You are on page 1of 4

CCR 20th Anniversary Commentary

20th Anniversary Commentary

CCR 20th Anniversary Commentary: RAS as a


Biomarker for EGFR-Targeted Therapy for
Colorectal Cancer—From Concept to Practice
E. Ramsay Camp1,2,3 and Lee M. Ellis4,5

Clinical data support the use of EGFR mAbs in patients with mCRC with Ras-mutated tumors from therapy with EGFR mAbs
metastatic colorectal cancer (mCRC) with wild-type RAS status. has led to improved outcomes while sparing patients unnecessary
This notion, hypothesized in the review article by Camp, Ellis, and and potentially harmful therapy. Clin Cancer Res; 21(16); 3578–80.
colleagues in the January 1, 2005, issue of Clinical Cancer Research, 2015 AACR.
serves as an example of the successful application of basic science See related article by Camp et al., Clin Cancer Res 2005;11(1):
principles to clinical practice. The exclusion of patients with January 1, 2005;397–405

Introduction therapies. At this time, EGFR mutations in lung cancer were being
identified as markers of sensitivity to EGFR tyrosine kinase recep-
The therapeutic potential of mAbs targeting the EGFR was
tor inhibitors (2). Our laboratory had been interested in growth
established by promising preclinical studies and subsequent
factor receptors and downstream intracellular signaling, with a
early-phase clinical trials in patients with metastatic colorectal
focus on Src in collaboration with G. Gallick at The University of
cancer (mCRC; ref. 1). Ultimately, in 2004, the FDA approved the
Texas MD Anderson Cancer Center (Houston, TX). In reviewing
human–mouse chimeric EGFR mAb, cetuximab, for use in che-
the literature and applying our own knowledge of cell signaling,
motherapy-refractory patients with mCRC. Similarly, panitumu-
the general topics of this review in Clinical Cancer Research (CCR)
mab, a fully human EGFR mAb, was FDA approved for similar
focused on the following as resistance factors to EGFR-targeted
indications in 2006, and ultimately both drugs were subsequently
therapies: (i) the presence of redundant tyrosine kinase receptors;
approved in the first-line setting in combination with chemother-
(ii) increased angiogenic signaling; (iii) existence of specific EGFR
apy. However, the earliest clinical trials investigating EGFR mAbs
mutations; and (iv) constitutive activation of downstream med-
in mCRC demonstrated that these agents only provided modest
iators (3). This last proposed mechanism of resistance was not
benefit when used in combination with chemotherapy. Around
based on actual data, but a simple hypothesis: If an important
this time, a concerted effort to identify predictive markers
intracellular pathway is constitutively activated by a mutation,
for EGFR-targeted therapies was undertaken simultaneous to
then blocking signaling at the cell surface would be ineffective.
advances in genomic sequencing.
In our original schematic drawing describing this hypothesis,
As principal investigator of a laboratory, the senior author
we listed several activated downstream intermediates as potential
of this article (L.M. Ellis) learned early on in his career that the
resistance pathways. One, in particular, has demonstrated clinical
best way to educate a trainee on a topic was to have this individual
utility in selecting patients with mCRC for EGFR mAb therapy.
write a review article. This strategy exposes trainees to the entire
The therapeutic relevance of downstream RAS mutations on EGFR
field of the topic of interest, requiring a critical evaluation of the
mAb efficacy in patients with mCRC highlights the importance of
literature, and identifying knowledge gaps. In 2004, the first
understanding the molecular basis of malignant disease to
author of this article (E.R. Camp) entered the laboratory and was
improve personalized medicine. We recognize that this concept
given the task of writing a review on resistance to EGFR-targeted
is the basis of many preclinical and clinical studies, but at the time
of writing this review, there were no publications that had dis-
1
Department of Surgery, Medical University of South Carolina, Charles- cussed or hypothesized that mutated RAS or other constitutively
ton, South Carolina. 2Hollings Cancer Center, Medical University of activated signaling intermediates could serve as markers of resis-
South Carolina, Charleston, South Carolina. 3Ralph H. Johnson VA tance (or even detriment) in patients with mCRC. However, at
Medical Center, Charleston, South Carolina. 4Department of Surgical
Oncology, The University of Texas MD Anderson Cancer Center, that time, several studies were examining the role of mutated RAS
Houston, Texas. 5Department of Molecular and Cellular Oncology, The as a resistance marker for EGFR tyrosine kinase inhibitors in lung
University of Texas MD Anderson Cancer Center, Houston, Texas. cancer (4, 5); other investigators had hypothesized that RAS could
Corresponding Author: Lee M. Ellis, Department of Surgical Oncology, Unit be a resistance marker for EGFR-targeted therapies, and this work
1484, PO Box 301402, The University of Texas MD Anderson Cancer Center, 1515 occurred simultaneous to, or shortly after, our publication. Soon
Holcombe Boulevard, Houston, TX 77230. Phone: 713-792-6926; Fax: 713-792- after the publication of our review in January 2005, several studies
4689; E-mail: lellis@mdanderson.org
were examining the role of KRAS in EGFR mAb resistance, sug-
doi: 10.1158/1078-0432.CCR-14-2900 gesting that others had this idea and had already begun studies.
2015 American Association for Cancer Research. However, we have not identified any publication prior to ours in

3578 Clin Cancer Res; 21(16) August 15, 2015

Downloaded from clincancerres.aacrjournals.org on April 15, 2021. © 2015 American Association for Cancer Research.
RAS as a Biomarker for EGFR-Targeted Therapy

CCR addressing this topic. Of course, since then we have learned study to identify the negative predictive value of driver mutations,
that additional RAS mutations may render patients resistant to including NRAS, reported on a large retrospective cohort of
EGFR mAb therapy (6). chemotherapy-refractory patients with mCRC who were subse-
quently treated with irinotecan plus cetuximab (11). In this study,
assessment of data from patients with tumors harboring driver
Constitutive RAS Pathway Activation and mutations beyond KRAS identified even more patients unlikely to
Its Impact on EGFR mAb Therapy respond to EGFR mAbs, thus improving response rates in those
In 2006, a retrospective study of 30 patients with mCRC treated patients with tumors without these mutations. Subsequently, a
with cetuximab (mostly combined with chemotherapy) was the retrospective analysis of 1,060 patient tumor samples from the
first clinical study to correlate KRAS mutational status with randomized PRIME trial in patients with mCRC treated with first-
resistance to EGFR mAb therapy (7). In this small study, patients line FOLFOX4 with or without panitumumab highlighted the
with tumors harboring a KRAS mutation showed a 0% response significance of an expanded RAS assessment to predict response
rate. Overall survival (OS) in cetuximab-treated patients was for EGFR mAb-containing regimens (6). In this trial, 52% of the
significantly shorter in those with KRAS mutations in comparison analyzed tumors were identified as RAS mutated, including any
with patients with wild-type (WT) KRAS (6.9 vs. 16.3 months; P ¼ KRAS or NRAS mutations in exon 2, 3, or 4. Of the original 641
0.016). A subsequent investigation confirmed the clinical signif- patients categorized as having exon 2 WT KRAS tumors, an
icance of KRAS mutations as predictive markers for cetuximab or additional 108 (17%) had an alternative RAS mutation. Patients
panitumumab therapy for patients with mCRC (8). Furthermore, with tumors expressing any RAS mutation who received panitu-
this study was the first to demonstrate a causal relationship mumab with FOLFOX4 experienced a significantly decreased
between activating KRAS mutation (Gly12Val) and decreased in progression-free survival (PFS) as well as OS compared with
vitro response to cetuximab therapy in human colon cancer cell patients with WT RAS tumors. The survival outcomes were similar
lines. between exon 2 KRAS mutations and tumors with alternative RAS
A landmark retrospective analysis of a prospective randomized mutations, suggesting that any RAS mutation equally negates the
trial in 392 chemotherapy-refractory patients with mCRC com- therapeutic benefit achieved with EGFR mAbs. However, the
paring cetuximab treatment with best supportive care (BSC) numbers of patients with these less frequent RAS mutations limit
solidified the significance of KRAS mutational status in clinical the ability to firmly state that they are indeed markers of resistance.
practice (9). OS in patients with WT KRAS tumors was signifi- Similar to the findings from the OPUS trial, patients with any RAS
cantly longer in those treated with cetuximab than in those who mutation actually experienced a significantly decreased PFS and
received BSC (9.5 vs. 4.8 months; P < 0.001). In contrast, patients OS with the addition of panitumumab to FOLFOX4, reinforcing
with mutated KRAS tumors experienced an equivalent OS the concern that EGFR mAbs may actually be detrimental in this
between cetuximab therapy and BSC. A larger concern was that setting. Similarly, a recent meta-analysis of nine randomized,
results from the randomized phase II OPUS trial comparing first- controlled trials with EGFR mAbs for mCRC patients evaluating
line FOLFOX4 with or without cetuximab in patients with mCRC RAS mutational status further solidified the predictive impact of
suggested that the addition of an EGFR mAb might actually have a an expanded RAS mutational profile on PFS as well as OS (12). In
detrimental effect on outcomes in mCRC patients with mutated general, expanded RAS analysis may increase the percentage of
KRAS tumors (10) and other studies have confirmed these find- patients ineligible for EGFR mAb therapy by approximately 15%
ings (6). These initial translational investigations laid the foun- compared with KRAS exon 2 mutations alone. Based on the
dation for personalized molecular medicine in mCRC by dem- potential detrimental impact of any RAS mutation in patients
onstrating the predictive capability of KRAS mutational status for receiving EGFR mAb therapy, the FDA endorsed an extended RAS
improving patient selection and outcomes for EGFR mAb therapy. assessment for EGFR mAb therapy in 2013 and, subsequently, the
In response to the growing evidence supporting the predictive National Comprehensive Cancer Network (NCCN) published
implications of KRAS status for EGFR mAb therapy, the FDA clinical guidelines recommending prospective expanded RAS
restricted the use of cetuximab and panitumumab to patients with mutational assessment for all mCRC patients considered candi-
WT KRAS mCRC in 2009. dates for EGFR mAb therapy.
The failure of efficacy with EGFR mAbs in a statistically signi-
The Clinical Implication of Expanded RAS ficant number of patients with WT KRAS tumors fueled further
investigations evaluating alternative downstream mediators of
Status on Cetuximab Therapy EGFR. The first report to extend the molecular analysis beyond
The initial studies investigating the role of KRAS mutation RAS in patients with mCRC investigated the role of mutated
status on the efficacy of cetuximab in patients with mCRC focused BRAFv600e as a marker of cetuximab resistance in WT KRAS tumors
on the most prevalent KRAS mutations involving codons 12 and (13). BRAF is a downstream mediator in the RAS pathway, but, of
13 in exon 2, which are present in approximately 40% of patients course, can be constitutively activated as it is mutated in approx-
with mCRC. Even though multiple studies confirmed that exon 2 imately 5% to 10% of colorectal cancers. In this study, patients
mutated KRAS rendered patients with mCRC resistant to EGFR with tumors expressing the BRAFv600e mutation experienced a
mAb therapy, EGFR mAbs were found to be effective in some, but shorter PFS and OS in response to cetuximab compared with WT
not all, patients with KRAS WT tumors; this suggested that BRAF tumors. Although initial reports suggested that BRAF might
alternative molecular abnormalities might be influencing thera- be a predictive marker for EGFR mAbs, subsequent studies have
peutic response. Extending this line of investigation further, an not uniformly confirmed this association. For example, in the
expanded or "all-RAS" mutation analysis including all KRAS and retrospective analysis of the PRIME trial, patients with tumors
NRAS mutations has been proposed for identifying patients with expressing the BRAFv600e mutation showed an equally poor PFS
mCRC who would benefit from EGFR mAb therapy. The first and OS in response to FOLFOX4 alone compared with the

www.aacrjournals.org Clin Cancer Res; 21(16) August 15, 2015 3579

Downloaded from clincancerres.aacrjournals.org on April 15, 2021. © 2015 American Association for Cancer Research.
CCR 20th Anniversary Commentary

addition of panitumumab (6). In contrast, the randomized PIC- median PFS and OS of 11.4 and 32 months, respectively, com-
COLO trial investigating the role of irinotecan plus panitumumab pared with historical controls.
in fluorouracil-refractory patients with mCRC demonstrated a Taken together, the evidence is growing that mutations in RAS
significantly worse OS if patients' tumors harbored a BRAF muta- can predict for resistance to EGFR mAbs in patients with mCRC.
tion. Despite the inconsistent results regarding BRAF as an EGFR NCCN clinical guidelines, the FDA, and the European Medicines
mAb predictive marker, BRAF mutational status appears to have Agency mandate restricted use of EGFR mAbs for patients with
strong negative prognostic significance, as highlighted by results RAS WT mCRC. The foundation of this clinical practice change can
from several trials. Compared with patients with WT RAS/BRAF be traced back to the fundamentals learned from preclinical
tumors, BRAFv600e mutations conferred an approximately 50% studies of signaling pathways. This dramatic change in clinical
reduced PFS and OS in both arms of the trial. Given the aggressive practice highlights the continued need for adequate funding for
biology associated with BRAFv600e mutation, the actual impact on translational research to improve oncologic outcomes for our
the therapeutic efficacy of EGFR mAbs may be difficult to deter- patients.
mine. Similar to BRAF, PIK3CA mutations and loss of PTEN have
shown an inconsistent impact on the therapeutic efficacy of EGFR Disclosure of Potential Conflicts of Interest
mAbs. Currently, testing for BRAF and PIK3CA/PTEN is not L.M. Ellis is a consultant/advisory board member for Celgene, Eli Lilly, and
recommended for EGFR mAb mCRC patient selection, although Genentech/Roche. No potential conflicts of interest were disclosed by the other
these mutations/alterations may be beneficial in determining author.
prognosis and eligibility for clinical trials.
The improved objective response with the addition of EGFR Authors' Contributions
mAbs observed in "all-RAS" WT tumors highlights the potential of Conception and design: E.R. Camp, L.M. Ellis
Writing, review, and/or revision of the manuscript: E.R. Camp, L.M. Ellis
molecularly driven personalized cancer therapy. The role for
prospective RAS mutational analysis for cetuximab selection has
been explored in recent cooperative group clinical trials that
analyzed a comprehensive ("all-RAS") molecular profile to assess Grant Support
E.R. Camp was supported by NIH grant K08CA142904. L.M. Ellis was
the benefit of using cetuximab in combination with FOLFIRI or supported by NIH grant R01CA157880, Department of Defense grant
mFOLFOX6 (14). The CALGB/SWOG 80405 trial evaluated CA100879, and the William C. Liedtke Jr Chair in Cancer Research.
response to cetuximab-containing regimens in 526 "all-RAS" WT
colorectal cancer patients. In the "all-RAS" WT cohort, patients Received February 6, 2015; revised February 27, 2015; accepted February 28,
receiving cetuximab plus chemotherapy achieved an improved 2015; published online August 14, 2015.

References
1. Goldstein NI, Prewett M, Zuklys K, Rockwell P, Mendelsohn J. Biological 9. Karapetis CS, Khambata-Ford S, Jonker DJ, O'Callaghan CJ, Tu D, Tebbutt
efficacy of a chimeric antibody to the epidermal growth factor receptor in a NC, et al. K-ras mutations and benefit from cetuximab in advanced
human tumor xenograft model. Clin Cancer Res 1995;1:1311–8. colorectal cancer. N Engl J Med 2008;359:1757–65.
2. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan 10. Bokemeyer C, Bondarenko I, Makhson A, Hartmann JT, Aparicio J, de
BW, et al. Activating mutations in the epidermal growth factor receptor Braud F, et al. Fluorouracil, leucovorin, and oxaliplatin with and without
underlying responsiveness of non-small-cell lung cancer to gefitinib. cetuximab in the first-line treatment of metastatic colorectal cancer. J Clin
N Engl J Med 2004;350:2129–39. Oncol 2009;27:663–71.
3. Camp ER, Summy J, Bauer TW, Liu W, Gallick GE, Ellis LM. Molecular 11. De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas
mechanisms of resistance to therapies targeting the epidermal growth G, et al. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the
factor receptor. Clin Cancer Res 2005;11:397–405. efficacy of cetuximab plus chemotherapy in chemotherapy-refractory met-
4. Pao W, Wang TY, Riely GJ, Miller VA, Pan Q, Ladanyi M, et al. KRAS astatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol
mutations and primary resistance of lung adenocarcinomas to gefitinib or 2010;11:753–62.
erlotinib. PLoS Med 2005;2:e17. 12. Sorich MJ, Wiese MD, Rowland A, Kichenadasse G, McKinnon RA, Kar-
5. Eberhard DA, Johnson BE, Amler LC, Goddard AD, Heldens SL, Herbst RS, apetis CS. Extended RAS mutations and anti-EGFR monoclonal antibody
et al. Mutations in the epidermal growth factor receptor and in KRAS are survival benefit in metastatic colorectal cancer: a meta-analysis of random-
predictive and prognostic indicators in patients with non–small-cell lung ized, controlled trials. Ann Oncol 2015;26:13–21.
cancer treated with chemotherapy alone and in combination with erloti- 13. Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S,
nib. J Clin Oncol 2005;23:5900–9. Saletti P, et al. Wild-type BRAF is required for response to panitumumab
6. Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, et al. or cetuximab in metastatic colorectal cancer. J Clin Oncol 2008;26:
Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal can- 5705–12.
cer. N Engl J Med 2013;369:1023–34. 14. Lenz H, Niedzwiecki D, Innocenti F, Blanke C, Mahony MR, O'Neil BH,
7. Lievre A, Bachet JB, Le Corre D, Boige V, Landi B, Emile JF, et al. KRAS et al. CALGB/SWOG 80405: Phase III trial of irinotecan/5-FU/leucov-
mutation status is predictive of response to cetuximab therapy in colorectal orin (FOLFIRI) or oxaliplatin/5-FY/leucovorin (mFOLFOX6) with
cancer. Cancer Res 2006;66:3992–5. bevacizumab or cetuximab for patients with expanded ras analyses
8. Benvenuti S, Sartore-Bianchi A, Di Nicolantonio F, Zanon C, Moroni M, untreated metastatic adenocarcinoma of the colon or rectum [abstract].
Veronese S, et al. Oncogenic activation of the RAS/RAF signaling pathway In: Proceedings of the ESMO 2014 Congress, 2014 Sep 26–30, Madrid,
impairs the response of metastatic colorectal cancers to anti-epidermal Spain. Lugano (Switzerland): European Society for Medical Oncology;
growth factor receptor antibody therapies. Cancer Res 2007;67:2643–8. 2014. Abstract nr 501O.

3580 Clin Cancer Res; 21(16) August 15, 2015 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on April 15, 2021. © 2015 American Association for Cancer Research.
CCR 20th Anniversary Commentary: RAS as a Biomarker for
EGFR-Targeted Therapy for Colorectal Cancer−−From Concept to
Practice
E. Ramsay Camp and Lee M. Ellis

Clin Cancer Res 2015;21:3578-3580.

Updated version Access the most recent version of this article at:
http://clincancerres.aacrjournals.org/content/21/16/3578

Cited articles This article cites 13 articles, 7 of which you can access for free at:
http://clincancerres.aacrjournals.org/content/21/16/3578.full#ref-list-1

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at
Subscriptions pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://clincancerres.aacrjournals.org/content/21/16/3578.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)
Rightslink site.

Downloaded from clincancerres.aacrjournals.org on April 15, 2021. © 2015 American Association for Cancer Research.

You might also like