Skip to main content
Indian Journal of Surgical Oncology logoLink to Indian Journal of Surgical Oncology
. 2016 Jul 27;8(4):580–590. doi: 10.1007/s13193-016-0543-z

Molecular Landscape and Treatment Options for Patients with Metastatic Colorectal Cancer

Yuji Miyamoto 1,2, Wu Zhang 1, Heinz-Josef Lenz 1,
PMCID: PMC5705494  PMID: 29203992

Abstract

Over the last 20 years, median survival for patients with metastatic colorectal cancer (CRC) has remarkably improved from about 12 to over 30 months, mainly because of the development of new agents and patient selection using predictive biomarkers. However, the identification of the most effective treatment for an individual patient is still a challenge. Molecular profiling of CRC has made great progress, but it is limited by tumor heterogeneity and absence of driver mutation. However, RAS, BRAF, and microsatellite instability are validated biomarker recommended by NCCN and ESMO. In this review, we discuss recent advances and future developments.

Keywords: Colorectal cancer, Chemotherapy, Targeted therapy, Biomarker, Personalized therapy, Molecular pathway

Introduction

Over the past several years, survival outcomes for patients with colorectal cancer (CRC) have remarkably improved because of the approval and incorporation of multiple new chemotherapeutic agents. Combined with conventional cytotoxic and targeted drugs, overall survival (OS) has increased to over 30 months in patients with metastatic CRC (mCRC) [13].

Despite those improvements, identification of the most effective treatment for an individual patient is still mainly based on clinical considerations. Advances in molecular biology of CRC have led to attempts to classify CRC into subtypes based on various clinical and molecular characteristics that reflect DNA, and RNA features and protein expression [4, 5]. The Cancer Genome Atlas Network (TCGA) conducted a comprehensive molecular characterization of CRC, demonstrating the biological differences in CRC and the suggestion about therapeutic approaches to CRC. These molecularly comprehensive investigations indicate that CRC is a heterogeneous disease. Therapies targeting specific molecular characteristic show promise for CRC patients.

In this review, we focus on recent advances in the molecular characteristics of CRC, especially microsatellite instability (MSI) and somatic mutations (RAS, BRAF, and PI3KCA), next-generation sequencing (NGS) technologies, molecular classification, and their implications in treatment options for both conventional cytotoxic chemotherapy and targeted therapies for CRC.

MSI Phenotype

Microsatellite instability (MSI) is a genetic hypermutable condition that results from inactivation of the DNA mismatch repair (MMR) system. Dysfunction in genes involved in the MMR system, MLH1, MSH2, MSH6, and PMS2, results in alterations in highly repeated DNA sequences (microsatellites) [6]. A germinal mutation that inactivates MMR genes may lead to a hereditary form of MSI (“termed Lynch syndrome”). MSI is detected in approximately 15 % of sporadic CRC [7], although somatic mutations in MMR-related genes are rarely found. Methylation-induced silencing of MLH1causes most sporadic CRC with MSI [8, 9]. MSI is classified into MSI-high (MSI-H), and MSI-low (MSI-L), depending on the percentage of loci that correlate to MSI characteristics. Tumors with no positive markers for MSI are designated as microsatellite stable (MSS). Sporadic MSI-H CRC tumors are associated with a specific phenotype that includes higher frequency in older people and females, proximal location, poor differentiation, mucinous and inflammatory features, and enrichment with BRAF mutations and CpG island methylator phenotype (CIMP) status [10].

Several studies have assessed the prognostic value of MSI status in CRC (Table 1), including multiple retrospective studies and meta-analyses that show MSI-H tumors to have a more favorable stage-adjusted prognosis than MSS [12, 17, 18]. A large meta-analysis of data from 10 trials with 4014 stages II–III CRC patients, including 645 with MSI, showed an association between MSI and longer OS (OR = 0.65, 95 % CI 0.53–0.79, P < 0.0001) relative to other CRC types [19]. This relatively favorable effect of defective MMR (dMMR) on outcomes is less pronounced in stage III CRC. For metastatic disease, a pooled analysis of four randomized first-line phase III trials (CAIRO, CAIRO2, FOCUS, and COIN) that assessed the effects of both BRAF and MSI on survival found that the progression free survival (PFS) and OS were significantly shorter for patients with dMMR tumor compared with proficient MMR (pMMR) tumors (median PFS 6.2 vs 7.6 months respectively; HR = 1.33, 95 % CI 1.12–1.57, P = 0.001; median OS 13.6 vs 16.8 months respectively; HR = 1.35, 95 % CI 1.13–1.61; P = 0.001) [20]. Given the absence of a significant interaction between BRAF mutations and dMMR, these data imply that the poor prognoses associated with MSI are driven by BRAF mutation status in these patients.

Table 1.

Clinical trials that evaluated the prognostic and predictive significance of mismatch repair status in colorectal cancer

Study [reference] Stage Treatment MMR status N 5-year DFS/TTR /RFS (%) HR 95 % CI P 5-year OS (%) HR 95 % CI P
Sinicrope et al. [11] II/III dMMR
pMMR
344
1797
72
63
0.73 0.59–0.91
70
0.004 77
70
0.73 0.59–0.90 0.004
Sargent et al. [12] II/III Surgery alone dMMR
pMMR
37
191
76
53
0.46 0.22–0.95 0.03 81
62
0.51 0.24–1.10 0.06
5-FU dMMR
pMMR
33
196
71
64
0.90 0.44–1.82 0.77 75
71
0.73 0.35–1.54 0.41
ACCENT [13] II Surgery alone dMMR
pMMR
63
244
89
74
0.35 0.15–0.80 0.013 90
78
0.37 0.17–0.81 0.013
5-FU dMMR
pMMR
235
920
88
83
0.84 0.57–1.24 0.37 88
87
0.91 0.63–1.31 0.62
III Surgery alone dMMR
pMMR
37
277
60
47
0.79 0.45–1.39 0.41 59
54
0.84 0.49–1.43 0.51
5-FU dMMR
pMMR
390
2333
72
64
0.82 0.67–0.99 0.04 77
71
0.81 0.67–0.99 0.039
NSABP C-07 [14] II/III 5-FU
FLOX
dMMR 86
85
NA
NA
1.01 0.45–2.25 0.98 NA
NA
NA
NA
NA
NA
NA
NA
5-FU
FLOX
pMMR 635
675
NA
NA
0.82 0.67–1.00 0.054 NA
NA
NA
NA
NA
NA
NA
NA
MOSAIC [15] II/III 5-FU
FOLFOX
dMMR 51
44
74
86
0.48 0.21–1.12 0.088 82
91
0.41 0.16–1.07 0.069
5-FU
FOLFOX
pMMR 442
471
65
72
0.87 0.70–1.07 0.185 79
81
0.91 0.72–1.15 0.428
N0147 [16] dMMR
pMMR
314
2266
NA
NA
0.82 0.64–1.04 0.1055 NA
NA
0.83 0.63–1.09 0.1822

On the other hand, the predictive value of MMR status on adjuvant chemotherapy outcome is still controversial. Some studies imply that dMMR status is a predictor of decreased benefit from adjuvant 5-FU-based therapy in stage II CRC patients [12, 21]. In addition, a meta-analysis of pooled data from seven trials that included 454 MSI out of 3690 stage II-III CRC patients also showed no significant differences for recurrence-free survival (RFS: HR = 0.96, 95 % CI 0.62–1.49, P = 0.86) and OS (HR = 0.70, 95 % CI 0.44–1.09, P = 0.12) whether or not they received 5-FU based chemotherapy, whereas MSS patients had better responses to chemotherapy [22]. In contrast, a study of 1913 patients with stage II CRC from the QUASAR study showed that dMMR did not predict benefit or detrimental impact of chemotherapy [23]. Another analysis from the ACCENT database involving 7803 stage II–III CRC cases revealed that among patients with stage II CRC treated with 5-FU adjuvant chemotherapy, time to recurrence (TTR) or OS did not differ between dMMR and pMMR (TTR: HR = 0.84, 95 % CI 0.57–1.24; P = 0.37; OS, HR = 0.91; 95 % CI 0.63–1.31; P = 0.62). In stage III CRC, however, patients with dMMR cancers treated with adjuvant 5-FU had better outcome compared to pMMR tumors (TTR: HR = 0.82, 95 % CI 0.67–0.99; P = 0.04; OS, HR = 0.81; 95 % CI 0.67–0.99; P = 0.039) [24]. In summary, because patients with stage II MSI-H tumors may have good prognoses and do not benefit from 5-FU adjuvant therapy, the NCCN panel recommends that MMR or MSI testing be performed for all patients with stage II disease, and adjuvant therapy should not be given to patients with low-risk stage II MSI-H tumors [25].

Although retrospective analyses of patients with stage III colon cancer who received adjuvant FOLFOX imply that dMMR CRCs are sensitive to oxaliplatin [26, 27], data from prospective clinical trials of oxaliplatin-based treatment are limited [28].

Significant responses to anti-PD-1 inhibitors in patients with MSI (+) cancers who failed standard chemotherapy were recently reported [29]. MSI was a significant predictor of the immune-related objective response rate (ORR; dMMR CRC 40 %; pMMR CRC 0 %) and also the immune-related PFS rate (dMMR 78 %, pMMR 11 %). The immune microenvironment of MSI CRC has been found to contain strong Th1 and CTL components not found in the vast majority of MSS tumors [30], which could significantly improve therapeutic options for mCRC [31] although the MSI rates in mCRC are very low (5 %).

Ras

RAS proteins (H-, K-, and N-Ras) are low-molecular-weight GTP-binding proteins that affect cell differentiation, proliferation, and survival [32]. Approximately 40 % of CRCs are characterized by mutations in codons 12 and 13 in exon 2 of the coding region of the KRAS gene [3335]. A landmark finding showed that tumor KRAS mutations do not respond to anti-EGFR therapies, owing to direct activation of the MAPK signaling pathway [36]. More recent studies have also associated lower-frequency (10 %) mutations in KRAS exon 3 or 4 or in NRAS exons 2, 3, and 4 to resistance to anti-EGFR therapies [3739]. A meta-analysis of nine randomized, controlled trials of EGFR mAbs for mCRC that evaluated RAS mutational status supported the predictive value of RAS mutational profiles for both PFS and OS [40]. Because of potential detrimental effects of anti-EGFR therapies on CRC patients whose tumors have any RAS mutations, such patients should be excluded from anti-EGFR treatments (Table 2) [40, 45].

Table 2.

Summary of phase III trial according to RAS mutational status

Study Phase RAS type Therapy n PFS (M) HR 95 % CI P OS (M) HR 95 % CI P P ORR (%) P
OPUS [41] II KRASWt FOLFOX + Cmab
FOLFOX
82
97
8.3
7.2
0.57 0.38–0.86 0.0064 22.8 0.86 0.60–1.22 0.39 57 0.0027
All RASWt FOLFOX + Cmab
FOLFOX
38
49
12.0
5.8
0.53 0.27–1.04 0.0615 19.8
17.8
0.94 0.56–1.56 0.80 58
29
0.0084
CRYSTAL [42] III KRASWt FOLFIRI + Cmab
FOLFIRI
316
350
9.9
8.4
0.70 0.56–0.87 0.0012 23.5
20.0
0.80 0.67–0.95 0.0093 57
40
<0.001
All RASWt FOLFIRI + Cmab
FOLFIRI
178
189
11.4
8.4
0.56 0.41–0.76 <0.001 28.4
20.0
0.69 0.54–0.88 0.0024 66 <0.001
PRIME [38] III KRASWt FOLFOX + Pmab
FOLFOX
325
331
9.6
8.0
0.80 0.66–0.97 0.02 23.8
19.4
0.83 0.70–0.98 0.03 NA NA
All RASWt FOLFOX + Pmab
FOLFOX + Bmab
259
253
10.1
7.9
0.72 0.58–0.90 0.004 25.8
20.2
0.77 0.64–0.94 0.009 NA NA
PEAK [43] III KRASWt FOLFOX + Pmab
FOLFOX + Bmab
142
143
10.9
10.1
0.87 0.65–1.17 0.353 34.2
24.3
0.62 0.44–0.89 0.009 58
54
NA
All RASWt FOLFOX + Pmab
FOLFOX
88
82
13.0
9.5
0.65 0.44–0.96 0.029 41.3
28.9
0.63 0.39–1.02 0.058 64
61
NA
FIRE-3
[3]
III KRASWt FOLFIRI + Cmab
FOLFIRI + Bmab
297
295
10.0
10.3
1.06 0.88–1.26 0.55 28.7
25.0
0.77 0.62–0.96 0.017 62 0.18
All RASWt FOLFIRI + Cmab
FOLFIRI + Bmab
171
171
10.4
10.2
0.93 0.74–1.17 0.54 33.1
25.6
0.70 0.53–0.92 0.011 65
60
0.32
CALGB/SWOG
80405
[44]
III KRASWt FOLFIRI/FOLFOX + Cmab
FOLFIRI/FOLFOX + Bmab
578
599
10.4
10.8
1.04 0.91–1.17 0.55 29.9
29.0
0.92 0.78–1.09 0.34 66
57
0.02
All RASWt FOLFIRI/FOLFOX + Cmab
FOLFIRI/FOLFOX + Bmab
270
256
11.4
11.3
1.10 NA 0.31 32.0
31.2
0.9 0.7–1.1 0.40 69
54
<0.01

PFS progression free survival, M months, HR hazard ratio, CI confidence interval, OS overall survival, ORR objective response rate, Wt wild-type, Cmab cetuximab, Pmab panitumumab, Bmab bevacizumab

The prognostic significance of KRAS mutations have also been evaluated; however, some systemic reviews indicate no association between KRAS mutant status and short- or long-term prognosis of patients with CRC [46]. Possibly, different mutations within the gene could have disparate prognostic influences. However, these tests are not currently recommended for prognostic purposes [25].

Retrospective data from large phase III trials suggested that patients who carry the KRAS G13D mutation might benefit from cetuximab [47] and panitumumab [48] in first and advanced treatment lines [49], which implied that the KRAS G13D mutation is clinically significant. Because of these results, two prospective phase II trials [50, 51] were performed; however, neither trial showed activity from cetuximab monotherapy in patients with KRAS G13D-mutated mCRC. Currently, anti-EGFR therapy for patients whose tumors have G13D mutations is not routinely recommended.

Even among patients with RAS wild-type (wt) mCRC that initially responds to anti-EGFR therapies, essentially all eventually develop secondary resistance to these agents through numerous mechanisms. Diaz et al. assessed circulating tumor DNA (ctDNA) in the blood of mCRC patients during panitumumab therapy and found that preexisting low-abundance KRAS-mutant clones may be selected through anti-EGFR treatment [52]. Additionally, Bertotti et al. used large-scale genomic and targeted therapeutic analyses of CRC patients who underwent anti-EGFR therapy to show that insulin receptor substrate 2 (IRS2) mediates a novel mechanism of sensitivity to anti-EGFR therapy [53]. IRS2 is regulated by insulin receptor (IR)/insulin-like growth factor (IGF) receptors [54]. IRS2 amplification and mutations were identified in tumors with increased sensitivity to anti-EGFR therapy. Another apparent mechanism of resistance to cetuximab is an EGFR extracellular domain (ECD) mutation that prevents binding to cetuximab but not to panitumumab [55]. The EGFR S492R mutation alters the epitope by which cetuximab binds EGFR, but because panitumumab binds a different epitope than cetuximab, patients with these mutations might benefit from subsequent panitumumab therapy.

Some new EGFR inhibitors appear to overcome resistance to cetuximab or panitumumab due to the emergence of EGFR ECD mutations. MM-151 is a third-generation EGFR inhibitor that uses three distinct antibodies that bind different EGFR epitopes to block potently signaling downstream of EGFR, despite escalating doses of EGF [56]. A preclinical study showed MM-151 to inhibit cell signaling and proliferation in cells from a patient whose tumor developed an EGFR ECD mutation during cetuximab treatment [57]. Moreover, after MM-151 treatment, liquid biopsies of patients with EGFR ECD mutations at baseline showed decreased or stabilized EGFR ECD mutant DNA concentrations that paralleled response assessments after using radiological methods.

Sym004 is another mixture of two synergistic nonoverlapping anti-EGFR antibodies that target different epitopes of the EGFR ECD [58]. Sym004 synergistically induces EGFR internalization and degradation in vitro and inhibits tumor growth in vivo [59]. Colorectal tumors of patients who develop secondary resistance to EGFR blockade often display heterogeneous resistance mechanisms, which would presumably be insensitive to EGFR-targeted monotherapy, including these new agents. Additional work is needed to elucidate relationships among these concurrent mechanisms of resistance.

BRAF

The BRAF protein is a serine/threonine kinase, downstream of KRAS, which acts as signal transducer from the membrane to the nucleus [60], and is mutated in approximately 10 % of CRC [61]. The most common BRAF mutation is at position V600E and causes a constitutive activation of the MAPK pathway [62]. In addition, BRAF mutations are mutually exclusive with RAS mutations [63]. In CRC, BRAF mutations are found more commonly in tumors with proximal locations, poor differentiation, mucinous histology, CIMP, or MSI [64, 65]. Results from a recent systematic review indicate that active BRAF mutations occur mostly in tumors with proximal location (OR 5.22; 95 % CI 3.80–7.17; P < 0.001), T4 grade (OR 1.76; 95 % CI 1.16–2.66; P = 0.007), and/or poor differentiation (OR 3.82; 95 % CI 2.71–5.36; P < 0.001) [66].

Evidence from retrospective analyses and randomized clinical trials has associated BRAF mutations with poor prognosis in patients with pMMR tumors (Table 3) [75, 76]. A prospective analysis of tissues from patients with stage II–III colon cancer enrolled in the PETACC-3 trial showed that the BRAF mutation is prognostic for shorter OS in patients with MSI-L or MSS tumors (HR 2.2; 95 % CI 1.4–3.4; P = 0.0003) [75]. A recent meta-analysis also showed that patients with advanced RAS-wt/BRAF-mutant CRC did not benefit from cetuximab or panitumumab treatments [77]. BRAF mutation status cannot be shown to predict benefit from cetuximab treatment [67]. Therefore, this testing is currently optional and not a necessary part of deciding whether to use anti-EGFR agents.

Table 3.

Summary of clinical trials in BRAF mutant tumor

Treatment line Trial (reference) BRAF mutation Regimen N PFS (M) HR 95 % CI P OS (M) HR 95 % CI P ORR (%) P
1st CRYSTAL [67] 6 % FOLFIRI + Cmab
FOLFIRI
26
33
8.0
5.6
0.93 0.43–2.06 0.87 14.1
10.3
0.91 0.51–1.62 0.74 15.2
19.2
0.91
1st OPUS [68] 4 % FOLFOX + Cmab
FOLFOX
6
5
NA NA NA NA 20.7
4.4
NA NA NA NA NA
1st PRIME [38] 8 % FOLFOX + Pmab
FOLFOX
24
29
6.1
5.4
0.58 0.29–1.15 0.12 10.5
9.2
0.90 0.46–1.76 0.76 NA NA
1st COIN [69] 8 % FOLFOX/XELOX + Cmab FOLFOX/XELOX 45
57
NA NA NA NA 7.2
10.0
1.18 0.76–1.81 0.46 NA NA
1st FIRE-3 [70] 12 % FOLFIRI + Cmab
FOLFIRI + Bmab
23
25
4.9
6.0
0.87 0.49–1.57 0.65 12.3
13.7
0.87 0.47–1.61 0.65 52.2
40.0
0.29
2nd 20050181 [71] 8 % FOLFIRI + Pmab
FOLFIRI
22
23
2.5
1.8
0.69 0.32–1.49 0.34 4.7
5.7
0.64 0.32–1.28 0.20 NA NA
2nd> PICCOLO [72] 15 % CPT-11 + Pmab
CPT-11
31
37
NA 1.40 0.82–2.39 NA NA 1.84 1.10–3.08 NA 6.5
10.8
NA
2nd> CO.17 [73] 5 % Cmab
BSC
4
6
NA 0.76 NA 0.69 1.8
3.0
0.84 0.20–3.58 0.81 0 NA
3rd 20020408 [74] 12 % Pmab
BSC
9
6
NA 0.34 0.09–1.24 NA NA NA NA NA 0
0
NA

PFS progression-free survival, M months, HR hazard ratio, CI confidence interval, OS overall survival, ORR objective response rate, wt wild-type, Cmab cetuximab, Pmab panitumumab, Bmab bevacizumab, NA not available

BRAF inhibitors, such as vemurafenib and dabrafenib, have produced dramatic response rates of 50–80 % in BRAF V600 mutant melanoma. In contrast, only a 5 % response rate was seen in patients treated with vemurafenib who had metastatic CRC that harbored the same BRAF V600 mutation [78, 79]. Corcoran et al. investigated this differential outcome and found that MAPK suppression by BRAF inhibitor alone in BRAF-mutant CRC cells was transient, followed by rapid reactivation of MAPK signaling and re-accumulation of phosphorylated ERK, despite continued presence of the drug [80]. Therefore, combination strategies to inhibit both BRAF and EGFR, or other approaches that target interacting pathways (i.e., that co-targets BRAF plus MEK or PI3K), are required. Other findings highlighted hyperactivation of the PI3K/Akt/PTEN pathway as a mechanism of resistance to BRAF inhibition in CRC [79, 81]. Vemurafenib with an AKT inhibitor was found to inhibit growth of BRAF-mutant CRC xenografts in a murine model [79]. Ongoing trials in patients with BRAF-mutant CRC will provide important insights on this issue (Table 4).

Table 4.

Recent and ongoing clinical trials of BRAF inhibitor for mCRC

Strategy Therapy Genomic profile Phase N ORR (%) DCR (%) PFS (M) OS (M) Clinical development Reference
BRAF monotherapy Vemurafenib BRAFMut I 21 5 84 2.1 7.7 Complete NCT00405587 [78]
BRAF monotherapy Vemurafenib BRAFMut BRAFMut II 10 0 50 4.5 9.3 Complete NCT01524978 [82]
BRAF + MEK Dabrafenib + trametnib BRAFMut I/II 43 12 63 3.5 NA Complete NCT01726738 [83]
BRAF + EGFR Vemurafenib + cetuximab BRAFMut II 27 4 73 3.7 7.1 Complete NCT01524978 [82]
BRAF + EGFR Vemurafenib + panitumumab BRAFMut Pilot 15 13 67 3.2 7.6 Complete NCT01791309 [84]
BRAF + EGFR Encorafenib + cetuximab BRAFMut + KRASWt I/II 26 23 NA 3.7 NA Ongoing NCT01719380 [85]
BRAF + EGFR Dabrafenib + panitumumab BRAFMut I/II 20 10 90 NA NA Ongoing NCT01750918 [86]
BRAF + EGFR + MEK Dabrafenib + panitumumab + trametnib BRAFMut I/II 24 21 83 NA NA Ongoing NCT01750918 [86]
BRAF + EGFR + PI3K Encorafenib + cetuximab + alpelisib BRAFMut + KRASWt I/II 28 25 NA 4.3 NA Ongoing NCT01719380 [85]
BRAF + EGFR + CT Vemurafenib + cetuximab + irinotecan BRAFMut I 12 44 89 NA NA Ongoing NCT01787500 [87]
BRAF + EGFR + CT Vemurafenib + cetuximab + irinotecan BRAFMut II (RCT) 78* NA NA NA NA Ongoing NCT02164916
BRAF + EGFR + WNT Encorafenib + cetuximab + WNT974 BRAFMut + WNTMut I/II 60* NA NA NA NA Ongoing NCT02278133

Recent data suggests that FOLFOXIRI + bevacizumab (Bmab) might be a reasonable first-line treatment for BRAF-mutant mCRC [88, 89]. Although its treatment effect did not significantly differ from that of FOLFIRI + Bmab, its survival HRs were OS of 0.54 (95 % CI: 0.24–1.20) and PFS of 0.57 (95 % CI 0.27–1.23) compared with FOLFIRI + Bmab.

PIK3CA

As with the MAPK pathway, EGFR signaling is mediated also by the PI3K-AKT pathway which is important for cancer cell survival [90]. PI3KCA is a heterodimeric protein, composed of a regulatory subunit (p85) and a catalytic subunit (p110); it functions as a lipid kinase. PIK3CA mutations are present in approximately 15 % of CRCs [34]; most of these mutations are single amino-acid substitutions located in hot spots in the helical (exon 9) or kinase domains (exon 20) [91]. These mutations may also affect anti-EGFR therapy responsiveness. Mao et al. conducted a systematic review and meta-analysis to investigate the association between PIK3CA mutations and resistance to anti-EGFR therapy in mCRC, by mutated PIK3CA exon. Their systematic review of 13 studies showed that PIK3CA mutation alone did not correlate with survival of patients with metastatic disease. Among patients with KRAS-wt mCRC who underwent anti-EGFR therapy, PIK3CA mutation on exon 20 was related to worse prognosis, but not significantly so. This result suggests that PIK3CA exon 20 mutations is a biomarker for resistance to anti-EGFR therapy in KRAS-wt mCRC; further studies are needed to confirm this relation and clarify its clinical implications.

In addition to the adjuvant setting, two large observational studies suggested that aspirin use was associated with significantly increased survival in patients with PIK3CA-mutant CRC but not PIK3CA-wt CRC [92, 93]. The underlying mechanism behind the benefit of aspirin in this setting is unclear. PI3K mutations result in constitutive activation of the AKT pathway, which enhances NF-kB, a transcription factor that plays a pivotal role in upregulating pro-inflammatory genes, including COX-2. PI3KCA mutations may also activate signaling of Wnt, which is an essential oncogenic pathway in CRC. Aspirin inhibits activity of both NF-kB and the Wnt/β-catenin pathway; patients with PIK3CA-mutant CRC may therefore benefit from this mechanism.

Molecular Classification of CRC

The development of convincing NGS technologies has revolutionized DNA sequencing by reducing both its time and cost in the past few years, offering the opportunity for a comprehensive description of the different CRC subtypes [94]. Pivotal comprehensive study conducted by TCGA showed the first tumor dataset with complete molecular measurements, including exome sequencing, DNA copy number, promoter methylation, and mRNA and miRNA expression analysis [4, 95]. According to whole genome sequencing, 16 % were found to be hypermutated, which were associated with MSI and/or CIMP. Among the nonhypermutated cancers, the four most frequently mutated genes are APC (81 %), p53 (59 %), KRAS (50 %), and PIK3CA (18 %). Among the hypermutated cancers, the four most frequently mutated genes are ACVR2A (63 %), APC (51 %), TGFBR2 (51 %), and BRAF (46 %). The WNT signaling pathway was altered in 93 % of all tumors, including biallelic inactivation of APC or activating mutations of CTNNB1. Furthermore, genomic alterations involved in TGF-beta signaling pathway were found in 27 % of the nonhypermutated and 87 % of the hypermutated tumors. Recurrent genetic alterations are eventually grouped according to the activity of four pathways (WNT, MAPK-PI3K, TGF-B, and p53) by hypermutation status. The findings from the TCGA have provided a useful resource for understanding CRC and, at the same time, have required the development of new classification schemes.

Recently, an international consortium has proposed a gene-expression-based subtyping classification system for CRC that defines four consensus molecular subtypes (CMSs) of CRC with distinguishing features: CMS1 (MSI–immune, 14 %), demonstrating hypermutation, MSI, and strong immune activation; CMS2 (canonical, 37 %), epithelial tumors with marked activation of WNT and MYC signaling; CMS3 (metabolic, 13 %), epithelial tumors with evident metabolic dysregulation; and CMS4 (mesenchymal, 23 %), with prominent activation of TGFβ signaling, stromal invasion, and angiogenesis [5]. In addition, the CMS groups have important associations with some clinical variables and prognosis. The next step should be to link these subtype classifications to response to therapy to identify predictive biomarkers.

Liquid Biopsy

This new genetic technology has allowed to identify cell-free circulating tumor DNA (ctDNA) and RNA (ctRNA) which are released from tumor cells into bloodstream. A BEAMing digital PCR technology (beads, emulsion, amplification, and magnetics) is the methods of detecting somatic mutations in small amounts of ctDNA. This system amplifies nucleic acids in the presence of magnetic beads and assesses its quantity by using a flow cytometry [96], allowing to identify genetic variations of ctDNA, but also to precisely quantify their number in comparison to the number of wild-type sequences [97]. However, this method interrogates only a few loci, and mutations in genes that lack mutational hotspots, such as tumor suppressors, are missed. On the other hand, NGS technologies have been used in the field of liquid biopsy. The main advantage of NGS of a liquid biopsy is that this approach is applicable to all patients because it does not rely on recurrent genetic changes [98].

Many studies suggested that ctDNA levels might be useful for monitoring patients with CRC and for recognizing individuals with high-risk recurrence [99]. Frattini et al. showed that ctDNA levels were significantly higher in patients with CRC; they decreased during the follow-up period in tumor-free patients and increased again at tumor recurrence [100]. These results are in line with the findings of Spindler et al. which demonstrated that median ctDNA levels were significantly higher in mCRC compared to healthy individuals and that confirmed a shorter OS with increasing levels of baseline ctDNA [101]. The correlation between the ctDNA level and clinical response was also assessed in a retrospective study of the CORRECT study cohort [102]. Although a high ctDNA level does not predict regorafenib clinical response, ctDNA level was found as prognostic factor, with shorter median survival observed in patients with a higher ctDNA level than in patients with a lower ctDNA level regardless of treatment.

The detection of somatic point mutations in ctDNA was reported by in 1994 [103]. Technological advances have improved the analytical sensitivity of detection RAS and BRAF mutations in the plasma samples of mCRC patients [104106]. KRAS mutant fragments were detected in the blood of patients with KRAS-mutant colorectal tumors, with high specificity (98 %) and sensitivity (92 %) [107]. These results suggest that the “liquid biopsy” is a feasible alternative to a solid tissue biopsy for identifying specific mutations. When tumor tissue specimens from metastatic cancer patients are unavailable, liquid biopsies offer an alternative that can be rapidly implemented without the pain, risk, and expense entailed by a biopsy of one of the metastatic lesions [105].

Furthermore, liquid biopsies allow the identification of resistance mutations that occur when patients first respond to therapy and then progress [108, 109]. Diaz et al. showed that 38 % of patients whose tumor were initially KRAS wild type developed detectable mutation in KRAS in their serum. In the same way, Bettegowda et al. reported that 96 % of patients, who objectively responded to anti-EGFR therapy but subsequently relapsed, developed one or more mutations in genes involved in the mitogen-activated protein kinase pathway [105]. These results suggest that liquid biopsy could detect the emergence of KRAS mutant clones months before radiographic progression [110].

Conclusions

With increasing number of new drugs in clinical studies and development of new technologies such as liquid biopsies, personalized therapies are becoming more and more a reality. We begin to understand that CRC is a heterogeneous disease with no driver mutations and most of the genetic aberrations are not sufficient to guide treatment decisions. We only have a few predictive and prognostic markers we can clinically use. In the future, NGS technology will help our progress toward understanding CRC genomes and molecular classifications, which will lead to a better personalized targeted therapeutics for CRC management. There is growing need for translational research focused on the early identification of biomarkers, and predictive biomarkers require even more extensive data for validation derived from large randomized clinical trials and meta-analysis.

Compliance with Ethical Standards

Conflict of Interest

HJ Lenz has received honoraria from Merck Serono, Roche, Celgene, Bayer, and Boehringer Ingelheim. The other authors have no conflict of interest.

References

  • 1.Loupakis F, Cremolini C, Masi G, et al. Initial therapy with FOLFOXIRI and bevacizumab for metastatic colorectal cancer. N Engl J Med. 2014;371:1609–1618. doi: 10.1056/NEJMoa1403108. [DOI] [PubMed] [Google Scholar]
  • 2.Yamada Y, Takahari D, Matsumoto H, et al. Leucovorin, fluorouracil, and oxaliplatin plus bevacizumab versus S-1 and oxaliplatin plus bevacizumab in patients with metastatic colorectal cancer (SOFT): an open-label, non-inferiority, randomised phase 3 trial. Lancet Oncol. 2013;14:1278–1286. doi: 10.1016/S1470-2045(13)70490-X. [DOI] [PubMed] [Google Scholar]
  • 3.Heinemann V, von Weikersthal LF, Decker T, et al. FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer (FIRE-3): a randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15:1065–1075. doi: 10.1016/S1470-2045(14)70330-4. [DOI] [PubMed] [Google Scholar]
  • 4.Cancer Genome Atlas Network Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487:330–337. doi: 10.1038/nature11252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Guinney J, Dienstmann R, Wang X, et al. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21:1350–1356. doi: 10.1038/nm.3967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Thibodeau SN, Bren G, Schaid D. Microsatellite instability in cancer of the proximal colon. Science. 1993;260:816–819. doi: 10.1126/science.8484122. [DOI] [PubMed] [Google Scholar]
  • 7.Ionov Y, Peinado MA, Malkhosyan S, Shibata D, Perucho M. Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis. Nature. 1993;363:558–561. doi: 10.1038/363558a0. [DOI] [PubMed] [Google Scholar]
  • 8.Kane MF, Loda M, Gaida GM, et al. Methylation of the hMLH1 promoter correlates with lack of expression of hMLH1 in sporadic colon tumors and mismatch repair-defective human tumor cell lines. Cancer Res. 1997;57:808–811. [PubMed] [Google Scholar]
  • 9.Hawkins N, Norrie M, Cheong K, et al. CpG island methylation in sporadic colorectal cancers and its relationship to microsatellite instability. Gastroenterology. 2002;122:1376–1387. doi: 10.1053/gast.2002.32997. [DOI] [PubMed] [Google Scholar]
  • 10.Weisenberger DJ, Siegmund KD, Campan M, et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet. 2006;38:787–793. doi: 10.1038/ng1834. [DOI] [PubMed] [Google Scholar]
  • 11.Sinicrope FA, Foster NR, Thibodeau SN, et al. DNA mismatch repair status and colon cancer recurrence and survival in clinical trials of 5-fluorouracil-based adjuvant therapy. J Natl Cancer Inst. 2011;103:863–875. doi: 10.1093/jnci/djr153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Sargent DJ, Marsoni S, Monges G, et al. Defective mismatch repair as a predictive marker for lack of efficacy of fluorouracil-based adjuvant therapy in colon cancer. J Clin Oncol. 2010;28:3219–3226. doi: 10.1200/JCO.2009.27.1825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sargent DJ, Shi Q, Yothers G et. al (2014) Prognostic impact of deficient mismatch repair (dMMR) in 7,803 stage II/III colon cancer (CC) patients (pts): a pooled individual pt data analysis of 17 adjuvant trials in the ACCENT database. J Clin Oncol 32:5s (suppl; abstr 3507).
  • 14.Gavin PG, Paik S, Yothers G, Pogue-Geile KL. Colon cancer mutation: prognosis/prediction--response. Clin Cancer Res. 2013;19:1301. doi: 10.1158/1078-0432.CCR-13-0020. [DOI] [PubMed] [Google Scholar]
  • 15.Andre T, de Gramont A, Vernerey D, et al. Adjuvant fluorouracil, leucovorin, and oxaliplatin in stage II to III colon cancer: updated 10-year survival and outcomes according to BRAF mutation and mismatch repair status of the MOSAIC study. J Clin Oncol. 2015;33:4176–4187. doi: 10.1200/JCO.2015.63.4238. [DOI] [PubMed] [Google Scholar]
  • 16.Sinicrope FA, Yoon HH, Mahoney MR et. al (2014) Overall survival result and outcomes by KRAS, BRAF, and DNA mismatch repair in relation to primary tumor site in colon cancers from a randomized trial of adjuvant chemotherapy: NCCTG (Alliance) N0147. J Clin Oncol 32:5s (suppl; abstr 3525)
  • 17.Lanza G, Gafa R, Santini A, Maestri I, Guerzoni L, Cavazzini L. Immunohistochemical test for MLH1 and MSH2 expression predicts clinical outcome in stage II and III colorectal cancer patients. J Clin Oncol. 2006;24:2359–2367. doi: 10.1200/JCO.2005.03.2433. [DOI] [PubMed] [Google Scholar]
  • 18.Sinicrope FA, Rego RL, Halling KC, et al. Prognostic impact of microsatellite instability and DNA ploidy in human colon carcinoma patients. Gastroenterology. 2006;131:729–737. doi: 10.1053/j.gastro.2006.06.005. [DOI] [PubMed] [Google Scholar]
  • 19.Guastadisegni C, Colafranceschi M, Ottini L, Dogliotti E. Microsatellite instability as a marker of prognosis and response to therapy: a meta-analysis of colorectal cancer survival data. Eur J Cancer. 2010;46:2788–2798. doi: 10.1016/j.ejca.2010.05.009. [DOI] [PubMed] [Google Scholar]
  • 20.Venderbosch S, Nagtegaal ID, Maughan TS, et al (2014) Mismatch repair status and BRAF mutation status in metastatic colorectal cancer patients: a pooled analysis of the CAIRO, CAIRO2, COIN, and FOCUS studies. Clin Cancer Res 20:5322–5330 [DOI] [PMC free article] [PubMed]
  • 21.Ribic CM, Sargent DJ, Moore MJ, et al. Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J Med. 2003;349:247–257. doi: 10.1056/NEJMoa022289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Des Guetz G, Schischmanoff O, Nicolas P, Perret GY, Morere JF, Uzzan B. Does microsatellite instability predict the efficacy of adjuvant chemotherapy in colorectal cancer? A systematic review with meta-analysis. Eur J Cancer. 2009;45:1890–1896. doi: 10.1016/j.ejca.2009.04.018. [DOI] [PubMed] [Google Scholar]
  • 23.Hutchins G, Southward K, Handley K, et al. Value of mismatch repair, KRAS, and BRAF mutations in predicting recurrence and benefits from chemotherapy in colorectal cancer. J Clin Oncol. 2011;29:1261–1270. doi: 10.1200/JCO.2010.30.1366. [DOI] [PubMed] [Google Scholar]
  • 24.Sargent DJ, Shi Q, Yothers G et. al (2014) Prognostic impact of deficient mismatch repair (dMMR) in 7,803 stage II/III colon cancer (CC) patients (pts): a pooled individual pt data analysis of 17 adjuvant trials in the ACCENT database. J Clin Oncol 32:5s (suppl; abstr 3507)
  • 25.National Comprehensive Cancer Network (NCCN). Clinical practice guidelines in oncology: colon cancer. Version II. 2016. [DOI] [PubMed]
  • 26.Zaanan A, Flejou JF, Emile JF, et al. Defective mismatch repair status as a prognostic biomarker of disease-free survival in stage III colon cancer patients treated with adjuvant FOLFOX chemotherapy. Clin Cancer Res. 2011;17:7470–7478. doi: 10.1158/1078-0432.CCR-11-1048. [DOI] [PubMed] [Google Scholar]
  • 27.Zaanan A, Cuilliere-Dartigues P, Guilloux A, et al. Impact of p53 expression and microsatellite instability on stage III colon cancer disease-free survival in patients treated by 5-fluorouracil and leucovorin with or without oxaliplatin. Ann Oncol. 2010;21:772–780. doi: 10.1093/annonc/mdp383. [DOI] [PubMed] [Google Scholar]
  • 28.Kim ST, Lee J, Park SH, et al. Clinical impact of microsatellite instability in colon cancer following adjuvant FOLFOX therapy. Cancer Chemother Pharmacol. 2010;66:659–667. doi: 10.1007/s00280-009-1206-3. [DOI] [PubMed] [Google Scholar]
  • 29.Le DT, Uram JN, Wang H, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med. 2015;372:2509–2520. doi: 10.1056/NEJMoa1500596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Llosa NJ, Cruise M, Tam A, et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015;5:43–51. doi: 10.1158/2159-8290.CD-14-0863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Dudley JC, Lin MT, Le DT, Eshleman JR. Microsatellite Instability as a Biomarker for PD-1 Blockade. Clin Cancer Res. 2016;22:813–820. doi: 10.1158/1078-0432.CCR-15-1678. [DOI] [PubMed] [Google Scholar]
  • 32.Bourne HR, Sanders DA, McCormick F. The GTPase superfamily: conserved structure and molecular mechanism. Nature. 1991;349:117–127. doi: 10.1038/349117a0. [DOI] [PubMed] [Google Scholar]
  • 33.Amado RG, Wolf M, Peeters M, et al. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26:1626–1634. doi: 10.1200/JCO.2007.14.7116. [DOI] [PubMed] [Google Scholar]
  • 34.Martincorena I, Campbell PJ. Somatic mutation in cancer and normal cells. Science. 2015;349:1483–1489. doi: 10.1126/science.aab4082. [DOI] [PubMed] [Google Scholar]
  • 35.Van Cutsem E, Kohne CH, Hitre E, et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 2009;360:1408–1417. doi: 10.1056/NEJMoa0805019. [DOI] [PubMed] [Google Scholar]
  • 36.Lievre A, Bachet JB, Le Corre D, et al. KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 2006;66:3992–3995. doi: 10.1158/0008-5472.CAN-06-0191. [DOI] [PubMed] [Google Scholar]
  • 37.Loupakis F, Ruzzo A, Cremolini C, et al. KRAS codon 61, 146 and BRAF mutations predict resistance to cetuximab plus irinotecan in KRAS codon 12 and 13 wild-type metastatic colorectal cancer. Br J Cancer. 2009;101:715–721. doi: 10.1038/sj.bjc.6605177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Douillard JY, Oliner KS, Siena S, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369:1023–1034. doi: 10.1056/NEJMoa1305275. [DOI] [PubMed] [Google Scholar]
  • 39.De Roock W, Claes B, Bernasconi D, et al. 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. doi: 10.1016/S1470-2045(10)70130-3. [DOI] [PubMed] [Google Scholar]
  • 40.Sorich MJ, Wiese MD, Rowland A, Kichenadasse G, McKinnon RA, Karapetis CS. Extended RAS mutations and anti-EGFR monoclonal antibody survival benefit in metastatic colorectal cancer: a meta-analysis of randomized, controlled trials. Ann Oncol. 2015;26:13–21. doi: 10.1093/annonc/mdu378. [DOI] [PubMed] [Google Scholar]
  • 41.Bokemeyer C, Kohne CH, Ciardiello F, et al. FOLFOX4 plus cetuximab treatment and RAS mutations in colorectal cancer. Eur J Cancer. 2015;51:1243–1252. doi: 10.1016/j.ejca.2015.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Van Cutsem E, Lenz HJ, Kohne CH, et al. Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J Clin Oncol. 2015;33:692–700. doi: 10.1200/JCO.2014.59.4812. [DOI] [PubMed] [Google Scholar]
  • 43.Schwartzberg LS, Rivera F, Karthaus M, et al. PEAK: a randomized, multicenter phase II study of panitumumab plus modified fluorouracil, 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;32:2240–2247. doi: 10.1200/JCO.2013.53.2473. [DOI] [PubMed] [Google Scholar]
  • 44.Lenz HJ, Niedzwiecki D, Innocenti F et. al (2014) CALGB/SWOG 80405: phase III trial of FOLFIRI or mFOLFOX6 with bevacizumab or cetuximab for patients with expanded RAS analyses in untreated metastatic adenocarcinoma of the colon or rectum. Ann Oncol 25 (suppl 4; abstr 5010)
  • 45.Allegra CJ, Rumble RB, Hamilton SR, Mangu PB, Roach N, Hantel A, Schilsky RL. Extended RAS gene mutation testing in metastatic colorectal carcinoma to predict response to anti-epidermal growth factor receptor monoclonal antibody therapy: American Society of Clinical Oncology Provisional Clinical Opinion Update 2015. J Clin Oncol. 2016;34:179–185. doi: 10.1200/JCO.2015.63.9674. [DOI] [PubMed] [Google Scholar]
  • 46.Ren J, Li G, Ge J, Li X, Zhao Y. Is K-ras gene mutation a prognostic factor for colorectal cancer: a systematic review and meta-analysis. Dis Colon Rectum. 2012;55:913–923. doi: 10.1097/DCR.0b013e318251d8d9. [DOI] [PubMed] [Google Scholar]
  • 47.Tejpar S, Celik I, Schlichting M, Sartorius U, Bokemeyer C, Van Cutsem E. Association of KRAS G13D tumor mutations with outcome in patients with metastatic colorectal cancer treated with first-line chemotherapy with or without cetuximab. J Clin Oncol. 2012;30:3570–3577. doi: 10.1200/JCO.2012.42.2592. [DOI] [PubMed] [Google Scholar]
  • 48.Rowland A, Dias MM, Wiese MD, Kichenadasse G, McKinnon RA, Karapetis CS, Sorich MJ. Meta-analysis comparing the efficacy of anti-EGFR monoclonal antibody therapy between KRAS G13D and other KRAS mutant metastatic colorectal cancer tumours. Eur J Cancer. 2016;55:122–130. doi: 10.1016/j.ejca.2015.11.025. [DOI] [PubMed] [Google Scholar]
  • 49.De Roock W, Jonker DJ, Di Nicolantonio F, et al. Association of KRAS p.G13D mutation with outcome in patients with chemotherapy-refractory metastatic colorectal cancer treated with cetuximab. JAMA. 2010;304:1812–1820. doi: 10.1001/jama.2010.1535. [DOI] [PubMed] [Google Scholar]
  • 50.Schirripa M, Loupakis F, Lonardi S, Cremolini C, Bergamo F, Zagonel V, Falcone A. Phase II study of single-agent cetuximab in KRAS G13D mutant metastatic colorectal cancer. Ann Oncol. 2015;26:2503. doi: 10.1093/annonc/mdv233.196. [DOI] [PubMed] [Google Scholar]
  • 51.E. Segelov, S. Thavaneswaran, P. Waring et. al (2015) The AGITG ICECREAM study: the irinotecan cetuximab evaluation and cetuximab response evaluation amongst patients with a G13D mutation-analysis of outcomes in patients with refractory metastatic colorectal cancer harbouring the KRAS G13D mutation. ESMO European Cancer Congress:32LBA
  • 52.Diaz LA, Jr, Williams RT, Wu J, et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012;486:537–540. doi: 10.1038/nature11219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Bertotti A, Papp E, Jones S, et al. The genomic landscape of response to EGFR blockade in colorectal cancer. Nature. 2015;526:263–267. doi: 10.1038/nature14969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.White MF. Insulin signaling in health and disease. Science. 2003;302:1710–1711. doi: 10.1126/science.1092952. [DOI] [PubMed] [Google Scholar]
  • 55.Montagut C, Dalmases A, Bellosillo B, et al. Identification of a mutation in the extracellular domain of the Epidermal Growth Factor Receptor conferring cetuximab resistance in colorectal cancer. Nat Med. 2012;18:221–223. doi: 10.1038/nm.2609. [DOI] [PubMed] [Google Scholar]
  • 56.Kearns JD, Bukhalid R, Sevecka M, et al. Enhanced Targeting of the EGFR Network with MM-151, an Oligoclonal Anti-EGFR Antibody Therapeutic. Mol Cancer Ther. 2015;14:1625–1636. doi: 10.1158/1535-7163.MCT-14-0772. [DOI] [PubMed] [Google Scholar]
  • 57.Arena S, Siravegna G, Mussolin B, et al. MM-151 overcomes acquired resistance to cetuximab and panitumumab in colorectal cancers harboring EGFR extracellular domain mutations. Sci Transl Med. 2016;8:324ra14. doi: 10.1126/scitranslmed.aad5640. [DOI] [PubMed] [Google Scholar]
  • 58.Pedersen MW, Jacobsen HJ, Koefoed K, Hey A, Pyke C, Haurum JS, Kragh M. Sym004: a novel synergistic anti-epidermal growth factor receptor antibody mixture with superior anticancer efficacy. Cancer Res. 2010;70:588–597. doi: 10.1158/0008-5472.CAN-09-1417. [DOI] [PubMed] [Google Scholar]
  • 59.Sanchez-Martin FJ, Bellosillo B, Gelabert M et. al (2016) The first-in-class anti-EGFR antibody mixture Sym004 overcomes cetuximab-resistance mediated by EGFR extracellular domain mutations in colorectal cancer. Clin Cancer Res [DOI] [PubMed]
  • 60.Li P, Wood K, Mamon H, Haser W, Roberts T. Raf-1: a kinase currently without a cause but not lacking in effects. Cell. 1991;64:479–482. doi: 10.1016/0092-8674(91)90228-Q. [DOI] [PubMed] [Google Scholar]
  • 61.Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–954. doi: 10.1038/nature00766. [DOI] [PubMed] [Google Scholar]
  • 62.Montagut C, Settleman J. Targeting the RAF-MEK-ERK pathway in cancer therapy. Cancer Lett. 2009;283:125–134. doi: 10.1016/j.canlet.2009.01.022. [DOI] [PubMed] [Google Scholar]
  • 63.Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat Rev Cancer. 2003;3:459–465. doi: 10.1038/nrc1097. [DOI] [PubMed] [Google Scholar]
  • 64.Rajagopalan H, Bardelli A, Lengauer C, Kinzler KW, Vogelstein B, Velculescu VE. Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature. 2002;418:934. doi: 10.1038/418934a. [DOI] [PubMed] [Google Scholar]
  • 65.Barault L, Veyrie N, Jooste V, et al. Mutations in the RAS-MAPK, PI(3)K (phosphatidylinositol-3-OH kinase) signaling network correlate with poor survival in a population-based series of colon cancers. Int J Cancer. 2008;122:2255–2259. doi: 10.1002/ijc.23388. [DOI] [PubMed] [Google Scholar]
  • 66.Clancy C, Burke JP, Kalady MF, et al (2013) BRAF mutation is associated with distinct clinicopathological characteristics in colorectal cancer: a systematic review and meta-analysis. Colorectal Dis 15:e711–8 [DOI] [PubMed]
  • 67.Van Cutsem E, Kohne CH, Lang I, et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol. 2011;29:2011–2019. doi: 10.1200/JCO.2010.33.5091. [DOI] [PubMed] [Google Scholar]
  • 68.Bokemeyer C, Bondarenko I, Hartmann JT, et al. Efficacy according to biomarker status of cetuximab plus FOLFOX-4 as first-line treatment for metastatic colorectal cancer: the OPUS study. Ann Oncol. 2011;22:1535–1546. doi: 10.1093/annonc/mdq632. [DOI] [PubMed] [Google Scholar]
  • 69.Maughan TS, Adams RA, Smith CG, et al. Addition of cetuximab to oxaliplatin-based first-line combination chemotherapy for treatment of advanced colorectal cancer: results of the randomised phase 3 MRC COIN trial. Lancet. 2011;377:2103–2114. doi: 10.1016/S0140-6736(11)60613-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Stintzing S, Jung A, Rossius L et. al (2014) Mutations within the EGFR signaling pathway: influence on efficacy in FIRE-3—a randomized phase III study of FOLFIRI plus cetuximab or bevacizumab as first-line treatment for wild-type (WT) KRAS (exon 2) metastatic colorectal cancer (mCRC) patients. J Clin Oncol 32 (suppl 3; abstr 445) :
  • 71.Peeters M, Oliner KS, Price TJ, et al. Analysis of KRAS/NRAS Mutations in a Phase III Study of Panitumumab with FOLFIRI Compared with FOLFIRI Alone as Second-line Treatment for Metastatic Colorectal Cancer. Clin Cancer Res. 2015;21:5469–5479. doi: 10.1158/1078-0432.CCR-15-0526. [DOI] [PubMed] [Google Scholar]
  • 72.Seymour MT, Brown SR, Middleton G, et al. Panitumumab and irinotecan versus irinotecan alone for patients with KRAS wild-type, fluorouracil-resistant advanced colorectal cancer (PICCOLO): a prospectively stratified randomised trial. Lancet Oncol. 2013;14:749–759. doi: 10.1016/S1470-2045(13)70163-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Karapetis CS, Jonker D, Daneshmand M, et al. PIK3CA, BRAF, and PTEN status and benefit from cetuximab in the treatment of advanced colorectal cancer--results from NCIC CTG/AGITG CO.17. Clin Cancer Res. 2014;20:744–753. doi: 10.1158/1078-0432.CCR-13-0606. [DOI] [PubMed] [Google Scholar]
  • 74.Patterson SD, Peeters M, Siena S, Van Cutsem E, Humblet Y, Van Laethem J-L, et al (2013) Comprehensive analysis of KRAS and NRAS mutations as predictive biomarkers for single agent panitumumab (pmab) response in a randomized, phase 3 metastatic colorectal cancer (mCRC) study (20020408) J Clin Oncol. 31 (supple; abstr 3617)
  • 75.Roth AD, Tejpar S, Delorenzi M, et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J Clin Oncol. 2010;28:466–474. doi: 10.1200/JCO.2009.23.3452. [DOI] [PubMed] [Google Scholar]
  • 76.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;53:852–864. doi: 10.3109/0284186X.2014.895036. [DOI] [PubMed] [Google Scholar]
  • 77.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;51:587–594. doi: 10.1016/j.ejca.2015.01.054. [DOI] [PubMed] [Google Scholar]
  • 78.Kopetz S, Desai J, Chan E, et al. Phase II Pilot Study of Vemurafenib in Patients With Metastatic BRAF-Mutated Colorectal Cancer. J Clin Oncol. 2015;33:4032–4038. doi: 10.1200/JCO.2015.63.2497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Yang H, Higgins B, Kolinsky K, et al. Antitumor activity of BRAF inhibitor vemurafenib in preclinical models of BRAF-mutant colorectal cancer. Cancer Res. 2012;72:779–789. doi: 10.1158/0008-5472.CAN-11-2941. [DOI] [PubMed] [Google Scholar]
  • 80.Corcoran RB, Ebi H, Turke AB, et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2012;2:227–235. doi: 10.1158/2159-8290.CD-11-0341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Oikonomou E, Koc M, Sourkova V, Andera L, Pintzas A. Selective BRAFV600E inhibitor PLX4720, requires TRAIL assistance to overcome oncogenic PIK3CA resistance. PLoS One. 2011;6:e21632. doi: 10.1371/journal.pone.0021632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Hyman DM, Puzanov I, Subbiah V, et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N Engl J Med. 2015;373:726–736. doi: 10.1056/NEJMoa1502309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Corcoran RB, Atreya CE, Falchook GS, et al. Combined BRAF and MEK Inhibition With Dabrafenib and Trametinib in BRAF V600-Mutant Colorectal Cancer. J Clin Oncol. 2015;33:4023–4031. doi: 10.1200/JCO.2015.63.2471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Yaeger R, Cercek A, O’Reilly EM, et al. Pilot trial of combined BRAF and EGFR inhibition in BRAF-mutant metastatic colorectal cancer patients. Clin Cancer Res. 2015;21:1313–1320. doi: 10.1158/1078-0432.CCR-14-2779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Tabernero J, van Geel R, Bendell JC et. al (2014) Phase I study of the selective BRAFV600 inhibitor encorafenib (LGX818) combined with cetuximab and with or without the a-specific PI3K inhibitor alpelisib (BYL719) in patients with advanced BRAF mutant colorectal cancer. Eur J Cancer 50:6s (suppl; abstr LBA 11) :
  • 86.Atreya CE, Van Cutsem E, Bendell JC et. al (2015) Updated efficacy of the MEK inhibitor trametinib (T), BRAF inhibitor dabrafenib (D), and anti-EGFR antibody panitumumab (P) in patients (pts) with BRAF V600E mutated (BRAFm) metastatic colorectal cancer (mCRC). J Clin Oncol 33 (suppl; abstr 103)
  • 87.David S. Hong, Van Karlyle Morris, Siqing Fu et. al (2014) Phase 1B study of vemurafenib in combination with irinotecan and cetuximab in patients with BRAF-mutated advanced cancers and metastatic colorectal cancer. J Clin Oncol 32:5s (suppl; abstr 3516)
  • 88.Cremolini C, Loupakis F, Antoniotti C, et al. FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab as first-line treatment of patients with metastatic colorectal cancer: updated overall survival and molecular subgroup analyses of the open-label, phase 3 TRIBE study. Lancet Oncol. 2015;16:1306–1315. doi: 10.1016/S1470-2045(15)00122-9. [DOI] [PubMed] [Google Scholar]
  • 89.Loupakis F, Cremolini C, Salvatore L, et al. FOLFOXIRI plus bevacizumab as first-line treatment in BRAF mutant metastatic colorectal cancer. Eur J Cancer. 2014;50:57–63. doi: 10.1016/j.ejca.2013.08.024. [DOI] [PubMed] [Google Scholar]
  • 90.Engelman JA. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer. 2009;9:550–562. doi: 10.1038/nrc2664. [DOI] [PubMed] [Google Scholar]
  • 91.Samuels Y, Wang Z, Bardelli A, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304:554. doi: 10.1126/science.1096502. [DOI] [PubMed] [Google Scholar]
  • 92.Liao X, Lochhead P, Nishihara R, et al. Aspirin use, tumor PIK3CA mutation, and colorectal-cancer survival. N Engl J Med. 2012;367:1596–1606. doi: 10.1056/NEJMoa1207756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Domingo E, Church DN, Sieber O, et al. Evaluation of PIK3CA mutation as a predictor of benefit from nonsteroidal anti-inflammatory drug therapy in colorectal cancer. J Clin Oncol. 2013;31:4297–4305. doi: 10.1200/JCO.2013.50.0322. [DOI] [PubMed] [Google Scholar]
  • 94.Kim TM, Lee SH, Chung YJ. Clinical applications of next-generation sequencing in colorectal cancers. World J Gastroenterol. 2013;19:6784–6793. doi: 10.3748/wjg.v19.i40.6784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Zhang B, Wang J, Wang X, et al. Proteogenomic characterization of human colon and rectal cancer. Nature. 2014;513:382–387. doi: 10.1038/nature13438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Hudecova I. Digital PCR analysis of circulating nucleic acids. Clin Biochem. 2015;48:948–956. doi: 10.1016/j.clinbiochem.2015.03.015. [DOI] [PubMed] [Google Scholar]
  • 97.Li M, Diehl F, Dressman D, Vogelstein B, Kinzler KW. BEAMing up for detection and quantification of rare sequence variants. Nat Methods. 2006;3:95–97. doi: 10.1038/nmeth850. [DOI] [PubMed] [Google Scholar]
  • 98.Heitzer E, Ulz P, Geigl JB. Circulating tumor DNA as a liquid biopsy for cancer. Clin Chem. 2015;61:112–123. doi: 10.1373/clinchem.2014.222679. [DOI] [PubMed] [Google Scholar]
  • 99.Frattini M, Gallino G, Signoroni S, et al. Quantitative analysis of plasma DNA in colorectal cancer patients: a novel prognostic tool. Ann N Y Acad Sci. 2006;1075:185–190. doi: 10.1196/annals.1368.025. [DOI] [PubMed] [Google Scholar]
  • 100.Frattini M, Balestra D, Verderio P, et al. Reproducibility of a semiquantitative measurement of circulating DNA in plasma from neoplastic patients. J Clin Oncol. 2005;23:3163–3164. doi: 10.1200/JCO.2005.05.430. [DOI] [PubMed] [Google Scholar]
  • 101.Spindler KL, Pallisgaard N, Andersen RF, Brandslund I, Jakobsen A. Circulating free DNA as biomarker and source for mutation detection in metastatic colorectal cancer. PLoS One. 2015;10:e0108247. doi: 10.1371/journal.pone.0108247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Tabernero J, Lenz HJ, Siena S, et al. Analysis of circulating DNA and protein biomarkers to predict the clinical activity of regorafenib and assess prognosis in patients with metastatic colorectal cancer: a retrospective, exploratory analysis of the CORRECT trial. Lancet Oncol. 2015;16:937–948. doi: 10.1016/S1470-2045(15)00138-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Vasioukhin V, Anker P, Maurice P, Lyautey J, Lederrey C, Stroun M. Point mutations of the N-ras gene in the blood plasma DNA of patients with myelodysplastic syndrome or acute myelogenous leukaemia. Br J Haematol. 1994;86:774–779. doi: 10.1111/j.1365-2141.1994.tb04828.x. [DOI] [PubMed] [Google Scholar]
  • 104.Taly V, Pekin D, Benhaim L, et al. Multiplex picodroplet digital PCR to detect KRAS mutations in circulating DNA from the plasma of colorectal cancer patients. Clin Chem. 2013;59:1722–1731. doi: 10.1373/clinchem.2013.206359. [DOI] [PubMed] [Google Scholar]
  • 105.Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra24. doi: 10.1126/scitranslmed.3007094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Mouliere F, El Messaoudi S, Gongora C, et al. Circulating Cell-Free DNA from Colorectal Cancer Patients May Reveal High KRAS or BRAF Mutation Load. Transl Oncol. 2013;6:319–328. doi: 10.1593/tlo.12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Thierry AR, Mouliere F, El Messaoudi S, et al. Clinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNA. Nat Med. 2014;20:430–435. doi: 10.1038/nm.3511. [DOI] [PubMed] [Google Scholar]
  • 108.Misale S, Yaeger R, Hobor S, et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012;486:532–536. doi: 10.1038/nature11156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.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;21:795–801. doi: 10.1038/nm.3870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Esposito A, Bardelli A, Criscitiello C, et al. Monitoring tumor-derived cell-free DNA in patients with solid tumors: clinical perspectives and research opportunities. Cancer Treat Rev. 2014;40:648–655. doi: 10.1016/j.ctrv.2013.10.003. [DOI] [PubMed] [Google Scholar]

Articles from Indian Journal of Surgical Oncology are provided here courtesy of Springer

RESOURCES