118
Views
13
CrossRef citations to date
0
Altmetric
Review

Molecular characterization and biomarker identification in colorectal cancer: Toward realization of the precision medicine dream

Pages 5895-5908 | Published online: 19 Nov 2018

Abstract

Colorectal cancer (CRC) is a major public health problem, both in the USA and globally. Over the past 20 years, significant advances have been made in the treatment of patients with metastatic CRC (mCRC). Recent efforts in the field of biomarkers have focused on the development of molecular diagnostics to define the subset of patients with mCRC that is likely to derive most benefit from anti-EGFR therapy. Herein, we review the recent advancements in molecular stratification of CRC and the role of current as well as emerging biomarkers in this disease. It is now clear that the presence of activating mutations in the KRAS and NRAS genes serves as reliable predictive markers for resistance to anti-EGFR therapy in mCRC. It is also clear that further improvements in the survival of mCRC patients will probably be made possible only with identification of new predictive molecular biomarkers and their evaluation using rational and innovative clinical trials. The recent advances in DNA sequencing technology and “omics”-based approaches have provided promising new strategies for the development of novel molecular biomarkers in this disease.

Introduction

Colorectal cancer (CRC) is a major public health problem, both in the USA and globally. In the USA, it is the second leading cause of cancer-related mortality, and in 2018, it is estimated that nearly 50,630 deaths will be attributed to this disease.Citation1 When metastatic disease is diagnosed, CRC is usually associated with poor prognosis with 5-year survival rates less than 15%.Citation1 With the advances in the treatment of metastatic CRC (mCRC) over the past 20 years, the median overall survival (OS) has been steadily increasing, and it has now reached beyond 40 months in a select group of patients.Citation2Citation4 This improvement in OS has been made possible with the identification of new agents and incorporation of targeted biologic agents into the cytotoxic chemotherapy regimens used for treatment of mCRC patients.Citation5,Citation6 Although the progress made thus far is encouraging, the existing treatment paradigm usually employs a “one-size-fits-all” approach, which translates into demonstrable clinical benefit from any given chemotherapeutic regimen in only a small subset of treated patients.

It is now being increasingly realized that CRC is not a single disease entity, but a heterogeneous group of tumors, both at the intertumoral and intratumoral levels. Given the complex molecular makeup of these tumors, significant efforts have been focused in recent years on identifying potential molecular markers that would provide further information regarding tumor response to anticancer therapies, which in turn would help personalize treatment for mCRC. “Predictive” biomarkers identify patients who are most likely to benefit from a specific treatment, and they can, therefore, play a critical role in guiding treatment decisions. Research in the identification of predictive biomarkers for mCRC has been directed toward the targets that are downstream in the EGFR signal transduction cascade, including mutations in the KRAS, NRAS, BRAF, and PIK3CA genes, and PTEN protein expression. “Prognostic” biomarkers, on the other hand, provide information about the tumor’s aggressiveness, and they help distinguish patients based on their expected disease outcome, independent of the treatment received. It is believed that molecular stratification of CRC can identify biomarkers to provide such additional prognostic and predictive information, over and beyond the conventional TNM staging system. Such validated biomarkers will also help optimize efficacy, minimize toxicity, and reduce treatment costs.

This article will review the recent advancements in molecular stratification of CRC and the role of current as well as emerging biomarkers in this disease.

Subtypes of colorectal tumors

CRC subtyping based on traditional oncogenic pathways

CRC has been theorized as a stepwise model of genetic and epigenetic events by Fearon and Vogelstein, nearly three decades ago.Citation7 It usually begins with the inactivation of APC tumor suppressor gene, resulting in increased Wnt pathway signaling.Citation8 Thereafter, depending on the mechanism of underlying genomic alterations, CRC develops through one of the three distinct molecular pathways – chromosomal instability (CIN), microsatellite instability (MSI), and CpG island methylator phenotype (CIMP) pathways. Of these, CIN pathway is the most commonly occurring (85%), and it involves widespread loss of heterozygosity and gross chromosomal abnormalities leading to hypermutability. MSI and CIMP pathways result in development of colorectal tumors due to a defective DNA mismatch repair (MMR) system. MMR system is a multiprotein system responsible for proofreading and eliminating any nucleotide mismatches that escape the checking system of DNA polymerase during DNA replication process. While the MSI pathway is characterized by the presence of genetic or sporadic mutations in the MMR genes, the CIMP pathway involves epigenetic silencing of the MMR gene MLH1 by causing hypermethylation of the CpG dinucleotide sequences located in the gene’s promoter region.

Transcriptome-based CRC subtypes

The international Colorectal Cancer Subtyping Consortium studied six independent transcriptome-based classification systems of CRC, and the resulting collection of 27 subtypes were coalesced into four distinct Consensus Molecular Subtype (CMS) groups.Citation9 Transcriptional molecular signatures refine disease subclassification and help to identify subtypes with distinct biologic behavior. CMS1 group (MSI immune subtype, 14%) consists of hypermutated MSI tumors with strong immune activation. These tumors have overexpression of proteins involved in DNA damage repair consistent with defective DNA mismatch repair. BRAF mutations occur frequently in these tumors, and an increased expression of genes associated with diffuse immune infiltrate is noted. CMS2 (canonical subtype, 37%) group is characterized by epithelial differentiation and chromosomally unstable tumors. The tumors in this group display high somatic copy number alterations, and upregulation of WNT and MYC downstream targets. CMS3 (metabolic subtype, 13%) tumors show epithelial differentiation and marked metabolic dysregulation. Nearly 30% of these tumors are hypermutated as well. There is a higher prevalence of CIMP and KRAS activating mutations in CMS3. CMS4 (mesenchymal subtype, 23%) consists of tumors with prominent TGF-β activation, stromal invasion, and angiogenesis.

Established biomarkers in current clinical practice

RAS (KRAS and NRAS) mutations

The RAS gene family is composed of three well-known genes: KRAS, NRAS, and HRAS.Citation10 KRAS mutations are estimated to be present in ~30%–45% of CRC patients and include single-point mutations in exons 2, 3, and 4.Citation11 Another 5% of CRC tumors harbor activating mutations in NRAS, in the same exons as KRAS. These genes encode GTP/GDP binding protein, which is a key player in the MAP kinase signal transduction pathway.

The role of RAS mutations as a negative predictive marker for anti-EGFR therapy in the treatment of mCRC has been confirmed in multiple clinical trials and is now well established.Citation12Citation18 Several systematic reviews and meta-analyses have also confirmed that among the anti-EGFR monoclonal antibody-treated mCRC patients, KRAS wild-type patients perform better than KRAS-mutant population; and that among the KRAS wild-type patients, treatment with anti-EGFR monoclonal antibody-containing regimen is associated with better response than chemotherapy alone.Citation19Citation22 Studies have also clearly established that in contrast to the KRAS wild-type tumors, the addition of anti-EGFR therapy to chemotherapy does not improve the outcomes in KRAS-mutant population and is even potentially harmful.Citation16,Citation17

It is, however, worth noting that in these earlier studies, only mutations in exon 2 of the KRAS gene were assessed. More recent data suggest that in addition to mutations in KRAS exon 2, other RAS mutations in KRAS exons 3 and 4, and NRAS exons 2, 3, and 4 are also associated with resistance to anti-EGFR therapy in mCRC.Citation23Citation25 The most convincing evidence establishing the predictive value of “expanded” RAS testing is provided by the meta-analysis of nine randomized clinical trials consisting of nearly 6,000 patients.Citation24 In this meta-analysis, the tumors with new RAS mutations (KRAS exons 3 and 4, NRAS exons 2, 3, and 4) were compared with tumors without any RAS mutation, with respect to anti-EGFR treatment-related progression-free survival (PFS) and OS benefit. Patients with tumors that were KRAS exon 2 wild-type, but harbored RAS mutations in KRAS exons 3 and 4, or NRAS exons 2, 3, and 4 had significantly inferior survival outcomes compared to those without any RAS mutation.

Taken together, it is now well established that presence of any activating mutation in KRAS or NRAS predicts for resistance to anti-EGFR therapy in mCRC. A recent guideline on molecular testing for CRC issued jointly by the American Society for Clinical Pathology (ASCP), College of American Pathologists (CAP), Association of Molecular Pathology (AMP), and American Society of Clinical Oncology (ASCO), recommends expanded RAS testing, which includes mutational analysis of KRAS and NRAS codons 12 and 13 of exon 2, codons 59 and 61 of exon 3, and codons 117 and 146 of exon 4 for mCRC patients being considered for anti-EGFR therapy.Citation26 The mCRC patients whose tumors harbor these mutations should not receive anti-EGFR therapy as they will not derive clinical benefit from such treatment. Perhaps, a greater concern is that such patients are likely to experience an even worse clinical outcome. It is also worth noting that the absence of RAS mutation is not the most accurate predictor of tumor response to anti-EGFR monoclonal antibody, as only 40%–50% of patients with KRAS/NRAS wild-type disease will ultimately respond to such therapy.

In contrast to the predictive value, the role of KRAS mutation in CRC as an “independent” prognostic marker has yielded inconsistent results. In one of the early efforts, several researchers with information about the KRAS tumor genotype and outcome of CRC patients came together to form a collaborative database.Citation27 The data on 3,439 patients recruited from 42 centers in 21 countries were entered into a multivariate analysis, and showed that KRAS mutation in codon 12 was associated with an increased risk of tumor recurrence and worse survival. In another retrospective analysis of tumor samples obtained from CRC patients treated with chemotherapy (without anti-EGFR monoclonal antibody) in the MRC FOCUS trial, the presence of mutation in either KRAS (codons 12, 13, and 61) or BRAF was associated with significantly decreased survival when compared to the wild-type tumors.Citation28 In contrast, the CO.17 studyCitation13 and the study by Kim et alCitation29 did not demonstrate any significant survival difference between the wild-type KRAS and mutant-KRAS subgroups. In a systematic review with meta-analysis also, KRAS mutation status did not correlate with prognosis in CRC patients.Citation30 Due to the inconsistencies in data, KRAS mutational status cannot be used reliably as an independent prognostic marker for mCRC patients at this time.

provides a summary of established and emerging biomarkers in CRC and their potential role in clinical practice.

Table 1 Summary of established and emerging biomarkers in the management of colorectal cancer

BRAF mutations

BRAF is a member of the RAF proto-oncogene family, and a key player of the RAS/RAF/MAP kinase pathway. Activating point mutations in BRAF are present in ~5%–10% of CRC.Citation31 This mutation results in constitutive activation of BRAF kinase, which then leads to activation of the downstream RAS/RAF/MAP kinase pathway.

Several independent studies have consistently demonstrated that the presence of BRAFV600E mutations is associated with poor prognosis in patients with mCRC.Citation23,Citation31Citation35 As the population of BRAF-mutant mCRC patients is relatively small due to the low mutation prevalence, several meta-analyses and systematic reviews have been performed to provide more conclusive evidence on the prognostic and predictive value of BRAF mutation as it relates to anti-EGFR therapy.Citation36Citation41 In one of the largest meta-analyses of 21 clinical trials that included 5,229 mCRC patients treated with anti-EGFR monoclonal antibody, 343 patients displayed BRAF mutations out of 4,616 (7.4%) patients with known BRAF status.Citation38 Patients with BRAF wild-type showed an improved PFS (HR, 0.38; 95% CI 0.29–0.51) and an improved OS (HR, 0.35; 95% CI 0.29–0.42), compared to the BRAF-mutant group. These results indicate that the presence of BRAF mutation is associated with poor prognosis in mCRC patients.

In mCRC, the presence of BRAF mutation has been suggested as a negative predictor of response toward anti-EGFR therapy, based on the results from the retrospective study by Di Nicolantonio et alCitation32 and the PICCOLO trial.Citation42 On the other hand, in a meta-analysis of eight randomized clinical trials that included RAS wild-type mCRC patients treated with anti-EGFR therapy, there was lack of statistically significant difference in OS (P=0.43) between the BRAF-mutant and BRAF wild-type subgroups.Citation40 In another meta-analysis, the addition of anti-EGFR therapy in the BRAF-mutant advanced CRC patients did not significantly improve the survival compared to the control regimens.Citation41

The available clinical evidence suggests that the presence of BRAF mutations has a clear prognostic value in mCRC as it confers a significantly worse clinical outcome. Recent guidelines recommend that BRAFV600E mutational analysis should be performed in all mCRC patients for prognostic stratification.Citation26 However, the available data on BRAF mutations as a predictive marker for anti-EGFR therapy are limited due to being largely retrospective in nature. The relatively low prevalence of BRAF mutation in colon cancer poses the greatest challenge in addressing this question prospectively. Therefore, at the present time, there is insufficient evidence to recommend BRAF mutation testing on colorectal tumors to predict response to anti-EGFR therapy.Citation26

MSI and tumor mutational burden

The loss of MMR proteins leads to MMR deficiency, and these tumors are also referred to as microsatellite instability-high (MSI-Hi) tumors. In fact, MSI is considered as the molecular fingerprint of a deficient MMR system. The Cancer Genome Atlas (TCGA) project reported that ~16% of all CRCs are hypermutated or MSI-Hi.Citation43 The majority of MSI-Hi CRCs (11%–13%) are caused by somatic changes, either resulting from epigenetic silencing of the MLH1 gene in the majority of cases or due to somatic mutation in any of the four most common MMR genes (MLH1, MSH2, MSH6, and PMS2) or POLE gene.Citation43Citation45 The remaining 3%–5% MSI-Hi CRCs are caused by germline mutation in the MMR genes, and these patients are referred to as having the hereditary nonpolyposis CRC (Lynch syndrome).Citation44Citation47

MSI status has been evaluated as a prognostic and predictive biomarker in CRC. MSI-Hi CRCs are more often located in the proximal colon, are poorly differentiated, and have mucinous or medullary histology. These tumors are also considered highly immunogenic.Citation48 It is now understood that the length variations in the microsatellites of coding sequence in MSI-Hi tumors lead to frame-shift mutations. The high mutational burden associated with these tumors results in formation of tumor-specific neoantigens and subsequent T-cell infiltration, which suggests presence of a robust anti-tumoral immune response.Citation48 This explains the observation that MSI-Hi is associated with favorable prognosis in patients with early stage colon cancer.Citation49 It is now recommended that MMR status should be checked in patients with CRC for prognostic stratification.Citation26

MSI status also has predictive value, both in early stage and advanced CRC.Citation50 The presence of MSI-Hi has been shown to be predictive for lack of efficacy toward 5-FU adjuvant chemotherapy in early-stage colon cancer.Citation51 Therefore, adjuvant 5-FU should be avoided for stage II MSI-Hi colon cancer. In patients with advanced CRC, the data from early clinical trials suggest that the presence of MSI-Hi status is a positive predictor of durable tumor response to anti-programmed death-1 (PD-1) immune-checkpoint therapy.Citation52Citation55 A Phase II study evaluated the clinical efficacy of anti-PD-1 agent pembrolizumab in patients with chemo-refractory mCRC, with and without MMR deficiency.Citation54 In this study, the MSI-Hi CRCs were found to have encouraging responses to pembrolizumab. The overall response rate (ORR) was 50% for MSI-Hi CRC and 0% for microsatellite stable (MSS) CRC.Citation55 The median OS was not reached in the MSI-Hi CRC cohort, as opposed to 6 months in the MSS CRC cohort (HR, 0.247; P=0.001). Based on these encouraging results, a phase III study (KEYNOTE-177, NCT02563002) of pembrolizumab in MSI-Hi advanced CRC is ongoing to confirm these early observations.

Similarly, in another phase II study (CheckMate-142), nivolumab monotherapy was shown to provide clinical benefit (ORR, 31%; 12-month PFS, 50%; 12-month OS, 73%) in previously treated patients with MSI-Hi mCRC patients.Citation56 Nivolumab was also combined with the CTLA-4 inhibitor ipilimumab to synergistically promote T-cell antitumor activity and further improve the outcomes among MSI-Hi mCRC patients.Citation57 In the nivolumab plus ipilimumab cohort of the CheckMate-142 study, patients received nivolumab plus ipilimumab every 3 weeks for four doses, followed by nivolumab once every 2 weeks. Among the 119 patients treated with this combination, ORR was 55% at a median follow-up of 13.4 months. The 12-month PFS and OS rates were 71% and 85%, respectively. Based on these encouraging results, the Food and Drug Administration granted accelerated approval to the combination of nivolumab and ipilimumab in MSI-Hi mCRC patients that have progressed beyond first-line chemotherapy.

Recent evidence suggests that the presence of a high tumor mutational load might be a more important and accurate predictive marker of response to immunotherapy than the tumor’s MSI status itself. This is illustrated by the fact that PD-1 immunotherapy has shown efficacy in colorectal tumors carrying the POLE mutation.Citation58 This type of tumor is classified as an MSS tumor but is characterized by the presence of very high mutational burden and tumor-infiltrating lymphocytes in its microenvironment.

Gene expression profiling-based recurrence score assays

Multigene expression signatures have been developed for use as biomarkers in early stage colon cancer, such as stage II disease where the role of adjuvant chemotherapy is still unclear. Examples of gene expression signatures that can guide CRC prognosis include Oncotype DX and ColoPrint assays. The Oncotype DX is a 12-gene RT-PCR-based recurrence score assay that was shown to have prognostic value in patients with stage II colon cancer, independent of the traditional clinicopathologic factors.Citation59,Citation60 ColoPrint is an 18-gene microarray-based assay that differentiates stage II tumors into low and high risk of recurrence.Citation61 Stage II colon cancer patients who were identified as having low-risk disease based on the ColoPrint assay had significantly improved disease-free survival (DFS) than those in the high-risk group (87.6% vs 67.2%; HR, 2.5; P=0.005). Although these recurrence score assays have a definite prognostic value in early stage colon cancer, their ability to predict benefit from adjuvant chemotherapy has not been established yet.Citation62

Emerging biomarkers

PIK3CA mutations

PIK3CA gene encodes for the p110α catalytic subunit of the PI3K protein, which is a component of the PI3K/PTEN/AKT signaling pathway. PIK3CA mutations occur in 10%–18% of CRCs, mainly in exons 9 and 20.Citation31,Citation43,Citation63,Citation64 When mutated, PIK3CA induces phosphorylation of AKT, which promotes cell growth and suppresses apoptosis in CRCs.

The impact of PIK3CA mutations on the outcome of mCRC has been evaluated in retrospective studies. In the study by De Roock et al, it was suggested that not all PIK3CA mutations behave similarly in terms of response to anti-EGFR therapy.Citation31 PIK3CA exon 9 mutations had no effect, whereas exon 20 mutations were associated with poor clinical response to cetuximab compared to wild-type PIK3CA tumors (OS, 34 weeks vs 51 weeks [HR, 3.29; P=0.0057]). Several meta-analyses have also been performed to study the association between PIK3CA mutational status and clinical response to anti-EGFR therapy in mCRC patients.Citation39,Citation65,Citation66 In one such systematic review with meta-analysis, Yang et al evaluated the predictive value of specific exon mutations of PIK3CA (exons 9 and 20) in response to anti-EGFR therapy in KRAS wild-type mCRC patients.Citation39 PIK3CA exon 20 mutations (but not PIK3CA exon 9 mutations) were associated with shorter PFS and OS. In another meta-analysis by Wu et al, the presence of a PIK3CA mutation was associated with poorer PFS (HR, 1.53; P<0.001) and OS (HR, 1.28; P=0.015) in the unselected mCRC patients treated with anti-EGFR therapy.Citation66 The negative predictive value of PIK3CA mutation toward anti-EGFR therapy was even stronger when the analysis was restricted to patients with only KRAS wild-type disease (HR for PFS was 2.44; P=0.004).

Based on these results, the presence of PIK3CA mutations may be considered a negative predictive biomarker for anti-EGFR therapy in patients with wild-type KRAS mCRC, and this is largely driven by PIK3CA mutations in exon 20. However, as the available clinical data are largely retrospective in nature, the recent CRC biomarker guideline recommends against routine PIK3CA mutational analysis for selection of therapy outside of a clinical trial.Citation26

Loss of PTEN expression

PTEN acts as a tumor suppressor gene, where its normal function is to inhibit PI3K-initiated signaling. Accordingly, loss of PTEN expression results in activation of the PI3K/AKT pathway. PTEN loss is present in 20%–40% of the unselected CRC cases when evaluated by immunohistochemistry (IHC). The available data surrounding the role of PTEN expression as a biomarker in CRC are inconsistent and discordant.

Data from a few retrospective studies suggest that PTEN expression in CRC might have a prognostic value, such that low PTEN expression is associated with aggressive tumor behavior, liver metastases, lymphatic invasion, venous invasion, and poor survival.Citation67Citation70 On the other hand, there are several studies that refute the prognostic influence of PTEN in CRC.Citation71,Citation72

Similarly, PTEN expression has also been evaluated for its predictive value toward anti-EGFR therapy in mCRC but has shown inconsistent results. In a few studies, the loss of PTEN expression was associated with resistance to anti-EGFR therapy.Citation73Citation76 The largest of these is the retrospective study by Loupakis et al,Citation75 which evaluated the effect of PTEN expression loss on the clinical outcome of mCRC patients treated with cetuximab-based therapy. Patients with PTEN loss had significantly shorter PFS than those with PTEN-positive tumors (3.3 months vs 4.7 months; HR, 0.49; P<0.005). In contrast, several other published reports suggest that PTEN loss is not predictive of response to anti-EGFR therapy.Citation33,Citation77Citation79

Due to these conflicting results, the precise role of PTEN status as a prognostic and predictive biomarker of CRC remains unclear. In addition to the inconsistency in data, there are other potential issues that limit the use of PTEN loss as a biomarker. PTEN loss is usually measured by IHC or fluorescence in situ hybridization (deletion), and at present, there is no international standard for IHC assessment of PTEN loss. As a result, there can be significant interobserver variation among the different clinical trials.Citation80,Citation81 Moreover, there appears to be a significant discordance (66%) in PTEN expression between the primary tumor and the matched metastases.Citation82

Therefore, PTEN loss cannot be viewed as a reliable predictive marker for anti-EGFR therapy at this time. The recent CRC biomarker guideline also does not recommend routine analysis of PTEN for the purposes of therapy selection.Citation26

HER2 amplification

HER2 (also known as ERBB2) is a proto-oncogene that belongs to the human EGFR family. It is amplified iñ5% of patients with KRAS exon 2 wild-type mCRC and has a high concordance between the primary tumor and the metastases.Citation83 It is also mutually exclusive with alterations to KRAS, NRAS, and BRAF in CRC.Citation43

HER2 amplification has been studied as a biomarker to predict response toward HER2-directed therapy in mCRC.Citation84 The phase II HERACLES trial evaluated the activity of dual HER2 inhibition using trastuzumab plus lapatinib in chemo-refractory mCRC patients with HER2-positive, KRAS exon 2 (codons 12 and 13) wild-type disease.Citation84 Of the 27 patients enrolled in the study, eight patients showed a response (ORR 30%). Moreover, the responses were noted to be durable with a median duration of 38 weeks, and the treatment combination was well tolerated. These results show that HER2 amplification is probably a clinically relevant genetic alteration that has a potential role as a predictive biomarker in CRC. However, the extremely low prevalence of this genetic alteration in colorectal tumors presents a significant challenge in validating these results in a confirmatory phase III clinical trial setting.

Immunologic characteristics

Galon and his colleaguesCitation85 were the first to demonstrate the importance of immune signatures as a prognostic marker in colon cancer, beyond the conventional TNM staging. The distribution of total T-cells (CD3+), cytotoxic T-effector cells (CD8+), and memory T-cells (CD45RO+) was evaluated in the tumor core and at the invasive margin. The investigators demonstrated that tumors with elevated levels of CD3+ T-cells in the core as well as at the invasive margin were associated with the best clinical outcome, independent of the TNM stage. In a subsequent study, the prognostic role of tumor-infiltrating lymphocytes was also demonstrated in mCRC.Citation86 In this study, high tumor-infiltrating lymphocyte density in the metastatic sites conferred a greater response to chemotherapy and was associated with a longer PFS.

Recently, an “Immunoscore” methodology has been defined with an intent to quantify the immune infiltrate inside the tumor.Citation87 This scoring system is based on the density of CD8+ cytotoxic T-cells and CD45RO+ memory cells at the invasive margin and in the center of the tumor. In a worldwide consortium-based analysis of tissue samples from stage I–III colon cancer patients, this standardized immunoscore assay was recently validated as a prognostic biomarker in early stage colon cancer.Citation88,Citation89 In the training set consisting of 700 patients, the patients with “high” Immunoscore had the lowest risk of cancer recurrence at 5 years (HR for “high” vs “low” immunoscore, 0.20; P<0.0001).Citation89 These findings were confirmed independently in the internal and external validation sets, consisting of 636 and 1,345 patients, respectively. Moreover, the prognostic value of immunoscore assay was independent of the known existing prognostic factors including T stage, N stage, and MSI status.

CDX2 expression

CDX2 is an intestine-specific transcription factor and is a major regulator of intestinal development and oncogenesis.Citation90 In fact, it is a highly sensitive and specific marker of adenocarcinomas of intestinal origin. It is known that colon cancers that lack CDX2 expression are associated with advanced stage, poor differentiation, vascular invasion, BRAF mutation, and CIMP pathway.Citation91

CDX2 expression has been recently evaluated for its role as a prognostic and predictive biomarker in early stage colon cancer.Citation92 In this study’s validation data set, CDX2-negative colon cancers were associated with a lower 5-year DFS than the CDX2-positive tumors (12.1% vs 87.9%; HR, 2.42; P=0.003). These results were independent of the known prognostic factors including tumor stage and pathologic grade. Absence of CDX2 expression was also shown to be a potential predictive marker of response to adjuvant chemotherapy in colon cancer. Among the stage II CDX2-negative colon cancers, the 5-year DFS was found to be longer in patients who received adjuvant chemotherapy than those who did not (91% vs 56%; P=0.006). Although encouraging, these results are derived from retrospective patient cohorts and pooled data sets, and, therefore, require validation using prospective randomized trials before CDX2 expression can be incorporated as a biomarker in routine clinical practice.

Tumor location

It is now well established that there are distinct differences in the clinical and molecular characteristics of the right- vs left-sided colon tumors. The large genome-scale analysis of CRC samples performed by TCGA revealed that significant biologic differences exist between right-sided tumors and tumors originating from the other sites, such that the right-sided cancers were more frequently hypermethylated and hypermutated.Citation43

The location of colorectal primary tumor has prognostic, and potentially predictive value in CRC.Citation93Citation95 In the study by Loupakis et al, the left-sided tumors were found to have better prognosis than the right-sided tumors in three independent cohorts of mCRC patients derived from the PROVETTA, AVF2107g, and NO16966 trials.Citation93 The favorable outcome of left-sided tumors was independent of the known prognostic variables. In another study, Venook et alCitation95 studied the effect of primary tumor location on survival in mCRC patients from the CALGB/SWOG 80405 clinical trial. In the KRAS wild-type mCRC cohort, patients with left-sided primary tumor had superior survival than those with right-sided primary tumor. Post hoc subgroup analyses showed that among patients with right-sided tumors, treatment with bevacizumab was associated with longer survival than that seen with cetuximab. Conversely, among patients with left-sided tumors, treatment with cetuximab was associated with longer OS than with bevacizumab. These results suggest that in addition to the known prognostic role, the location of colorectal primary tumor might also have a predictive value toward anti-EGFR and anti-VEGF therapies.

Consensus molecular subtypes of CRC

The recently defined four transcriptome-based molecular subtypes of CRC also demonstrate clinical and prognostic associations.Citation9 For example, the CMS4 tumors are associated with worse OS and DFS even after adjustment for clinicopathologic features, MSI, BRAF, and KRAS mutation status. The CMS1 lesions are frequently right sided with high histopathologic grade, and these patients have very poor survival after disease relapse. Conversely, CMS2 tumors are predominantly left sided and have superior survival after relapse. Due to the biologic differences between the CMS subtypes, response to therapies is also expected to differ for each subtype. Future studies that prospectively compare the efficacy of therapies in different colorectal CMS subtypes will help establish its role as a predictive biomarker to guide therapeutic decisions.

MicroRNAs (miRNAs)

miRNAs are short (18–22 nucleotides), noncoding single-stranded RNA sequences involved in the posttranscriptional regulation of gene expression.Citation96 They cause either degradation or inhibition of translation by binding to the 3′-untranslated region of targeted mRNA. miRNAs are believed to play a significant role in colorectal tumorigenesis, and several candidate miRNAs are being evaluated for having either predictive or prognostic role in the treatment of CRC.Citation97Citation102 More recently, circulating exosomal miR-27a and miR-130a were demonstrated as novel diagnostic and prognostic biomarkers of CRC.Citation103

Novel molecular approaches in CRC management

Next-generation sequencing

Next-generation sequencing is non-Sanger-based, high-throughput DNA sequencing technology, where an extremely large number of DNA strands can be sequenced simultaneously in parallel, yielding substantially more throughput of several hundred billion bases in a single run than by conventional sequencing methods. Next-generation sequencing of entire CRC genome has shown that each tumor harbors around 75 mutations.Citation43 Whole-genome sequencing and whole-exome sequencing are two of the more recent advances in next-generation sequencing technology.Citation104,Citation105 Next-generation sequencing has utility in target identification for matched therapies. Specific examples include MSI-Hi for immune checkpoint inhibitor therapy, HER2 amplification for HER2-targeted therapy.

“Omics”-based approaches

Large-scale “omics” techniques have the ability to identify tumor biology with an extraordinarily high definition that enables understanding of complex pathologies and discriminate tumors with distinct molecular phenotypes. This growing knowledge has the potential to identify biomarkers leading to the development of pathway-directed targeted therapies and help individualize anticancer treatments to improve clinical outcomes.

Transcriptomics is the study of global mRNA expression of a particular tissue.Citation106 Recent transcriptomic and immune-based characterization of CRC has further improved our understanding of the colorectal tumor biology, and the precision medicine paradigm in this disease has now evolved and shifted from clonal perspective for targeted therapies, to a clonal–stromal–immune perspective, which involves multi-molecular, multidrug approach.Citation107 Epigenomics is the study of epigenetic modifications that are now widely known to play an important role in tumorigenesis and are prevalent in CRC.Citation108 These alterations include DNA promoter region methylation and histone modifications, and several biomarkers of these epigenetic changes have been described.Citation108 Proteomics refers to large-scale study of proteins to comprehensively map biologic processes such as molecular mechanisms of carcinogenesis.Citation109,Citation110 It involves identification and quantification of proteins, as well as the study of their location, modifications, interactions, and functions. Proteomics has identified five different CRC subtypes in TCGA cohort, two of which overlap with the MSI/CIMP transcriptomic subtype.Citation43,Citation109,Citation110 Omics strategy is now being explored to guide therapies in these distinct CRC subtypes. Finally, genomics, based on the advances in the next-generation sequencing technology, is providing a great opportunity for biomarker development and realization of precision medicine in CRC.

Circulating tumor DNA

Circulating cell-free DNA is referred to the DNA fragments found in cell-free components of peripheral blood, and it is largely derived from the normal tissue. A small fraction of circulating cell-free DNA in patients with malignancy is also derived from the tumor cells and is referred to as circulating tumor DNA (ctDNA). CtDNA is present in a majority of patients with mCRC, and molecular analysis of ctDNA from a patient’s peripheral blood is a considerably less invasive means of identifying specific genetic mutations in tumors.

CtDNA has high accuracy for the detection of KRAS and other mutations in CRC patients.Citation111,Citation112 Moreover, it does not have the limitation of spatial selection bias owing to tumor heterogeneity that is frequently associated with biopsy of the tumor tissue sample. CtDNA has also made it significantly easier to study the clonal evolution of CRC and detection of emerging KRAS-mutated clones during anti-EGFR therapy. For example, in the studies by Diaz et alCitation113 and Misale et al,Citation114 the emergence of KRAS mutations was detectable in ctDNA about 6–10 months prior to radiographic evidence of disease progression while on anti-EGFR therapy.

CtDNA has an immense potential to be a noninvasive molecular biomarker. Given its ease for sample collection, it can be used to estimate tumor burden, predict radiologic tumor progression, detect minimal residual disease, monitor both tumor progression and treatment response, and study acquired resistance mechanisms.Citation104,Citation115,Citation116 However, further development of ctDNA-based biomarkers would require standardization of the assay platform and prospective studies to validate ctDNA-based biomarkers in patient care.Citation115,Citation117

Circulating tumor cells

Tumor cells detected in the peripheral blood are referred to as circulating tumor cells (CTCs). CtDNA and CTCs are considered distinct entities, as ctDNA is often present in patients without detectable CTCs.Citation111 Several studies have suggested a potential prognostic role of CTCs in CRC.Citation118Citation120 In a prospective multicenter study, Cohen et alCitation118 examined CTCs from 430 patients with mCRC at baseline and after starting first-, second-, or third-line chemotherapy. Patients were stratified into unfavorable and favorable prognostic groups based on CTC levels of ≥3 or <3 CTCs/7.5 mL, respectively. Patients in the baseline unfavorable CTCs group had a shorter median PFS (4.5 months vs 7.9 months; P=0.0002) and OS (9.4 months vs 18.5 months; P<0.0001) compared to those in the favorable CTCs group. Patients whose CTC level converted from the unfavorable to the favorable group after the initial 3–5 weeks of treatment had a significantly improved PFS (6.2 months vs 1.6 months; P=0.02) and OS (11.0 months vs 3.7 months; P=0.0002).

Precision medicine – goals and challenges

The evolving paradigm

Precision medicine paradigm in CRC originally started with “one gene, one drug” approach, which was a considerable advancement over nonselective patient treatment based on the results of conventional population-level clinical trials. However, it was soon realized that such strategy is unlikely to result in durable remission given the complex molecular makeup of CRC with significant cross-talk between signaling pathways. As an example, there is virtually no clinical activity of BRAF inhibitors in BRAF-mutant mCRC, which is in strike contrast to their activity in metastatic melanoma.Citation121 This lack of response to BRAF inhibitors in CRC is due to the rapid activation of EGFR/PI3K pathway via feedback loop, resulting in the escape from downstream inhibition of targets because of the cross-talk between signaling pathways in colon cancer cells.Citation122,Citation123 Consequently, a “multi-gene, multi-drug” approach was evaluated and was the next big step to optimize outcomes. The encouraging activity observed with combined inhibition of BRAF, MEK, and EGFR in early trials of BRAF-mutant CRC is a perfect example of this concept.Citation124

Impact of tumor heterogeneity

CRC is a very heterogeneous disease, and tumor heterogeneity exists at several different levels. First, the genetic makeup of a given tumor type can vary significantly between one patient and another. Next, within an individual patient, there exists a high degree of genetic heterogeneity between the primary tumor and the metastasis as well as between the different metastatic sites (intertumoral heterogeneity).Citation125Citation127 Heterogeneity can also exist in different areas within a single tumor (intratumoral heterogeneity), in different anatomical regions of the same patient (spatial tumor heterogeneity), and at different time points within the same tumor (temporal tumor heterogeneity).

Intratumoral heterogeneity relates to genetic heterogeneity, functional heterogeneity, and nongenetic (epigenetic) heterogeneity.Citation128 Genetic heterogeneity can be attributed to varying number of cancer clones depending upon the mutation rates and differences in the ability of these clones to metastasize or/and their responsiveness to therapies. Genetic heterogeneity also arises due to clonal selection from treatment pressures. For example, mutations in the driver oncogenes KRAS, NRAS, and BRAF are found to emerge in patients who were initially diagnosed with wild-type disease and were treated with anti-EGFR antibody, but subsequently developed resistance to such therapy.Citation129 Even though nondominant, these dynamic clonal and subclonal alterations are responsible for sustaining CRC progression.

These different levels of tumor heterogeneity can affect the signaling of multiple key oncogenic pathways resulting in extensive phenotypic variation, such that each tumor clone even within the same tumor displays its own genetic, epigenetic, transcriptomic, and proteomic profile.Citation126,Citation130,Citation131 Consequently, intratumoral heterogeneity poses an enormous challenge to the practice of personalized medicine because any given therapeutic agent targeted at a specific molecular alteration is likely to be successful in only a minority of patients. The four CMS groups are currently the best representation of CRC heterogeneity at the gene-expression level.Citation107

Future directions and conclusion

Significant advances have been made in the development of molecular biomarkers for the treatment of mCRC. Unlike BRAF, mutations in RAS genes (KRAS and NRAS) do not have a definite prognostic significance in mCRC. However, the presence of activating mutations in these RAS genes is predictive for lack of response to anti-EGFR therapy. There is now a well-established body of evidence documenting that patients with RAS-mutated mCRC should not receive anti-EGFR therapy. However, it is worthwhile to note that the mere presence of wild-type RAS in the tumor does not guarantee a response to anti-EGFR therapy. This suggests that further refinement in the biomarker selection criteria beyond our current understanding is warranted.

CRC is a highly heterogeneous disease, and, therefore, molecular characterization of these tumors is necessary to determine molecularly homogeneous subtypes, understand tumor biology, and identify the most relevant driver events. Recent advances in DNA sequencing technology and “omics”-based approaches have provided promising new strategies for the development of molecular biomarkers in this disease. We have now identified and characterized genomic, transcriptomic, and immune-based subtypes of CRC. Our future success will depend on the ability to guide therapeutic decisions based on distinct biologic subtypes identified using such gene-expression signatures. Given the significant heterogeneity associated with colorectal tumors, the clonal perspective for targeted therapies is met with only limited success. Instead, a “multimolecular, multidrug” approach is now advocated that integrates knowledge from transcriptomic and immune subtyping of CRC to guide novel therapeutic strategies and precision medicine.Citation107 For example, combination of PD-L1 blockade plus immune stimulation with MEK inhibitor in MSS colon cancer has shown early signs of clinical efficacy.Citation132

To conclude, the goal of precision medicine is to personalize the therapeutic strategy in each given individual depending upon the clinicopathologic features, genomic profile, epigenetic changes, and other molecular characteristics of the tumor. Future clinical trials should, therefore, focus on testing therapies in molecularly defined subgroups of CRC to optimize efficacy, minimize toxicity, and reduce cost. Of course, such initiatives will require intimate collaborations between institutions in multiple countries across continents to ensure rapid clinical trial accrual and efficacy evaluation.

Disclosure

The author reports no conflicts of interest in this work.

References

  • SiegelRLMillerKDJemalACancer statisticsCA: Cancer J Clin201868173029313949
  • LenzHJNiedzwieckiDInnocentiFCALGB/SWOG 80405: phase III trial of irinotecan/5-FU/leucovorin (FOLFIRI) or oxaliplatin/5-FU/leucovorin (MFOLFOX6) with bevacizumab (BV) or cetuximab (CET) for patients with expanded RAS analyses untreated metastatic adenocarcinoma of the colon or rectum (mCRC)Paper presented at: ESMO CongressMadrid, Spain2014
  • HeinemannVvon WeikersthalLFDeckerTFOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer (FIRE-3): a randomised, open-label, phase 3 trialLancet Oncol201415101065107525088940
  • SchwartzbergLSRiveraFKarthausMPEAK: 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 cancerJ Clin Oncol201432212240224724687833
  • GoelGChauhanAHoseinPJRamucirumab: a novel anti-angiogenic agent in the treatment of metastatic colorectal cancerCurr Colorectal Cancer Rep2016125232240
  • GoelGEvolution of regorafenib from bench to bedside in colorectal cancer: is it an attractive option or merely a “me too” drug?Cancer Manag Res20181042543729563833
  • FearonERVogelsteinBA genetic model for colorectal tumorigenesisCell19906157597672188735
  • ShihIMZhouWGoodmanSNLengauerCKinzlerKWVogelsteinBEvidence that genetic instability occurs at an early stage of colorectal tumorigenesisCancer Res200161381882211221861
  • GuinneyJDienstmannRWangXThe consensus molecular subtypes of colorectal cancerNat Med201521111350135626457759
  • KarnoubAEWeinbergRARas oncogenes: split personalitiesNat Rev Mol Cell Biol20089751753118568040
  • WongRCunninghamDUsing predictive biomarkers to select patients with advanced colorectal cancer for treatment with epidermal growth factor receptor antibodiesJ Clin Oncol200826355668567019001346
  • LièvreABachetJBLe CorreDKRAS mutation status is predictive of response to cetuximab therapy in colorectal cancerCancer Res20066683992399516618717
  • KarapetisCSKhambata-FordSJonkerDJK-ras mutations and benefit from cetuximab in advanced colorectal cancerN Engl J Med2008359171757176518946061
  • AmadoRGWolfMPeetersMWild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancerJ Clin Oncol200826101626163418316791
  • van CutsemEKöhneC-HHitreECetuximab and chemotherapy as initial treatment for metastatic colorectal cancerN Engl J Med Overseas Ed20093601414081417
  • BokemeyerCBondarenkoIMakhsonAFluorouracil, leucovorin, and oxaliplatin with and without cetuximab in the first-line treatment of metastatic colorectal cancerJ Clin Oncol200927566367119114683
  • DouillardJYSienaSCassidyJRandomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME studyJ Clin Oncol201028314697470520921465
  • PeetersMPriceTJCervantesARandomized phase III study of panitumumab with fluorouracil, leucovorin, and irinotecan (FOLFIRI) compared with FOLFIRI alone as second-line treatment in patients with metastatic colorectal cancerJ Clin Oncol201028314706471320921462
  • AdelsteinB-ADobbinsTAHarrisCAMarschnerICWardRLA systematic review and meta-analysis of KRAS status as the determinant of response to anti-EGFR antibodies and the impact of partner chemotherapy in metastatic colorectal cancerEur J Cancer20114791343135421550229
  • DahabrehIJTerasawaTCastaldiPJTrikalinosTASystematic review: anti-epidermal growth factor receptor treatment effect modification by KRAS mutations in advanced colorectal cancerAnn Intern Med20111541374921200037
  • LoupakisFCremoliniCSalvatoreLClinical impact of anti-epidermal growth factor receptor monoclonal antibodies in first-line treatment of metastatic colorectal cancer: meta-analytical estimation and implications for therapeutic strategiesCancer201211861523153222009364
  • ValeCLTierneyJFFisherDDoes anti-EGFR therapy improve outcome in advanced colorectal cancer? A systematic review and meta-analysisCancer Treat Rev201238661862522118887
  • DouillardJ-YOlinerKSSienaSPanitumumab–FOLFOX4 treatment and RAS mutations in colorectal cancerN Engl J Med Overseas Ed20133691110231034
  • SorichMJWieseMDRowlandAKichenadasseGMckinnonRAKarapetisCSExtended RAS mutations and anti-EGFR monoclonal antibody survival benefit in metastatic colorectal cancer: a meta-analysis of randomized, controlled trialsAnn Oncol2015261132125115304
  • van CutsemELenzHJKöhneCHFluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancerJ Clin Oncol201533769270025605843
  • SepulvedaARHamiltonSRAllegraCJMolecular biomarkers for the evaluation of colorectal cancer: guideline from the American Society for Clinical Pathology, College of American Pathologists, Association for Molecular Pathology, and the American Society of Clinical OncologyJ Clin Oncol201735131453148628165299
  • AndreyevHJNormanARCunninghamDKirsten ras mutations in patients with colorectal cancer: the ‘RASCAL II’ studyBr J Cancer200185569269611531254
  • RichmanSDSeymourMTChambersPKRAS and BRAF mutations in advanced colorectal cancer are associated with poor prognosis but do not preclude benefit from oxaliplatin or irinotecan: results from the MRC FOCUS trialJ Clin Oncol200927355931593719884549
  • KimSTParkKHKimJSShinSWKimYHImpact of KRAS mutation status on outcomes in metastatic colon cancer patients without anti-epidermal growth factor receptor therapyCancer Res Treat2013451556223613671
  • RenJLiGGeJLiXZhaoYIs K-ras gene mutation a prognostic factor for colorectal cancer: a systematic review and meta-analysisDis Colon Rectum201255891392322810479
  • de RoockWClaesBBernasconiDEffects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysisLancet Oncol201011875376220619739
  • di NicolantonioFMartiniMMolinariFWild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancerJ Clin Oncol200826355705571219001320
  • TolJDijkstraJRKlompMMarkers for EGFR pathway activation as predictor of outcome in metastatic colorectal cancer patients treated with or without cetuximabEur J Cancer201046111997200920413299
  • van CutsemEKöhneCHLángICetuximab 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 statusJ Clin Oncol201129152011201921502544
  • BokemeyerCvan CutsemERougierPAddition of cetuximab to chemotherapy as first-line treatment for KRAS wild-type metastatic colorectal cancer: pooled analysis of the CRYSTAL and OPUS randomised clinical trialsEur J Cancer201248101466147522446022
  • LinJSWebberEMSengerCAHolmesRSWhitlockEPSystematic review of pharmacogenetic testing for predicting clinical benefit to anti-EGFR therapy in metastatic colorectal cancerAm J Cancer Res20111565066221779535
  • XuQXuATZhuMMTongJLXuXTRanZHPredictive and prognostic roles of BRAF mutation in patients with metastatic colorectal cancer treated with anti-epidermal growth factor receptor monoclonal antibodies: a meta-analysisJ Dig Dis201314840941623615046
  • YuanZXWangXYQinQYThe prognostic role of BRAF mutation in metastatic colorectal cancer receiving anti-EGFR monoclonal antibodies: a meta-analysisPLoS One201386e6599523776587
  • YangZYWuXYHuangYFPromising biomarkers for predicting the outcomes of patients with KRAS wild-type metastatic colorectal cancer treated with anti-epidermal growth factor receptor monoclonal antibodies: a systematic review with meta-analysisInt J Cancer201313381914192523494461
  • RowlandADiasMMWieseMDMeta-analysis of BRAF mutation as a predictive biomarker of benefit from anti-EGFR monoclonal antibody therapy for RAS wild-type metastatic colorectal cancerBr J Cancer2015112121888189425989278
  • PietrantonioFPetrelliFCoinuAPredictive role of BRAF mutations in patients with advanced colorectal cancer receiving cetuximab and panitumumab: a meta-analysisEur J Cancer201551558759425673558
  • SeymourMTBrownSRMiddletonGPanitumumab and irinotecan versus irinotecan alone for patients with KRAS wild-type, fluorouracil-resistant advanced colorectal cancer (PICCOLO): a prospectively stratified randomised trialLancet Oncol201314874975923725851
  • The Cancer Genome Atlas NetworkComprehensive molecular characterization of human colon and rectal cancerNature2012487740733033722810696
  • AaltonenLASalovaaraRKristoPIncidence of hereditary nonpolyposis colorectal cancer and the feasibility of molecular screening for the diseaseN Engl J Med Overseas Ed19983382114811487
  • Evaluation of Genomic Applications in Prevention Working GroupRecommendations from the EGAPP Working Group: genetic testing strategies in newly diagnosed individuals with colorectal cancer aimed at reducing morbidity and mortality from Lynch syndrome in relativesGenet Med2009111354119125126
  • HampelHFrankelWLMartinEFeasibility of screening for Lynch syndrome among patients with colorectal cancerJ Clin Oncol200826355783578818809606
  • SinicropeFAFosterNRThibodeauSNDNA mismatch repair status and colon cancer recurrence and survival in clinical trials of 5-fluorouracil-based adjuvant therapyJ Natl Cancer Inst20111031186387521597022
  • LanzaGGafàRMaestriISantiniAMatteuzziMCavazziniLImmunohistochemical pattern of MLH1/MSH2 expression is related to clinical and pathological features in colorectal adenocarcinomas with microsatellite instabilityMod Pathol200215774174912118112
  • PopatSHubnerRHoulstonRSSystematic review of microsatellite instability and colorectal cancer prognosisJ Clin Oncol200523360961815659508
  • MyintZWGoelGRole of modern immunotherapy in gastrointestinal malignancies: a review of current clinical progressJ Hematol Oncol20171018628434400
  • SargentDJMarsoniSMongesGDefective mismatch repair as a predictive marker for lack of efficacy of fluorouracil-based adjuvant therapy in colon cancerJ Clin Oncol201028203219322620498393
  • LipsonEJSharfmanWHDrakeCGDurable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibodyClin Cancer Res201319246246823169436
  • LlosaNJCruiseMTamAThe vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpointsCancer Discov201551435125358689
  • DtLUramJNWangHPD-1 blockade in tumors with mismatch-repair deficiencyN Engl J Med2015372262509252026028255
  • DtLUramJNWangHProgrammed death-1 blockade in mismatch repair deficient colorectal cancerJ Clin Oncol20163410326628472
  • OvermanMJMcdermottRLeachJLNivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 studyLancet Oncol20171891182119128734759
  • OvermanMJLonardiSWongKYMDurable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair–deficient/microsatellite instability–high metastatic colorectal cancerJ Clin Oncol201836877377929355075
  • GongJWangCLeePPChuPFakihMResponse to PD-1 blockade in microsatellite stable metastatic colorectal cancer harboring a POLE mutationJ Natl Compr Cancer Netw2017152142147
  • GrayRGQuirkePHandleyKValidation study of a quantitative multigene reverse transcriptase-polymerase chain reaction assay for assessment of recurrence risk in patients with stage II colon cancerJ Clin Oncol201129354611461922067390
  • VenookAPNiedzwieckiDLopatinMBiologic determinants of tumor recurrence in stage II colon cancer: validation study of the 12-gene recurrence score in cancer and leukemia group B (CALGB) 9581J Clin Oncol201331141775178123530100
  • SalazarRRoepmanPCapellaGGene expression signature to improve prognosis prediction of stage II and III colorectal cancerJ Clin Oncol2011291172421098318
  • GoelGEvolving role of gene expression signatures as bio-markers in early-stage colon cancerJ Gastrointest Cancer201445439940424989938
  • VelhoSOliveiraCFerreiraAThe prevalence of PIK3CA mutations in gastric and colon cancerEur J Cancer200541111649165415994075
  • BaraultLVeyrieNJoosteVMutations in the RAS-MAPK, PI(3)K (phosphatidylinositol-3-OH kinase) signaling network correlate with poor survival in a population-based series of colon cancersInt J Cancer2008122102255225918224685
  • MaoCYangZYHuXFChenQTangJLPIK3CA exon 20 mutations as a potential biomarker for resistance to anti-EGFR monoclonal antibodies in KRAS wild-type metastatic colorectal cancer: a systematic review and meta-analysisAnn Oncol20122361518152522039088
  • WuSGanYWangXLiuJLiMTangYPIK3CA mutation is associated with poor survival among patients with metastatic colorectal cancer following anti-EGFR monoclonal antibody therapy: a meta-analysisJ Cancer Res Clin Oncol2013139589190023435830
  • ColakogluTYildirimSKayaselcukFClinicopathological significance of PTEN loss and the phosphoinositide 3-kinase/Akt pathway in sporadic colorectal neoplasms: is PTEN loss predictor of local recurrence?Am J Surg2008195671972518440486
  • SawaiHYasudaAOchiNLoss of PTEN expression is associated with colorectal cancer liver metastasis and poor patient survivalBMC Gastroenterol2008815619036165
  • XhLZhengHCTakahashiHMasudaSYangXHTakanoYPTEN expression and mutation in colorectal carcinomasOncol Rep200922475776419724853
  • LinMSHuangJXChenWCExpression of PPARγ and PTEN in human colorectal cancer: an immunohistochemical study using tissue microarray methodologyOncol Lett2011261219122422848291
  • PriceTJHardinghamJELeeCKPrognostic impact and the relevance of PTEN copy number alterations in patients with advanced colorectal cancer (CRC) receiving bevacizumabCancer Med20132327728523930204
  • EklöfVWikbergMLEdinSThe prognostic role of KRAS, BRAF, PIK3CA and PTEN in colorectal cancerBr J Cancer2013108102153216323660947
  • FrattiniMSalettiPRomagnaniEPTEN loss of expression predicts cetuximab efficacy in metastatic colorectal cancer patientsBr J Cancer20079781139114517940504
  • PerroneFLampisAOrsenigoMPI3KCA/PTEN deregulation contributes to impaired responses to cetuximab in metastatic colorectal cancer patientsAnn Oncol2009201849018669866
  • LoupakisFPollinaLStasiIPTEN expression and KRAS mutations on primary tumors and metastases in the prediction of benefit from cetuximab plus irinotecan for patients with metastatic colorectal cancerJ Clin Oncol200927162622262919398573
  • NegriFVBozzettiCLagrastaCAPTEN status in advanced colorectal cancer treated with cetuximabBr J Cancer2010102116216419953097
  • Laurent-PuigPCayreAManceauGAnalysis of PTEN, BRAF, and EGFR status in determining benefit from cetuximab therapy in wild-type KRAS metastatic colon cancerJ Clin Oncol200927355924593019884556
  • UliviPCapelliLValgiustiMPredictive role of multiple gene alterations in response to cetuximab in metastatic colorectal cancer: a single center studyJ Transl Med20121018722569004
  • KarapetisCSJonkerDDaneshmandMPIK3CA, BRAF, and PTEN status and benefit from cetuximab in the treatment of advanced colorectal cancer—results from NCIC CTG/AGITG CO.17Clin Cancer Res201420374475324218517
  • MolinariFFrattiniMFunctions and regulation of the PTEN gene in colorectal cancerFront Oncol2013332624475377
  • SridharanMHubbardJMGrotheyAColorectal cancer: how emerging molecular understanding affects treatment decisionsOncology201428211011824701697
  • CejasPLopez-GomezMAguayoCAnalysis of the concordance in the EGFR pathway status between primary tumors and related metastases of colorectal cancer patients: implications for cancer therapyCurr Cancer Drug Targets201212212413122229245
  • ValtortaEMartinoCSartore-BianchiAAssessment of a HER2 scoring system for colorectal cancer: results from a validation studyMod Pathol201528111481149126449765
  • Sartore-BianchiATrusolinoLMartinoCDual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trialLancet Oncol201617673874627108243
  • GalonJCostesASanchez-CaboFType, density, and location of immune cells within human colorectal tumors predict clinical outcomeScience200631357951960196417008531
  • HalamaNMichelSKloorMLocalization and density of immune cells in the invasive margin of human colorectal cancer liver metastases are prognostic for response to chemotherapyCancer Res201171175670567721846824
  • GalonJPagèsFMarincolaFMCancer classification using the Immunoscore: a worldwide task forceJ Transl Med201210120523034130
  • GalonJMlecnikBMarliotFValidation of the Immunoscore (IM) as a prognostic marker in stage I/II/III colon cancer: results of a worldwide consortium-based analysis of 1,336 patientsJ Clin Oncol201634Suppl abstr 3500
  • PagèsFMlecnikBMarliotFInternational validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy studyLancet2018391101352128213929754777
  • WerlingRWYazijiHBacchiCEGownAMCDX2, a highly sensitive and specific marker of adenocarcinomas of intestinal origin: an immunohistochemical survey of 476 primary and metastatic carcinomasAm J Surg Pathol200327330331012604886
  • BabaYNoshoKShimaKRelationship of CDX2 loss with molecular features and prognosis in colorectal cancerClin Cancer Res200915144665467319584150
  • DalerbaPSahooDPaikSCDX2 as a prognostic biomarker in stage II and stage III colon cancerN Engl J Med Overseas Ed20163743211222
  • LoupakisFYangDYauLPrimary tumor location as a prognostic factor in metastatic colorectal cancerJ Natl Cancer Inst20151073dju42725713148
  • TejparSStintzingSCiardielloFPrognostic and predictive relevance of primary tumor location in patients with RAS wild-type metastatic colorectal cancer: retrospective analyses of the CRYSTAL and FIRE-3 trialsJAMA Oncol201632194201
  • VenookAPNiedzwieckiDInnocentiFImpact of primary (1°) tumor location on overall survival (OS) and progression-free survival (PFS) in patients (pts) with metastatic colorectal cancer (mCRC): analysis of CALGB/SWOG 80405 (Alliance)J Clin Oncol201634suppl abstr 3504
  • BartelDPMicroRNAs: target recognition and regulatory functionsCell2009136221523319167326
  • SchetterAJOkayamaHHarrisCCThe role of microRNAs in colorectal cancerCancer J201218324425222647361
  • LouXQiXZhangYLongHYangJDecreased expression of microRNA-625 is associated with tumor metastasis and poor prognosis in patients with colorectal cancerJ Surg Oncol2013108423023523861214
  • YangI-PTsaiH-LHuangC-WThe functional significance of microRNA-29c in patients with colorectal cancer: a potential circulating biomarker for predicting early relapsePLoS One201386e6684223840538
  • ZhangYUWangXXuBEpigenetic silencing of miR-126 contributes to tumor invasion and angiogenesis in colorectal cancerOncol Rep20133041976198423900443
  • DongYZhaoJWuCWCwWTumor suppressor functions of miR-133a in colorectal cancerMol Cancer Res20131191051106023723074
  • KaraayvazMZhaiHJuJmiR-129 promotes apoptosis and enhances chemosensitivity to 5-fluorouracil in colorectal cancerCell Death Dis20134e65923744359
  • LiuXPanBSunLCirculating exosomal miR-27a and miR-130a act as novel diagnostic and prognostic biomarkers of colorectal cancerCancer Epidemiol Biomarkers Prev201827774675429739748
  • WangLWheelerDAGenomic sequencing for cancer diagnosis and therapyAnnu Rev Med201465334824274147
  • WangXKuangYYHuXTXtHAdvances in epigenetic biomarker research in colorectal cancerWorld J Gastroenterol201420154276428724764665
  • DeySSKesterLSpanjaardBBienkoMvan OudenaardenAIntegrated genome and transcriptome sequencing of the same cellNat Biotechnol201533328528925599178
  • DienstmannRVermeulenLGuinneyJKopetzSTejparSTaberneroJConsensus molecular subtypes and the evolution of precision medicine in colorectal cancerNat Rev Cancer2017172799228050011
  • van EngelandMDerksSSmitsKMMeijerGAHermanJGColorectal cancer epigenetics: complex simplicityJ Clin Oncol201129101382139121220596
  • ZhangBWangJWangXProteogenomic characterization of human colon and rectal cancerNature2014513751838238725043054
  • UhlenMFagerbergLHallstromBMTissue-based map of the human proteomeScience20153476220126041925613900
  • BettegowdaCSausenMLearyRJDetection of circulating tumor DNA in early- and late-stage human malignanciesSci Transl Med20146224224ra224
  • ThierryARMouliereFEl MessaoudiSClinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNANat Med201420443043524658074
  • DiazLAWilliamsRTWuJThe molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancersNature2012486740453754022722843
  • MisaleSYaegerRHoborSEmergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancerNature2012486740453253622722830
  • NewmanAMBratmanSVToJAn ultrasensitive method for quantitating circulating tumor DNA with broad patient coverageNat Med201420554855424705333
  • DiazLABardelliALiquid biopsies: genotyping circulating tumor DNAJ Clin Oncol201432657958624449238
  • SausenMParpartSDiazLACirculating tumor DNA moves further into the spotlightGenome Med2014653524944584
  • CohenSJPuntCJIannottiNRelationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancerJ Clin Oncol200826193213322118591556
  • RomitiARaffaSdi RoccoRCirculating tumor cells count predicts survival in colorectal cancer patientsJ Gastrointestin Liver Dis201423327928425267956
  • HuangXGaoPSongYRelationship between circulating tumor cells and tumor response in colorectal cancer patients treated with chemotherapy: a meta-analysisBMC Cancer20141497625519477
  • KopetzSDesaiJChanEPLX4032 in metastatic colorectal cancer patients with mutant BRAF tumorsJ Clin Oncol20102815_Suppl3534
  • PrahalladASunCHuangSUnresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFRNature2012483738710010322281684
  • CorcoranRBEbiHTurkeABEGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenibCancer Discov20122322723522448344
  • Triple therapy improves colorectal cancer responseCancer Discov2016612OF6
  • GerlingerMRowanAJHorswellSIntratumor heterogeneity and branched evolution revealed by multiregion sequencingN Engl J Med20123661088389222397650
  • BedardPLHansenARRatainMJSiuLLTumour heterogeneity in the clinicNature2013501746735536424048068
  • VogelsteinBPapadopoulosNVelculescuVEZhouSDiazLAKinzlerKWCancer genome landscapesScience201333961271546155823539594
  • PuntCJKoopmanMVermeulenLFrom tumour heterogeneity to advances in precision treatment of colorectal cancerNat Rev Clin Oncol201714423524627922044
  • ArenaSBellosilloBSiravegnaGEmergence of multiple EGFR extracellular mutations during cetuximab treatment in colorectal cancerClin Cancer Res20152192157216625623215
  • BurrellRAMcgranahanNBartekJSwantonCThe causes and consequences of genetic heterogeneity in cancer evolutionNature2013501746733834524048066
  • MeachamCEMorrisonSJTumour heterogeneity and cancer cell plasticityNature2013501746732833724048065
  • BendellJCKimTWGohBCClinical activity and safety of cobimetinib (cobi) and atezolizumab in colorectal cancer (CRC)J Clin Oncol201634Supplabstr 3502