Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Aug 14, 2014; 20(30): 10288-10295
Published online Aug 14, 2014. doi: 10.3748/wjg.v20.i30.10288
Rationally designed treatment for metastatic colorectal cancer: Current drug development strategies
Pavlina Spiliopoulou, Hendrik-Tobias Arkenau, Drug Development Unit, Sarah Cannon Research Institute United Kingdom, London W1G 6AD, United Kingdom
Hendrik-Tobias Arkenau, UCL Cancer Institute, Medical Oncology, University College London, London WC1E 6BT, United Kingdom
Author contributions: Both authors contributed to the design of the manuscript.
Correspondence to: Hendrik-Tobias Arkenau, MD, PhD, Director, Drug Development Unit, Sarah Cannon Research Institute United Kingdom, 93 Harley Street, London W1G 6AD, United Kingdom. tobias.arkenau@sarahcannonresearch.co.uk
Telephone: +44-203-2195200 Fax: +44-203-2195239
Received: December 16, 2013
Revised: February 28, 2014
Accepted: April 15, 2014
Published online: August 14, 2014

Abstract

The therapeutic landscape of metastatic colorectal cancer (mCRC) has changed substantially with the emergence of new molecularly targeted agents (MTA) used as single agents or alongside standard chemotherapy. The use of these MTAs extended the overall survival of patients with mCRC to a level that current chemotherapeutics alone could not achieve. In addition, improvement in surgical techniques and ablation modalities offer cure to a limited subset of patients with mCRC and MTAs have been found to have a significant role here too, as they aid resectability. However, for the majority of patients, mCRC remains an invariably incurable disease necessitating continued courses of combined treatment modalities. During the course of these treatments, either cytotoxic or biological, cancer cells maintain their ability to acquire mitogenic mutations which render them resistant to treatment. Key challenges remain to identify appropriate subsets of patients who will most likely benefit from these new MTAs and effectively select these based on validated biomarkers. Moreover, better knowledge of the biology of colorectal cancer and the mechanisms via which it bypasses blockade of known signalling pathways will help us design better and more rational sequencing of these treatments, so that we can maximise the survivorship of mCRC patients. This review outlines treatment strategies for known molecular alterations with new MTAs and highlights some promising strategies.

Key Words: Colorectal cancer, Molecularly targeted therapies, Drug development

Core tip: Abrogation of the mitogen-activated protein kinase pathway downstream epidermal growth factor receptor (EGFR) has emerged as a new potential targetable pathway in the treatment of metastatic colorectal cancer. Similarly, a variety of agents blocking the PI3K/Akt/mTOR pathway are underway. At the same time, a combinatorial inhibition of angiogenesis is being attempted with dual blockade of vascular-endothelial growth factor and c-mesenchymal-epithelial transition factor. Indications that HER-2 overactivation can confer resistance to treatment to MoAb against EGFR has revealed yet another potential target whereas PARP inhibitors are being tested for their ability to cause synthetic lethality in cancer cells with established defects in MMR system.



INTRODUCTION

Colorectal cancer remains the third most common diagnosed cancer and the third leading cause of cancer death in the United States with 142820 estimated new cases in 2013[1].

The combinations of fluoropyrimidine with oxaliplatin (FOLFOX) or irinotecan (FOLFIRI) are all appropriate first-line treatments for metastatic colorectal cancer (mCRC) with the crossover to the alternate regimen upon progression[2].

The addition of bevacizumab, a humanized recombinant monoclonal antibody (moAb) which blocks the vascular-endothelial growth factor-A (VEGF-A or VEGF) to first line chemotherapy with either FOLFOX or XELOX (capecitabine, oxaliplatin) resulted in a better progression free survival (PFS) compared to chemotherapy alone[3]. There is also now evidence that the use of bevacizumab beyond first-line chemotherapy progression is increasing overall survival (OS) when added to second-line chemotherapy[4].

Similarly, the MoAb against the epidermal growth factor receptor (EGFR) cetuximab is approved for use in the first-line setting for KRAS wild-type (wt) mCRC combined with FOLFIRI, based on results of the CRYSTAL study[5]. Panitumumab, a fully humanised EGFR-antibody, showed superior PFS and overall survival (OS) compared with FOLFOX alone in the first-line setting[6]. The use of EGFR antibodies is restricted to patients without an activating mutation in exon 2 of KRAS gene (KRAS wt) which confers intrinsic resistance of cancer cells to EGFR blockade[7].

APPROVED NEW MOLECULARLY TARGETED AGENTS

Based on principles set by EGFR and VEGF inhibition, new targeted agents that abrogate multiple receptors emerged, in expectation of achieving greater results than their predecessors. And yet, their clinical effect can be characterised as modest. Questions arise as to whether these agents would exhibit their maximal effect only when used in selected patient subsets based on relevant biomarker.

Aflibercept is a recombinant fusion protein (VEGR-trap) that blocks the activity of VEGF-A, VEGF-B and placental growth factor (PlGF). In the VELOUR study, aflibercept was combined with FOLFIRI vs FOLFIRI/placebo in patients with mCRC patients who relapsed after treatment with oxaliplatin-containing regimen. Compared to the control group, it produced better PFS (6.9 mo vs 4.67 mo, HR = 0.758; P < 0.0001) and an overall response rate of 19.8% compared to 11.1% (P = 0.0001)[8].

Another new MTA is regorafenib, an inhibitor of several angiogenic and stromal receptor tyrosine kinases, including VEGFR-1, -2, -3, platelet-derived growth factor-β (PDGFR-β), fibroblast growth factor receptor 1 (FGFR-1), and TIE2. In addition, regorafenib inhibits various oncogenic receptor tyrosine kinases (RTKs:c-KIT and RET) and intracellular signaling kinases (cRAF/RAF-1, B-RAF, and B-RAFV600E mt)[9]. The multicentre CORRECT phase-3 study demonstrated a 23% risk reduction of death (HR = 0.77) in mCRC patients who progressed through standard therapeutic options compared to best supportive care[10]. Undoubtedly, this study yielded a new agent for patients with no remaining therapeutic options, but clinical benefit vs increased toxicity has to be considered (Table 1).

Table 1 Completed trials of approved targeted treatments.
DrugTargetStudyPhase patientsOutcome
BevacizumabVEGF-ABevacizumab in combination with Oxaliplatin-based chemotherapy as first-line therapy in metastatic CRC.Phase IIIMedian OS 21.3 mo Bev/chemo arm vs 19.9 mo chemo alone
n = 1401(HR = 0.89, 97.5%CI: 0.76-1.03, P = 0.077)
CetuximabEGFRCetuximab combined with FOLFIRI vs FOLFIRI alone as first line in mCRC.Phase III n = 599Median OS for KRAS WT patients: 23.5 mo (97.5%CI: 21.2-26.3) in Cetux/FOLFIRI arm vs 20 mo (97.5%CI: 17.4-21.7) in FOLFIRI alone
PanitumumabEGFRPanitumumab with FOLFOX vs FOLFOX alone in first line aCRCPhase III n = 656Median OS (KRAS wt) 23.8 mo (97.5%CI: 20.0–27.7) with panitum/FOLFOX vs 19.4 mo (97.5%CI: 17.4-22.6) with FOLFOX alone
Ziv-afliberceptVEGFAAddition of Aflibercept to FOLFIRI in patient with mCRC previously treated with Oxaliplatin-based chemotherapy (including patients who received Bevacizumab)Phase IIIMedian OS 13.5 mo (97.5%CI: 12.5-14.9) with Aflibercept/FOLFIRI vs 12 mo (97.5%CI: 11-13) with FOLFIRI alone
VEGFBn = 1226
PIGF
RegorafenibVEGFR -1, -2, -3Regorafenib monotherapy for previously treated metastatic colorectal cancer vs best supportive care (CORRECT)Phase IIIMedian OS 6.5 mo with Regorafenib vs 5 mo for best supportive care (HR = 0.77, 97.5%CI: 0.64-0.94, P = 0.0052)
TIE2n = 760
KIT, RET, RAF1, BRAF, and BRAFV600E
PDGFR and FGFR
NEW RATIONALLY DESIGNED TREATMENT MODALITIES

Two major pathways, the mitogen-activated protein kinase (MAPK-kinase) and the PI3K/Akt/mTOR pathways are commonly deregulated in mCRC (i.e., 40% KRAS mutant, 14.5% PIK3CA mutant and 4.7% BRAF mutant[11]) and offer a good clinical rationale to target these alterations. These pathways interact with a variety of receptor tyrosine kinases (RTK), including EGFR, HER-2, hepatocyte growth factor (HGF) and others.

MAPK pathway

MAPK pathway regulates various cellular functions including cell proliferation, differentiation, migration and apoptosis. With recent successes of selected BRAF and MEK inhibitors in metastatic cutaneous melanoma these drugs have also been explored in v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) mutant mCRC.

Vemurafenib, an oral selective inhibitor of BRAF oncoprotein was tested as single agent in a phase 1 study in patients with BRAF mutant colorectal cancer. In contrast to its pronounced activity in melanoma patients, the results of this study were comparably modest, with only 5% rate of partial responses[12]. Prahallad et al[13] investigated reasons for the limited single agent activity and revealed that BRAF inhibition leads to rapid upstream activation of EGFR with subsequent bypass signalling via the PI3K/Akt/mTOR pathway. Interestingly, preclinical BRAF mutant CRC models showed that combined BRAF/EGFR inhibition resulted in promising anti-proliferate effects and this approach is currently tested in a study, whereby vemurafenib is combined with panitumumab[14] (Table 2).

Table 2 On-going clinical trials with novel targeted therapeutics.
DrugTargetStudyPhaseObjective
Patients (est.)(primary, secondary)
Vemurafenib plusBRAF/EGFRA Pilot Study of Vemurafenib and Panitumumab Combination Therapy in Patients With BRAF V600E Mutated Metastatic Colorectal CancerI/IIORR
Panitumumabn = 15PFS/OS
Vemurafenib, Cetuximab plusBRAF/EGFR plus chemotherapyA Phase I Trial of Vemurafenib in Combination With Cetuximab and Irinotecan in Patients With BRAF V600 Mutant Advanced Solid MalignanciesIMTD
Irinotecann = 63
Dabrafenib, Trametinib panitumumabBRAF/MEK and EGFRAn Open-Label, Three-Part, Phase I/II Study to Investigate the Safety, Pharmacokinetics, Pharmacodynamics, and Clinical Activity of the MEK Inhibitor GSK1120212, BRAF Inhibitor GSK2118436 and the Anti-EGFR antibody Panitumumab in Combination in Subjects With BRAF-mutation V600E or V600K Positive Colorectal CancerI/IIDLTs
n = 200Pharmacokinenics
RR
LGX818BRAFA phase I, multicentre, open label, dose-escalation study of oral LGX818 in adult patients advanced colorectal cancer BRAF mutatedIDLT
n = 126Tumour response
LGX818BRAFA Phase Ib/II Multi-center, Open-label, Dose Escalation Study of LGX818 and Cetuximab or LGX818, BYL719, and Cetuximab in Patients With BRAF Mutant Metastatic Colorectal CancerI/IIDLT
BYL719PI3Kn = 124PFS
CetuximabEGFR
CEP-32496BRAFAn open-Label, Phase 1/2, Single-Agent Study of CEP-32496 in Patients With Advanced Solid Tumors in Phase 1 and in Patients With Advanced Melanoma and Metastatic Colorectal Cancer With BRAF Mutation in Phase 2I/IIn = 154ORRPFS
BKM120PI3KPhase I Trial of Irinotecan and BKM120 in Previously Treated Advanced Colorectal CancerIMTD
Irinotecanplus chemotherapyn = 30Pharmacokinetics
BKM120PI3KPhase I/II Study of the P13Kinase Inhibitor BKM120 Given in Combination With Panitumumab in Patients With Metastatic or Advanced RAS-Wild Type Colorectal CancerI/IIMTD
n = 40Antitumour activity
PanitumumabEGFRTranslational research
GSK- 2636771PI3K-betaA Phase I/IIa, First Time in Human, Open-label Dose-escalation Study of GSK2636771 in Subjects With Advanced Solid Tumors With PTEN DeficiencyI/IIMTD
n = 150Pharmacokinetics
Efficacy
BKM120PI3KA Phase Ib, Open-label, Multi-center, Dose-escalation and Expansion Study of an Orally Administered Combination of BKM120 Plus MEK162 in Adult Patients With Selected Advanced Solid TumorsIRate of DLTs
MEK162MEKn = 88ORR and PFS
MK-2206AktA Phase 2 Study of MK-2206 in Previously Treated Metastatic Colorectal Cancer Patients Enriched for PTEN Loss and PIK3CA MutationIIORR
n = 54Biomarker validation
MK-2206AktPilot Study of the Combination of MK-2206, an AKT Inhibitor, and AZD6244, a MEK Inhibitor, in Patients With Advanced Colorectal CarcinomaIIEvaluate reduction of pAKT, pERK and Ki-67
SelumetinibMEKn = 38
EverolimusmTORPhase I/Randomized Phase II Study of Second Line Therapy With Irinotecan and Cetuximab With or Without RAD001, an Oral mTOR Inhibitor for Patients With Metastatic Colorectal CancerI/IIMTD
IrinotecanEGFRn = 41ORR
Cetuximabplus chemotherapy
SAR245408 (XL147)mTORA Phase 1 Dose Escalation Study of Combination Therapy With Oral SAR245408 (XL147) and Oral MSC1936369B in Patients With Locally Advanced or Metastatic Solid TumorsIMTD
MSC 1936369BMEKn = 170Pharmacokinetics
Onartuzumabc-METRandomized, Double-Blind, Phase II Study of FOLFOX/Bevacizumab With Onartuzumab (MetMAb) vs Placebo as First-Line Treatment for Patients With Metastatic Colorectal CaIIPFS
BevacizumabVEGF-An = 196Response rate
FOLFOXplus chemotherapy
TrastuzumabHER2A Phase II multi-center 2-sequential cohorts trial, designed to assess the objective response rate of the anti HER2 monoclonal antibody trastuzumab, used in combination with either the small molecule tyrosine kinase inhibitor lapatinib (Cohort A) or the monoclonal antibody pertuzumab (Cohort B), in advanced disease CRC patients harbouring an amplified HER2 tumorIIORR
PertuzumabEGFRn = 54PFS
Lapatinib
OlaparibPARPPhase II, Open-Label, Multicenter Trial to Assess the Efficacy and Safety of the PARP Inhibitor, Olaparib, Alone in Previously-Treated Patients With Stage IV, Measurable Colorectal Cancer, Stratified by MSI StatusIITumour response
n = 33
ABT-888PARPA Phase I Study of ABT-888 in Combination With Oxaliplatin and Capecitabine in Advanced Solid TumorsIMTD/DLTs
Oxaliplatinn = 36Pharmacokinetics
CapecitabineTumour response
Brivanib AlaninateVEGFRA phase II Study of second-line irinotecan plus brivanib, a dual tyrosine inhibitor of VEGFR and FGFR, in metastatic colorectal cancer patients enriched for elevated levels of plasma FGF following progression on bevacizumab-based treatmentIIPFS
IrinotecanFGFR plus chemotherapyn = 60

Another approach to overcome the limited response to single agent BRAF inhibitors was the addition of MEK inhibitors which resulted in synergistic anti-tumour effect in BRAF mutant CRC cell lines[15]. The rationale of this approach was that the addition of a MEK inhibitor reduces signalling via RAF-isoforms, especially CRAF, via MEK. A dual BRAF/MEK combination was clinically tested with the BRAF inhibitor, dabrafenib, in combination with the MEK inhibitor, trametinib. Despite a combined response rate of only 12% (3%CR and 9%PR), there was a significant rate of disease stabilisation (53%) with a median PFS of 3.5 mo (95%CI: 1.8-4.9)[16], justifying to test this combination in later phase trials.

Similarly to the EGFR upregulation that is observed when BRAF is inhibited, abrogation of MEK activity with a MEK inhibitor stimulates AKT activity in an EGFR-dependent way[17]. Therefore, a combinatorial approach is currently underway whereby the BRAF/MEK inhibitor combination of dabrafenib/trametinib is compared to dabrafenib/trametinib plus panitumumab in an ongoing phase 2 study[18] (Table 2).

Interestingly, MEK inhibitors have not displayed promising activity as single agents. However, pre-clinical data showed that the MEK inhibitor selumetinib (AZD6244) can augment the efficacy of conventional chemotherapeutic agents such as temozolamide or docetaxel[19]. Early results from a small phase 2 study where reported when selumetinib was combined with irinotecan in patients with KRAS and/or BRAF mutant mCRC who progressed on oxaliplatin-containing regimen. Of a total of 32 patients, there was a 10% PR rate and disease stabilisation in 52% of patients. Interestingly, the median PFS was 3.4 mo compared with the historic PFS of 2.5 mo in second line FOLFIRI[4,20].

PI3K - AKT- mTOR pathway

The PI3K-Akt-mTOR pathway deregulation is often a result of alterations in the PIK3CA gene or loss of function/expression of the tumour suppressor gene PTEN, thereby promoting cancer cell survival and motility. As reported by Perrone et al[21], PIK3CA/PTEN deregulation can be found in up to 27% of patients not responding to classical epidermal growth factor receptor blockade with cetuximab.

BKM120 is a pan-PI3K inhibitor with activity against all PI3K isoforms and was tested in a classical dose-escalation phase 1 study of which 43% were patients with mCRC - promisingly, 33% of the mCRC cancer patients had stable disease with good tolerability[22]. This subsequently led to the initiation of a phase-1 b study in combination with irinotecan which is currently enrolling mCRC patients[23] (Table 2).

Loss of PTEN function has been reported in up to 31% of colon cancer[24] and specific isoform inhibition of the PI3K-beta subunit has been reported to prohibit proliferation in tumours with PTEN loss[25]. As part of an ongoing phase-1 study the PI3K-beta inhibitor, GSK2636711, is currently tested in patients with advanced tumours with PTEN loss - including patients with mCRC[26] (Table 2).

The multi-tyrosine kinase inhibitor, perifosine, exhibits its main action by inhibiting signalling via Akt[27]. Despite promising results of perifosine in a phase 2 study in combination with capecitabine a large phase 3 study (X-PECT study) failed to show overall survival benefit in unselected mCRC patients who failed standard therapies[28]. Further trials of targeting Akt-signalling are currently underway and one of the biological rational is to combine both, Akt and MEK-inhibitors, in an attempt to horizontally target both the PI3K/Akt/mTOR and the MAP-kinase axis (Table 2).

A pharmacokinetic, biomarker-driven study using the combination of the AKT-inhibitor (MK-2206) with selumetinib (MEK-inhibitor) is currently recruiting, however early results indicate that the combination is not well tolerated mandating several dose reductions and leading to limited drug exposure in target tissue[29]. Further results will clarify whether the drug combination can enter later stage trials.

Everolimus is an oral derivative of rapamycin which inhibits the mTOR1 complex, one of the major effectors of Akt-signalling. In a phase 2 study of heavily pre-treated mCRC patients, everolimus failed to produce a meaningful PFS and OS benefit[30]. In addition a combination of everolimus and bevacizumab resulted in modest clinical benefit and clinical programmes were stopped[31].

HGF and the receptor tyrosine kinase mesenchymal-epithelial transition factor

The receptor tyrosine kinase mesenchymal-epithelial transition factor mesenchymal-epithelial transition factor (c-MET) and its ligand HGF are currently investigated as potential active targets in mCRC. Activation of the axis through c-MET, triggers downstream effectors that promote mitogenesis and growth via the RAS/MAPK pathway and prevent apoptosis through activating the PI3K/AKT signalling route[32]. When colorectal tumours were examined in various stages including adenomas, carcinomas and liver metastases, overexpression of the MET oncogene was detected in more than 50% of each stage[33].

In preclinical studies, c-MET expressing cancer cell lines that were exposed to the small molecule ARQ-197 (tivantinib) showed inhibition of their proliferation, as well as induction of caspase-dependent apoptosis[34]. Tivantinib is a selective non-ATP competitive inhibitor of the c-MET receptor kinase and it was further investigated in combination with irinotecan and cetuximab in patients with KRAS wt mCRC. Although early results of a small phase 1/2 study demonstrated disease control in 8 out of 9 patients[35], a recently reported phase 2 study could not confirm these initial promising results (PFS: 8.3 mo vs 7.3 mo, HR = 0.85, 95%CI: 0.55-1.33, stratified log-rank P = 0.38)[36]. Whether the non-significant 1 mo PFS benefit will change in selected subgroups, i.e., high c-met expression is currently being investigated.

Cross-talk between c-MET and VEGF pathways led to the hypothesis that combined abrogation can inhibit tumour growth. c-MET promotes tumour vasculature formation through VEGF and conversely, VEGF results in HGF production which in turn activates c-MET. Onartuzumab (MetMab), a novel “one-armed” monovalent antibody which prevents antibody-induced dimerization of c-MET was tested in combination with bevacizumab in a phase 1 study and dual-agent safety was confirmed. A randomized, phase 2, placebo-controlled study evaluating onartuzumab in combination with mFOLFOX6 plus bevacizumab vs mFOLFOX6/bevacizumab/placebo in patients with mCRC is underway and results are awaited[37] (Table 2).

HER-2 (ERBB2) signalling pathway

The HER (ErbB) family of receptors consists of EGFR, HER2 (ErbB-2), HER3 (ErbB-3) and HER4 (ErbB-4). They are the main transmembrane mediators for cell proliferation and survival via the activation of the MAP-kinase and PI3K/Akt/mTOR pathways[38]. Between these receptors there is a fine interaction upon ligand binding including homodimerisation or heterodimerisation, whereby EGFR is a preferred heterodimerisation partner of HER2.

Preclinical studies showed that aberrant activation of HER2 receptor can confer resistance of colorectal cancer cells to EGFR MoAbs, either via HER2 amplification or excessive production of the ligand heregulin[39]. Surgical specimens from patients who did not harbour KRAS/NRAS/BRAF/PIK3CA mutations but overexpressed the HER2 receptor, showed substantial tumour response when they incurred combined EGFR and HER2 inhibition[40].

In this context, the HERACLES phase-2 study investigates the role of either MoAbs trastuzumab/pertuzumab or trastuzumab/lapatinib in patients with HER2 amplified mCRC[41] (Table 2).

Lapatinib is an oral dual EGFR and HER-2 tyrosine kinase inhibitor which is already known to act synergistically with capecitabine in breast cancer. In a small phase-2 study of 29 patients with heavily pretreated mCRC, the combination produced a non-significant median PFS of 2.1 mo[42]. Notably, the patients in this study were not stratified by KRAS status which might have masked potential activity of lapatinib/capecitabine on KRAS wt mCRC patients.

Microsatellite instability

Microsatellite instability (MSI) produces a phenotype which characterises hereditary colorectal cancer, but can also be found sporadically in approximately 4% of patients with mCRC; it is the consequence of a defective mismatch repair system of proteins (MLH1, MSH2, MSH6 and PMS2)[43,44]. Recent studies suggested that the sporadic MSI occurs as a consequence of CpG island methylator phenotype (CIMP) in which high rates of MLH1 DNA is hypermethylated. Interestingly, in this context, there is a strong correlation with BRAF mutations[45]. The role of demethylating agents is currently under clinical investigation and it could be of interest in selected subgroups, i.e., CIMP.

In addition, there is mounting evidence that MSI tumours are associated with a high rate of deficiency of homologous recombination due to mutations in coding microsatellites suggesting that synthetic lethality can be achieved by using Poly-(ADP-ribose) Polymerase (PARP) inhibitors[46]. In preclinical models increased cytotoxicity could be achieved with the PARP inhibitor (ABT888) in MSI cell lines with mutant copies of MRE11A compared to wild type MSI[47]. Results of clinical trials of single agent PARP inhibitors (e.g., Olaparib) or in combination with chemotherapy in MSI mCRC patients are awaited (Table 2).

FGFR plus VEGFR inhibition

Several data suggest that resistance to VEGFR inhibition can be promoted through hypoxia-mediated activation of other proangiogenic factors, independent of VEGF. Some of them are members of FGF family[48]. This can re-activate tumour revascularisation and growth despite VEGF blockade.

Dual inhibition of the fibroblast and vascular endothelial growth factors can be achieved with the oral small molecule tyrosine kinase inhibitor, brivanib alaninate. Brivanib alaninate produced promising phase-1 results in patients with advanced gastrointestinal malignancies with a median PFS in the range of 5 mo for responders. It progressed to phase-3 level combined with cetuximab vs cetuximab alone in KRAS-wild type patients with mCRC, but it failed to meet the end point of overall survival. The investigational arm had an OS of 8.8 mo vs 8.1 mo for cetuximab alone[49].

CONCLUSION

The integration of antibodies against EGFR and VEGF into clinical practice raised hopes that a new era of rationally designed treatments has started. The lessons learnt from the ‘‘one-size-fits-all’’ approach of EGFR-antibody therapies to selected subgroups (KRASwt) were tremendously helpful in understanding CRC tumour biology and led for example to identification of several molecular alterations in the MAP-kinase and PI3K-Akt-mTOR pathways. Moreover, further knowledge in particular on crosstalk between pathways, mechanisms of resistance and new targets for drug development have emerged.

Despite the recent progress, interpretation of current preclinical and clinical data is still limited by retrospective analyses, single centre experience, small sample size and lack of standardisation of diagnostic tools. As witnessed in this review several new MTAs in development were tested in unselected patient populations increasing the risk of failure. New trial designs focusing on biomarkers in selected subgroups will be essential not only for better understanding of the mechanisms of action, but also to make confident ‘‘go or no-go decisions’’ in the drug development process.

In this context a new “taxonomy-based” model for biomarker-driven drug development in mCRC has been suggested, whereby pathological and genomic classification of patient’s tumour takes place upfront, before initiation of treatment. This so-called multi-“omic” profiling, testing for multiple biomarkers instead of one, could help in creating a stratification model for colorectal cancer, in a similar way as breast or lung cancer has been stratified. A defined molecular “signature”, could be used for multiple therapeutic interventions or clinical trials.

Footnotes

P- Reviewer: Gagliardi G, Inoue M, Leggett BA S- Editor: Zhai HH L- Editor: A E- Editor: Ma S

References
1.  American Cancer Society Cancer stats and figures 2013. Accessed on 1st August. 2013; Available from: http://www.cancer.org/research/cancerfactsstatistics/cancerfactsfigures2013/index.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Tournigand C, André T, Achille E, Lledo G, Flesh M, Mery-Mignard D, Quinaux E, Couteau C, Buyse M, Ganem G. FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorectal cancer: a randomized GERCOR study. J Clin Oncol. 2004;22:229-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2181]  [Cited by in F6Publishing: 2147]  [Article Influence: 102.2]  [Reference Citation Analysis (1)]
3.  Saltz LB, Clarke S, Díaz-Rubio E, Scheithauer W, Figer A, Wong R, Koski S, Lichinitser M, Yang TS, Rivera F. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol. 2008;26:2013-2019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2147]  [Cited by in F6Publishing: 2218]  [Article Influence: 138.6]  [Reference Citation Analysis (0)]
4.  Bennouna J, Sastre J, Arnold D, Österlund P, Greil R, Van Cutsem E, von Moos R, Viéitez JM, Bouché O, Borg C. Continuation of bevacizumab after first progression in metastatic colorectal cancer (ML18147): a randomised phase 3 trial. Lancet Oncol. 2013;14:29-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 760]  [Cited by in F6Publishing: 827]  [Article Influence: 68.9]  [Reference Citation Analysis (0)]
5.  Van Cutsem E, Köhne CH, Hitre E, Zaluski J, Chang Chien CR, Makhson A, D’Haens G, Pintér T, Lim R, Bodoky G. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 2009;360:1408-1417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2901]  [Cited by in F6Publishing: 3021]  [Article Influence: 201.4]  [Reference Citation Analysis (1)]
6.  Douillard JY, Siena S, Tabernero J, Burkes RL, Barugel ME, Humblet Y, Sidhu R. Overall survival (OS) analysis from PRIME: Randomized phase III study of panitumumab (pmab) with FOLFOX4 for first-line metastatic colorectal cancer (mCRC). ASCO Annual Meeting. 2013;31:3620.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Lièvre A, Bachet JB, Le Corre D, Boige V, Landi B, Emile JF, Côté JF, Tomasic G, Penna C, Ducreux M. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 2006;66:3992-3995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1669]  [Cited by in F6Publishing: 1653]  [Article Influence: 91.8]  [Reference Citation Analysis (0)]
8.  Van Cutsem E, Tabernero J, Lakomy R, Prenen H, Prausová J, Macarulla T, Ruff P, van Hazel GA, Moiseyenko V, Ferry D. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol. 2012;30:3499-3506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 933]  [Cited by in F6Publishing: 954]  [Article Influence: 79.5]  [Reference Citation Analysis (0)]
9.  Wilhelm SM, Dumas J, Adnane L, Lynch M, Carter CA, Schütz G, Thierauch KH, Zopf D. Regorafenib (BAY 73-4506): a new oral multikinase inhibitor of angiogenic, stromal and oncogenic receptor tyrosine kinases with potent preclinical antitumor activity. Int J Cancer. 2011;129:245-255.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Grothey A, Van Cutsem E, Sobrero A, Siena S, Falcone A, Ychou M, Humblet Y, Bouché O, Mineur L, Barone C. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet. 2013;381:303-312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1808]  [Cited by in F6Publishing: 1899]  [Article Influence: 172.6]  [Reference Citation Analysis (0)]
11.  De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, Kalogeras KT, Kotoula V, Papamichael D, Laurent-Puig P. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol. 2010;11:753-762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1491]  [Cited by in F6Publishing: 1585]  [Article Influence: 113.2]  [Reference Citation Analysis (1)]
12.  Kopetz S, Desai J, Chan E, Hecht JR, O’Dwyer PJ, Lee RJ, Nolop KB, Saltz L. PLX4032 in metastatic colorectal cancer patients with mutant BRAF tumors. J Clin Oncol. 2010;28:3534.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, Beijersbergen RL, Bardelli A, Bernards R. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature. 2012;483:100-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1390]  [Cited by in F6Publishing: 1456]  [Article Influence: 121.3]  [Reference Citation Analysis (0)]
14.  Vemurafenib and Panitumumab Combination Therapy in Patients With BRAF V600E Mutated Metastatic Colorectal Cancer. Clinicaltrials.gov, accessed on 25th Aug. 2013; Available from: http://clinicaltrials.gov/ct2/results?term=vemurafenib,colorectal&Search=Search.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Higgins B, Kolinsky KD, Yangetal H. Efficacy of vemurafenib, a selective BRAFV600E inhibitor, in combination with a MEK inhibitor in BRAFV600E colorectal cancer models. J Clin Oncol. 2012;30:abstr 488.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Corcoran RB, Falchook GS, Infante JR. Pharmacodynamic and efficacy analysis of the BRAF inhibitor dabrafenib (GSK436) in combination with the MEK inhibitor trametinib (GSK212) in patients with BRAFV600 mutant colorectal cancer (CRC). J Clin Oncol. 2013;31:abstr 3507.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Klinger B, Sieber A, Fritsche-Guenther R, Witzel F, Berry L, Schumacher D, Yan Y, Durek P, Merchant M, Schäfer R. Network quantification of EGFR signaling unveils potential for targeted combination therapy. Mol Syst Biol. 2013;9:673.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Klinger B; Clinicaltrials. gov. BRAF/MEK/EGFR Inhibitor Combination Study in Colorectal Cancer. Accessed on 25/8/13.  Available from: http://clinicaltrials.gov/ct2/show/study/NCT01750918?term=dabrafenib AND trametinib plus&rank=1.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Holt SV, Logié A, Odedra R, Heier A, Heaton SP, Alferez D, Davies BR, Wilkinson RW, Smith PD. The MEK1/2 inhibitor, selumetinib (AZD6244; ARRY-142886), enhances anti-tumour efficacy when combined with conventional chemotherapeutic agents in human tumour xenograft models. Br J Cancer. 2012;106:858-866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 83]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
20.  Hochster HS, Messersmith WA, O’Neil BH. The MEK inhibitor selumetinib ([SEL], AZD6244, ARRY-142886) plus irinotecan (IRI) as second-line therapy for KRAS-mutated (KRASm) metastatic colorectal cancer (CRC). J Clin Oncol. 2012;31:3587.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Perrone F, Lampis A, Orsenigo M, Di Bartolomeo M, Gevorgyan A, Losa M, Frattini M, Riva C, Andreola S, Bajetta E. PI3KCA/PTEN deregulation contributes to impaired responses to cetuximab in metastatic colorectal cancer patients. Ann Oncol. 2009;20:84-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 306]  [Cited by in F6Publishing: 334]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
22.  Bendell JC, Rodon J, Burris HA, de Jonge M, Verweij J, Birle D, Demanse D, De Buck SS, Ru QC, Peters M. Phase I, dose-escalation study of BKM120, an oral pan-Class I PI3K inhibitor, in patients with advanced solid tumors. J Clin Oncol. 2012;30:282-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 530]  [Cited by in F6Publishing: 558]  [Article Influence: 42.9]  [Reference Citation Analysis (0)]
23.  Bendell JC; ClinicalTrials. gov. Identifier: NCT01304602.  Available from: http://www.clinicaltrials.gov/ct2/show/CT01304602.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Sanchez RM, Erhard K, Hardwicke MA, Lin H, McSurdy-Freed J, Plant R, Raha K, Rominger CM, Schaber MD, Spengler MD. Synthesis and structure-activity relationships of 1,2,4-triazolo[1,5-a]pyrimidin-7(3H)-ones as novel series of potent β isoform selective phosphatidylinositol 3-kinase inhibitors. Bioorg Med Chem Lett. 2012;22:3198-3202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
25.  Waniczek D, Śnietura M, Młynarczyk-Liszka J, Pigłowski W, Kopeć A, Lange D, Rudzki M, Arendt J. PTEN expression profiles in colorectal adenocarcinoma and its precancerous lesions. Pol J Pathol. 2013;64:15-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
26.  Waniczek D; ClinicalTrials. gov. Identifier: NCT01458067.  Available from: http://www.clinicaltrials.gov/ct2/show/NCT01458067.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Kondapaka SB, Singh SS, Dasmahapatra GP, Sausville EA, Roy KK. Perifosine, a novel alkylphospholipid, inhibits protein kinase B activation. Mol Cancer Ther. 2003;2:1093-1103.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Bendell J, Ervin TJ, Senzer N, Richards DA, Firdaus I, Lockhart AC, Eng C. Results of the X-PECT study: A phase III randomized double-blind, placebo-controlled study of perifosine plus capecitabine (P-CAP) vs placebo plus capecitabine (CAP) in patients (pts) with refractory metastatic colorectal cancer (mCRC). J Clin Oncol. 2012;30:abstr LBA3501.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Speranza G, Kinders RJ, Khin S, Weil MK, Do KT, Horneffer Y, Kummar S. Pharmacodynamic biomarker-driven trial of MK-2206, an AKT inhibitor, with AZD6244 (selumetinib), a MEK inhibitor, in patients with advanced colorectal carcinoma (CRC). J Clin Oncol. 2012;30:3529.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Ng K, Tabernero J, Hwang J, Bajetta E, Sharma S, Del Prete SA, Arrowsmith ER, Ryan DP, Sedova M, Jin J. Phase II study of everolimus in patients with metastatic colorectal adenocarcinoma previously treated with bevacizumab-, fluoropyrimidine-, oxaliplatin-, and irinotecan-based regimens. Clin Cancer Res. 2013;19:3987-3995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 51]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
31.  Altomare I, Bendell JC, Bullock KE, Uronis HE, Morse MA, Hsu SD, Zafar SY, Blobe GC, Pang H, Honeycutt W. A phase II trial of bevacizumab plus everolimus for patients with refractory metastatic colorectal cancer. Oncologist. 2011;16:1131-1137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 52]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
32.  Boccaccio C, Comoglio PM. Invasive growth: a MET-driven genetic programme for cancer and stem cells. Nat Rev Cancer. 2006;6:637-645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 412]  [Cited by in F6Publishing: 404]  [Article Influence: 22.4]  [Reference Citation Analysis (0)]
33.  Di Renzo MF, Olivero M, Giacomini A, Porte H, Chastre E, Mirossay L, Nordlinger B, Bretti S, Bottardi S, Giordano S. Overexpression and amplification of the met/HGF receptor gene during the progression of colorectal cancer. Clin Cancer Res. 1995;1:147-154.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Munshi N, Jeay S, Li Y, Chen CR, France DS, Ashwell MA, Hill J, Moussa MM, Leggett DS, Li CJ. ARQ 197, a novel and selective inhibitor of the human c-Met receptor tyrosine kinase with antitumor activity. Mol Cancer Ther. 2010;9:1544-1553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 269]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
35.  Bessudo A, Bendell JC, Gabrail N, Kopp MV, Mueller L, Hart LL, Eng C. Phase I results of the randomized, placebo controlled, phase I/II study of the novel oral c-MET inhibitor, ARQ 197, irinotecan (CPT-11), and cetuximab (C) in patients (pts) with wild-type (WT) KRAS metastatic colorectal cancer (mCRC) who have received front-line systemic therapy. J Clin Oncol. 2011;29:3582.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Eng C, Hart L, Severtsev A. A randomized, placebo-controlled, phase I/II study of tivantinib (ARQ 197) in combination with cetuximab and irinotecan in patients (pts) with KRAS wild-type (WT) metastatic colorectal cancer (CRC) who had received previous front-line systemic therapy. J Clin Oncol. 2013;31:abstr 3508.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Bendell JC, Ervin TJ, David H. A randomized, phase II, multicenter, double-blind, placebo-controlled study evaluating onartuzumab (MetMAb) in combination with mFOLFOX6 plus bevacizumab in patients with metastatic colorectal cancer. J Clin Oncol. 2012;30:abstr TPS3640.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Wells A. EGF receptor. Int J Biochem Cell Biol. 1999;31:637-643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 722]  [Cited by in F6Publishing: 707]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
39.  Yonesaka K, Zejnullahu K, Okamoto I, Satoh T, Cappuzzo F, Souglakos J, Ercan D, Rogers A, Roncalli M, Takeda M. Activation of ERBB2 signaling causes resistance to the EGFR-directed therapeutic antibody cetuximab. Sci Transl Med. 2011;3:99ra86.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Bertotti A, Migliardi G, Galimi F, Sassi F, Torti D, Isella C, Corà D, Di Nicolantonio F, Buscarino M, Petti C. A molecularly annotated platform of patient-derived xenografts (“xenopatients”) identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. Cancer Discov. 2011;1:508-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 645]  [Cited by in F6Publishing: 695]  [Article Influence: 53.5]  [Reference Citation Analysis (0)]
41.  Marsoni S, Bertotti A, Sartore-Bianchi A. Dual anti-HER2 treatment of patients with HER2-positive metastatic colorectal cancer: The HERACLES trial (HER2 Amplification for Colo-rectaL Cancer Enhanced Stratification). J Clin Oncol. 2013;31:abstr TPS3648.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Frank D, Jumonville A, Loconte NK, Schelman WR, Mulkerin D, Lubner S, Richter K, Winterle N, Wims MB, Dietrich L. A phase II study of capecitabine and lapatinib in advanced refractory colorectal adenocarcinoma: A Wisconsin Oncology Network study. J Gastrointest Oncol. 2012;3:90-96.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Hendriks YM, de Jong AE, Morreau H, Tops CM, Vasen HF, Wijnen JT, Breuning MH, Bröcker-Vriends AH. Diagnostic approach and management of Lynch syndrome (hereditary nonpolyposis colorectal carcinoma): a guide for clinicians. CA Cancer J Clin. 2006;56:213-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 108]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
44.  Vogelstein B, Kinzler KW.  The Genetic Basis of Human Cancer. New York: McGraw Hill, Medical 2002; .  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Hinoue T, Weisenberger DJ, Pan F, Campan M, Kim M, Young J, Whitehall VL, Leggett BA, Laird PW. Analysis of the association between CIMP and BRAF in colorectal cancer by DNA methylation profiling. PLoS One. 2009;4:e8357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 121]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
46.  Sinicrope FA, Sargent DJ. Molecular pathways: microsatellite instability in colorectal cancer: prognostic, predictive, and therapeutic implications. Clin Cancer Res. 2012;18:1506-1512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 188]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
47.  Vilar E, Bartnik CM, Stenzel SL, Raskin L, Ahn J, Moreno V, Mukherjee B, Iniesta MD, Morgan MA, Rennert G. MRE11 deficiency increases sensitivity to poly(ADP-ribose) polymerase inhibition in microsatellite unstable colorectal cancers. Cancer Res. 2011;71:2632-2642.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 120]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
48.  Casanovas O, Hicklin DJ, Bergers G, Hanahan D. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell. 2005;8:299-309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1215]  [Cited by in F6Publishing: 1231]  [Article Influence: 64.8]  [Reference Citation Analysis (0)]
49.  Siu LL, Shapiro JD, Jonker DJ, Karapetis CS, Zalcberg JR, Simes J, Couture F, Moore MJ, Price TJ, Siddiqui J. Phase III randomized, placebo-controlled study of cetuximab plus brivanib alaninate versus cetuximab plus placebo in patients with metastatic, chemotherapy-refractory, wild-type K-RAS colorectal carcinoma: the NCIC Clinical Trials Group and AGITG CO.20 Trial. J Clin Oncol. 2013;31:2477-2484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 100]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]