinocytes by inhibiting FGF-2-mediated mitogenic signaling, which offers the possibility of treating psoriasis.
Article
Full-text available
Colorectal cancer (CRC) is one of the most deaths causing diseases worldwide. Several risk factors including hormones like insulin and insulin like growth factors (e.g., IGF-1) have been considered responsible for growth and progression of colon cancer. Though there is a huge advancement in the available screening as well as treatment techniques for CRC. There is no significant decrease in the mortality of cancer patients. Moreover, the current treatment approaches for CRC are associated with serious challenges like drug resistance and cancer re�growth. Given the severity of the disease, there is an urgent need for novel therapeutic agents with ideal characteristics. Several pieces of evidence suggested that natural products, specifically medicinal plants, and derived phytochemicals may serve as potential sources for novel drug discovery for various diseases including cancer. On the other hand, cancer cells like colon cancer require a high basal level of reactive oxygen species (ROS) to maintain its own cellular functions. However, excess production of intracellular ROS leads to cancer cell death via disturbing cellular redox homeostasis. Therefore, medicinal Colorectal cancer (CRC) is one of the most deaths causing diseases worldwide. Several risk factors including hormones like insulin and insulin like growth factors (e.g., IGF-1) have been considered responsible for growth and progression of colon cancer. Though there is a huge advancement in the available screening as well as treatment techniques for CRC. There is no significant decrease in the mortality of cancer patients. Moreover, the current treatment approaches for CRC are associated with serious challenges like drug resistance and cancer regrowth. Given the severity of the disease, there is an urgent need for novel therapeutic agents with ideal characteristics. Several pieces of evidence suggested that natural products, specifically medicinal plants, and derived phytochemicals may serve as potential sources for novel drug discovery for various diseases including cancer. On the other hand, cancer cells like colon cancer require a high basal level of reactive oxygen species (ROS) to maintain its own cellular functions. However, excess production of intracellular ROS leads to cancer cell death via disturbing cellular redox homeostasis. Therefore, medicinal plants and derived phytocompounds that can enhance the intracellular ROS and induce apoptotic cell death in cancer cells via modulating various molecular targets including IGF-1 could be potential therapeutic agents. Alkaloids form a major class of such phytoconstituents that can play a key role in cancer prevention. Moreover, several preclinical and clinical studies have also evidenced that these compounds show potent anti-colon cancer effects and exhibit negligible toxicity towards the normal cells. Hence, the present evidence�based study aimed to provide an update on various alkaloids that have been reported to induce ROS-mediated apoptosis in colon cancer cells via targeting various cellular components including hormones and growth factors, which play a role in metastasis, angiogenesis, proliferation, and invasion. This study also provides an individual account on each such alkaloid that underwent clinical trials either alone or in combination with other clinical drugs. In addition, various classes of phytochemicals that induce ROS-mediated cell death in different kinds of cancers including colon cancer are discussed.
Article
Micro abstract: This retrospective observational study assessed real-world treatment patterns and clinical outcomes among first-line MSI-H/dMMR metastatic colorectal cancer patients. Of 150 patients in the study cohort, 38.7% were treated with chemotherapy and 61.3% with chemotherapy + EGFR/VEGF inhibitor (EGFRi/VEGFi). Clinical outcomes were better among patients who received chemotherapy + EGFR/VEGF inhibitor than those who received chemotherapy. Introduction: Prior to pembrolizumab approval in first-line (1L) treatment of MSI-H/dMMR metastatic colorectal cancer (mCRC), patients were managed with chemotherapy with or without an EGFRi or VEGFi, agnostic of biomarker testing or mutation status. This study assessed real-world treatment patterns and clinical outcomes among 1L MSI-H/dMMR mCRC patients treated with standard of care (SOC). Patients and methods: Retrospective observational evaluation of patients ≥18 years diagnosed with stage IV MSI-H/dMMR mCRC who received community-based oncology care. Eligible patients were identified (01-Jun-2017 - 29-Feb-2020) and followed longitudinally until 31-Aug-2020/the last patient record/date of death. Descriptive statistics and Kaplan-Meier analyses were conducted. Results: Of 150 1L MSI-H/dMMR mCRC patients, 38.7% were treated with chemotherapy and 61.3% with chemotherapy + EGFRi/VEGFi. Accounting for censoring, the overall median real-world time to treatment discontinuation (95% CI) was 5.3 (4.4, 5.8) months; 3.0 (2.1, 4.4) and 6.2 (5.5, 7.6) months in the chemotherapy and chemotherapy + EGFRi/VEGFi cohorts, respectively. The combined median overall survival was 27.7 (23.2, not reached [NR]) months; 25.3 (14.5, NR) and 29.8 (23.2, NR) months in the chemotherapy and chemotherapy + EGFRi/VEGFi cohorts, respectively. The overall median real-world progression-free survival was 6.8 (5.3, 7.8) months; 4.2 (2.8, 6.1) and 7.7 (6.1, 10.2) months in the chemotherapy and chemotherapy + EGFRi/VEGFi cohorts, respectively. Conclusions: 1L MSI-H/dMMR mCRC patients receiving chemotherapy with EGFRi/VEGFi had better outcomes than those receiving only chemotherapy. An unmet need and opportunity to improve outcomes exists in this population that may be addressed by newer treatments like immunotherapies.
Article
The established cancer treatment strategy in clinical setting is based on chemo and radiation therapy, having limitations due to severe side-effects and drug-resistance. Small molecule chemo-drugs target any fast-dividing cells irrespective of healthy or defective origin. As a result, a substantial amount of healthy tissue is also destroyed. Moreover, failure to recognize the heterogeneity of tumour tissue results in drug-resistance over the course of time. On the other hand, peptides and proteins actively target somatic changes that are signature to any specific tumour tissue. Development and metastasis of cancer cells require unique disruption/alteration of protein activity. Identification of those wild and cancerous genotypes and phenotypes is the key to establishing easy 'targets' for protein based targeted therapeutics. The approach is cytostatic and tissue specific, which reduces drug toxicity. Biopharmaceutical products based on proteins and peptides are slowly re-directing oncology from cytotoxic small molecular treatment approach to target oriented cytostatic strategy. This review focuses on current and upcoming peptide and protein-based precision therapeutics. At the same time, the study also shades light on the technological advancement in the field of protein and peptide-based therapeutics.
Article
Full-text available
Background: Several studies show the importance of accurately quantifying not only KRAS and other low-abundant mutations because benefits of anti-EGFR therapies may depend on certain sensitivity thresholds. We assessed whether ultra-selection of patients using a high-sensitive digital PCR (dPCR) to determine KRAS, NRAS, BRAF and PIK3CA status can improve clinical outcomes of panitumumab plus FOLFIRI. Patients and methods: This was a single-arm phase II trial that analysed 38 KRAS, NRAS, BRAF and PIK3CA hotspots in tumour tissues of irinotecan-resistant metastatic colorectal cancer patients who received panitumumab plus FOLFIRI until disease progression or early withdrawal. Mutation profiles were identified by nanofluidic dPCR and correlated with clinical outcomes (ORR, overall response rate; PFS, progression-free survival; OS, overall survival) using cut-offs from 0% to 5%. A quantitative PCR (qPCR) analysis was also performed. Results: Seventy-two evaluable patients were enrolled. RAS (KRAS/NRAS) mutations were detected in 23 (32%) patients and RAS/BRAF mutations in 25 (35%) by dPCR, while they were detected in 7 (10%) and 11 (15%) patients, respectively, by qPCR. PIK3CA mutations were not considered in the analyses as they were only detected in 2 (3%) patients by dPCR and in 1 (1%) patient by qPCR. The use of different dPCR cut-offs for RAS (KRAS/NRAS) and RAS/BRAF analyses translated into differential clinical outcomes. The highest ORR, PFS and OS in wild-type patients with their lowest values in patients with mutations were achieved with a 5% cut-off. We observed similar outcomes in RAS/BRAF wild-type and mutant patients defined by qPCR. Conclusions: High-sensitive dPCR accurately identified patients with KRAS, NRAS, BRAF and PIK3CA mutations. The optimal RAS/BRAF mutational cut-off for outcome prediction is 5%, which explains that the predictive performance of qPCR was not improved by dPCR. The biological and clinical implications of low frequent mutated alleles warrant further investigations.
Article
Full-text available
Panitumumab is approved for RAS wild‐type metastatic colorectal cancer and was evaluated in Phase III (PRIME, NCT00364013) and Phase II (PEAK, NCT00819780) first‐line randomised studies. This retrospective analysis of these trials investigated efficacy and toxicity of panitumumab‐based maintenance after oxaliplatin discontinuation in RAS wild‐type patients. First‐line regimens were FOLFOX4 ± panitumumab in PRIME and mFOLFOX6 plus panitumumab or mFOLFOX6 plus bevacizumab in PEAK. Outcomes included median progression‐free survival (PFS) and overall survival (OS), from randomisation and oxaliplatin discontinuation, and toxicity. Overall, median duration of panitumumab plus 5‐fluorouracil/leucovorin (5‐FU/LV) maintenance was 21 (interquartile range: 11–41) weeks; that of 5‐FU/LV ± bevacizumab maintenance was 16 (6–31) weeks. Median OS from randomisation was 40.2 (95% confidence interval: 30.3–50.4) and 39.1 (34.2–63.0) months for panitumumab plus 5‐FU/LV maintenance and 24.1 (17.7–33.0) and 28.9 (21.0–32.0) months for 5‐FU/LV ± bevacizumab maintenance in PRIME and PEAK, respectively. Median PFS from randomisation was 16.6 (11.3–23.6) and 15.4 (11.6–18.4) months for panitumumab plus 5‐FU/LV maintenance and 12.6 (9.4–16.2) and 13.1 (9.5–16.6) months for 5‐FU/LV ± bevacizumab maintenance in PRIME and PEAK, respectively. From oxaliplatin discontinuation, median OS was 33.9 (24.7–42.8) and 33.5 (24.5–54.9) months for panitumumab plus 5‐FU/LV maintenance and 16.4 (12.4–24.1) and 23.3 (15.7–26.3) months for 5‐FU/LV ± bevacizumab maintenance in PRIME and PEAK, respectively; PFS was 11.7 (7.8–19.2) and 9.7 (5.8–14.8) months and 7.1 (5.6–10.2) and 7.0 (3.9–10.6) months, respectively. The most frequently reported adverse events were rash, fatigue and diarrhoea. Maintenance of panitumumab plus 5‐FU/LV after oxaliplatin discontinuation was well tolerated and may be an acceptable treatment paradigm for patients demonstrating a good response to first‐line treatment. Prospective studies are warranted.
Article
Full-text available
Purpose: Mutations in EGFR pathway genes are poor prognostic indicators in patients with metastatic colorectal cancer. Plasma analysis of cell-free DNA is a minimally invasive and highly sensitive method to detect somatic mutations in tumors. Experimental Design: Plasma samples collected from panitumumab-treated patients in the ASPECCT study at baseline and safety follow-up (SFU) were analyzed by a next-generation sequencing–based approach for extended mutant allele frequency as a continuous variable and their association with clinical outcomes and the mutational prevalence of 63 cancer-related genes. The correlation between patient outcome and baseline mutational status of EGFR pathway genes was also examined. Results: Overall, 261 patients in the panitumumab arm had evaluable plasma samples. Patients with a higher mutant allele frequency at baseline had worse clinical outcomes than those with a lower frequency (P < 0.001, Cox PH model); however, mutations did not necessarily preclude patients from deriving benefits. The objective response rate (complete or partial response) was 10.8% for patients with baseline mutations and 21.7% for those with BRAF mutations. The 63-gene panel analysis revealed an increase in tumor mutational burden from baseline to SFU (P < 0.001, Wilcoxon signed rank test). Baseline mutations in EGFR pathway genes, when analyzed both categorically and continuously, were associated with shorter survival. Conclusions: When mutations in EGFR pathway genes were analyzed continuously, higher mutant allele frequency correlated with poorer outcomes. However, extended mutation, by itself, did not preclude clinical responses to panitumumab in a monotherapy setting.
Article
Full-text available
Background: Colorectal cancer (CRC) has been shown to acquire RAS and EGFR ectodomain mutations as mechanisms of resistance to EGFR inhibition (anti-EGFR). After anti-EGFR withdrawal, RAS and EGFR mutant clones lack a growth advantage relative to other clones and decay, however the kinetics of decay remain unclear. We sought to determine the kinetics of acquired RAS/EGFR mutations after discontinuation of anti-EGFR therapy. Patients and methods: We present the post-progression circulating tumor DNA (ctDNA) profiles of 135 patients with RAS/BRAF wild-type metastatic CRC treated with anti-EGFR who acquired RAS and/or EGFR mutations during therapy. Our validation cohort consisted of an external dataset of 73 patients with a ctDNA profile suggestive of prior anti-EGFR exposure and serial sampling. A separate retrospective cohort of 80 patients was used to evaluate overall response rate and progression free survival during re-challenge therapies. Results: Our analysis showed that RAS and EGFR relative mutant allele frequency (rMAF) decays exponentially (r2=0.93 for RAS; r2=0.94 for EGFR) with a cumulative half-life of 4.4 months. We validated our findings using an external dataset of 73 patients with a ctDNA profile suggestive of prior anti-EGFR exposure and serial sampling, confirming exponential decay with an estimated half-life of 4.3 months. A separate retrospective cohort of 80 patients showed that patients had a higher overall response rate during re-challenge therapies after increasing time intervals, as predicted by our model. Conclusion: These results provide scientific support for anti-EGFR re-challenge and guide the optimal timing of re-challenge initiation.
Article
729 Background: FOLFOX therapy, an infusion of 5-fluorouracil (5-FU) with leucovorin in combination with oxaliplatin (OXA), is a common first-line chemotherapy regimen for unresectable, advanced or recurrent colorectal carcinoma (CRC). However, long-term administration of OXA is associated with peripheral neuropathy (PN); decreasing treatment length of OXA may be beneficial without reducing its efficacy. Methods: Chemotherapy-naïve pts aged ≥20 yrs with RAS wild-type advanced/recurrent CRC were enrolled to receive 6 cycles of panitumumab (Pmab) + mFOLFOX6 once every 2 wks. Pts who completed 6 cycles of Pmab + mFOLFOX6 and confirmed no progressive disease were subsequently randomized 1:1 to continue to receive Pmab + mFOLFOX6 (arm 1) or Pmab + 5-FU/LV (arm 2). The primary endpoint was progression-free survival (PFS) rate at 9 mos after randomization. The threshold PFS rate was defined as 30%, and the expected rate was set at 50%, with a 90% power and a 1-sided alpha value of 0.10. In the primary analysis, a binomial test was conducted separately for each arm. This study was designed as a phase II randomized screening comparison study which does not use direct comparison for the primary analysis. Results: Of 164 enrolled pts who received initial Pmab + mFOLFOX6 treatment, 56 were randomized to arm 1 and 57 to arm 2. PFS rates at 9 mos after randomization were significantly higher than the defined threshold at 44.6% (80% CI, 36.4–53.2) in arm 1 and 47.4% (39.1–55.8) in arm 2. Median PFS after randomization was 9.1 (8.6–11.2) and 9.3 (6.0–13.0) mos, respectively. Grade ≥2 PNs occurred in 6 (10.7%) and 1 (1.8%) pts in arms 1 and 2, respectively. Serious AEs occurred in 14 (25.0%) pts in arm 1 and in 9 (16.7%) pts in arm 2. Conclusions: The results of this trial suggest that Pmab + 5-FU/LV after 6 fixed-cycles of Pmab + mFOLFOX6 may be a treatment option in pts with RAS wild type chemotherapy-naïve advanced/recurrent CRC. Pts treated with Pmab + 5-FU/LV had a lower occurrence of grade ≥2 PNs compared with Pmab + mFOLFOX6. Clinical trial information: NCT02337946.
Article
Background: Despite advances in precision oncology and immunotherapy of tumors, little progress has been made in metastatic colorectal cancer (mCRC) in recent years. Therefore, making the most of available therapies is a necessity. Several studies, based on the pulsatile behavior of RAS clones under EGFR blockade, investigated whether readministration of EGFR-targeted agents is effective beyond second line. Methods: A systematic review of studies of retreatment with anti-EGFR monoclonal antibodies has been performed from January 2005 to December 2018 according to PRISMA criteria from PubMed, ESMO and ASCO meetings libraries and Clinicaltrial.gov. Efficacy has been evaluated as objective response rate and survival in available publications. In addition, type and incidence of side effects occurring during on anti-EGFR retreatment have been considered. Results: 26 publications have been retrieved, of which 20 full-text articles and 6 abstracts and categorized as for the retreatment strategy into five groups: rechallenge (n = 10), reintroduction (n = 4), sequence (n = 5), dose escalation (n = 1) and mixed (n = 6). Data of efficacy displayed high heterogeneity across different strategies (objective response rate, ORR = 0.0–53.8%; disease control rate, DCR = 24.0–89.7%), with best results in the setting of rechallenge (ORR = 2.9–53.8%; DCR = 40.0–89.7%). Conclusions: Rechallenge with anti-EGFR provides clinical benefit in molecularly selected mCRC patients beyond second line. Further ctDNA-guided studies comparing this option of treatment with current approved advanced line treatments are warranted.
Article
Importance Based on a small retrospective study, rechallenge with cetuximab-based therapy for patients with KRAS wild-type metastatic colorectal cancer (mCRC) who were previously treated with the same anti–epidermal growth factor receptor–based regimen might be efficacious. Recent data suggest the role of liquid biopsy as a tool to track molecular events in circulating tumor DNA (ctDNA). Objective To prospectively assess the activity of cetuximab plus irinotecan as third-line treatment for patients with RAS and BRAF wild-type mCRC who were initially sensitive to and then resistant to first-line irinotecan- and cetuximab-based therapy. Design, Setting, and Participants Multicenter phase 2 single-arm trial conducted from January 7, 2015, to June 19, 2017. Liquid biopsies for analysis of ctDNA were collected at baseline. Main eligibility criteria included RAS and BRAF wild-type status on tissue samples; prior first-line irinotecan- and cetuximab-based regimen with at least partial response, progression-free survival of at least 6 months with first-line therapy, and progression within 4 weeks after last dose of cetuximab; and prior second-line oxaliplatin- and bevacizumab-based treatment. Interventions Biweekly cetuximab, 500 mg/m², plus irinotecan, 180 mg/m². Main Outcomes and Measures Overall response rate according to the Response Evaluation Criteria in Solid Tumors, version 1.1. Secondary end points included progression-free survival and overall survival and, as an exploratory analysis, RAS mutations in ctDNA. Results Twenty-eight patients (9 women and 19 men; median age, 69 years [range, 45-79 years]) were enrolled. Six partial responses (4 confirmed) and 9 disease stabilizations were reported (response rate, 21%; 95% CI, 10%-40%; disease control rate, 54%; 95% CI, 36%-70%). Primary end point was met because lower limit of 95% CI of response rate was higher than 5%. RAS mutations were found in ctDNA collected at rechallenge baseline in 12 of 25 evaluable patients (48%). No RAS mutations were detected in samples from patients who achieved confirmed partial response. Patients with RAS wild-type ctDNA had significantly longer progression-free survival than those with RAS mutated ctDNA (median progression-free survival, 4.0 vs 1.9 months; hazard ratio, 0.44; 95% CI, 0.18-0.98; P = .03). Conclusions and Relevance This is the first prospective demonstration that a rechallenge strategy with cetuximab and irinotecan may be active in patients with RAS and BRAF wild-type mCRC with acquired resistance to first-line irinotecan- and cetuximab-based therapy. The evaluation of RAS mutational status on ctDNA might be helpful in selecting candidate patients. Trial Registration ClinicalTrials.gov Identifier: NCT02296203
Article
Background: Although colorectal cancer (CRC) is a disease of the older patients, older patients are under-represented from randomized trials. Herein we conducted a retrospective analysis for the effect of panitumumab in the management of older patients (≥65 years) patients with metastatic CRC (mCRC) in the Hellenic Oncology Research Group's (HORG) database. Methods: Τhe efficacy of panitumumab-based chemotherapy as front-line treatment in older patients with mCRC was assessed. Results: In total, 110 older patients with KRAS exon 2 wild type tumors were treated with chemotherapy plus panitumumab. The median age was 74 years; 69.9% of the patients were male, with left-sided primary tumors (78.2%), ECOG Performance Status 0-1 (95.4%) and median number of metastatic sites 2. Sixty-two (Overall Response Rate-ORR: 56.4%; 95% CI: 48.8%-68.1%) achieved an objective response, while 21 (19.1%) had stable disease. Median Progression free survival (PFS) was 9.4 months (95% CI: 7.8-11.0 months) and median Overall survival (OS) 23.0 months (95% CI: 20.6-25.3 months). Additionally, a statistically significant difference in ORR (62.7% vs. 33.3%; p = .014), median PFS (12.9 vs. 5.7 months; p = .001) and median OS (31.6 vs. 16.7 months; p < .001) was observed in patients with left-sided compared to right-sided primary tumor. There was no treatment-related death. Grade 3-4 toxicities were neutropenia (8.9%) and diarrhea (14.5%) whereas skin rash grade 2 or 3 was recorded in 41.1% and 10.7%, respectively. Conclusions: The results of this retrospective study provide the evidence that combination chemotherapy plus panitumumab is active and well tolerated in older patients with mCRC.
Article
11508 Background: Almost half of RAS and BRAFwt mCRC patients do not respond to anti-EGFRs. Different molecular alterations suggested as predictors of resistance have not been validated. Methods: We conducted a case-control study to prospectively demonstrate the negative predictive impact of HER-2 amplification or mutations (mut), MET amplification, NTRK/ROS1/ALK/RET rearrangements, and mut activating MAPKs or PI3K/Akt axis. Patients with RAS and BRAFwt mCRC clearly resistant (cases) or clearly sensitive (controls) to anti-EGFRs were selected. Hypothesizing a prevalence of candidate alterations of 0% and 15% among controls and cases, 47 cases and 47 controls were needed to be able to reject the null hypothesis of equally prevalent alterations, with a- and b- error 0.05 and 0.20. Since hypermutated tumors may hardly rely on a single pathway for their growth, we also evaluated the impact of microsatellite instability. Results: 47 cases and 47 controls were included. Primary endpoint was met: mentioned alterations were reported in 20 (42.6%) cases and 1 (2.1%) control (p<0.001). MSI-high was significantly more frequent among resistant than sensitive tumos (15% vs 0%, p<0.001). Conclusions: This is the first prospective demonstration that the combined assessment of these rare alterations allows to better select patients for anti-EGFRs, while opening the way to other tailored therapies. [Table: see text]