Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jan 1.
Published in final edited form as: Nat Rev Clin Oncol. 2019 Jul 9;17(1):11–32. doi: 10.1038/s41571-019-0241-1

Biomarker-guided therapy for colorectal cancer: strength in complexity

Anita Sveen 1,2,*, Scott Kopetz 3, Ragnhild A Lothe 1,2
PMCID: PMC7577509  NIHMSID: NIHMS1599244  PMID: 31289352

Abstract

The number of molecularly stratified treatment options available to patients with colorectal cancer (CRC) is increasing, with a parallel increase in the use of biomarkers to guide prognostication and treatment decision making. This increase in both the number of biomarkers and their use has resulted in an increasingly complex situation, which is evident both from the extensive interactions between biomarkers and from their sometimes complex associations with patient prognosis and treatment benefit. Current and emerging biomarkers also reflect the genomic complexity of CRC, and include a wide range of aberrations such as point mutations, amplifications, fusions and hypermutator phenotypes, in addition to global gene expression subtypes. In this Review, we provide an overview of current and emerging clinically relevant biomarkers and their role in the management of patients with CRC, illustrating the intricacies of biomarker interactions and the growing treatment opportunities created by the availability of comprehensive molecular profiling.

Introduction

Clinical outcomes and treatment responses of patients with colorectal cancer (CRC) vary greatly1, and the use of stratified treatment options remains limited in standard practice2,3. In the primary setting, patients undergo surgery with a curative intent, as well as perioperative chemoradiation and/or adjuvant chemotherapy predominantly determined by cancer stage and tumour location4. However, considerable potential exists to improve the risk:benefit ratio of adjuvant chemotherapy regimens by adopting more personalized approaches. This potential is illustrated by the noninferiority of 3 months of adjuvant chemotherapy versus the standard-of-care treatment duration of 6 months in patients with low-risk stage III colon cancer5. In those with metastatic disease, the treatment repertoire has been extended to include biologically targeted agents including monoclonal antibodies targeting EGFR, such as cetuximab or panitumumab, and anti-angiogenic agents targeting VEGF signalling, such as bevacizumab or ramucirumab,6 as well as the broad-spectrum kinase inhibitor regorafenib7. As a result of improved treatment options, the overall survival (OS) of patients with metastatic CRC has increased from approximately 1 year in the era of 5-fluorouracil (5-FU) therapy alone, to approximately 3 years with currently available therapies8. No clinically validated predictive biomarkers of response to the anti-angiogenic agents are currently available, although VEGF-D expression9 and KDR mutations10 are potential candidates. By contrast, activating mutations in KRAS/NRAS are contraindications for the use of EGFR targeted therapies, and this approach remains the gold standard for the stratified treatment of patients with CRC11. Addition of cetuximab to 5-FU, leucovorin and irinotecan (FOLFIRI) has been shown to increase the median progression-free survival (PFS) of patients with previously untreated RAS wild type metastatic disease by ~3 months12. However, most patients either have a poor initial response to treatment, or develop secondary resistance to all standard therapies13. Mechanisms of resistance to targeted agents commonly include secondary, reactivating mutations in the affected signalling pathways, as demonstrated by the emergence of resistant subclones harbouring mutations in the MAPK signalling pathway during EGFR inhibition14,15.

Several advances from the past few years indicate the potential to improve the effectiveness of treatments through patient stratification based on tumour biology. Arguably, the most prominent examples are provided by the successes with anti-programmed cell death protein-1 (PD-1) and anti-cytotoxic T lymphocyte associated protein 4 (CTLA4) antibodies in metastatic cancers with a microsatellite instability (MSI) or hypermutator phenotype16-19, and the use of targeted combination therapies in patients with CRCs harbouring BRAFV600 mutations20,21. Furthermore, moving from the single-marker and single-agent approach to a more integrated perspective3, the gene expression-based consensus molecular subtypes (CMS) have provided a new and biologically rational stratification framework22. This framework defines four groups of CRCs based on intrinsic biological characteristics: CMS1-MSI/immune; CMS2-epithelial/canonical; CMS3-epithelial/metabolic; and CMS4-mesenchymal/stromal. Finally, the search for effective therapies has been greatly facilitated by the availability of ex vivo drug screening of patient-derived preclinical models in culture using tumour organoid models23.

Biomarker-guided treatment of patients with CRC is currently based on the presence or absence of individual markers that are associated with either prognosis or expected benefit from a specific therapy. However, as the treatment options are expanding, an increasing level of complexity is becoming apparent. Firstly, biomarkers often interact with each other, thus reinforcing the importance of comprehensive molecular profiling (FIG. 1). For example, the experience with therapies targeting alterations in the MAPK signalling pathway illustrates the potential to improve initial response rates and/or to identify effective drug combinations based on features associated with primary resistance. In addition, monitoring the dynamics of resistance-bearing subclones provides the potential for rechallenge with anti-EGFR antibodies in later lines of treatment15,24. Nonetheless, the majority of patients with CRC currently have no targeted therapy options available. Even low-prevalence markers can substantially increase the use of molecularly-guided treatments, as demonstrated by the introduction of anti-PD-1 antibodies for patients with MSI-high (MSI-H)/mismatch repair deficient (dMMR) cancers25. However, other subgroups provide examples of cancers with more than one molecular target, including MSI-H and RAS wild type cancers that also harbour either BRAFV600E mutations or kinase fusions. The presence of several targetable alterations might extend the range of treatment options when faced with resistance, but also emphasizes the need for appropriate treatment sequencing. Secondly, biomarkers might have complex associations with patient outcomes. Discriminating between prognostic and predictive biomarker values26 is sometimes challenging in clinical studies evaluating therapeutic benefit. This prognostic–predictive complexity is partly driven by the search for more effective therapies for patients who have a poor prognosis with standard treatments.

Figure 1. Clinical implications of biomarker interactions in CRC.

Figure 1.

Interactions between established and emerging clinical biomarkers suggest that more comprehensive molecular profiling would improve patient outcomes a. Detection of, and stratification based on genetic and/or clinical features associated with primary resistance, such as alterations in the MAPK signalling pathway, might improve responsiveness to targeted therapies and/or enable the identification of more effective drug combinations. b. The accumulation of low-prevalence ‘actionable’ alterations has the potential to increase the total use of biomarker-guided therapies c. The co-occurrence of more than one ‘actionable’ alteration might enable new treatment options when resistance develops, although the most appropriate treatment sequence and/or drug combinations need to be determined. BRAFi, BRAF inhibitor; ERBB2amp, amplification of ERBB2/HER2; Fusion+, positive for kinase gene fusions; MSI-H, microsatellite instability-high; RASwt, RAS wild-type.

In this Review, we provide an overview of current and emerging biomarkers with therapeutic implications in the treatment and management of patients with CRC. We illustrate biomarker complexity by highlighting interactions between different biomarkers and discuss both potential prognostic and predictive associations (TABLE 1).

Table1.

Biomarkers with therapeutic implications in patients with CRC

Biomarker Cancer stage Prevalence
in relevant
cancer
stage
Biomarker complexity
Patient prognosis Treatment
benefit
Interactions
with
therapeutic
implications
MSI-H/dMMR Stage II >15%46 aFavorable OS in meta-analysis of stage II or III (HR 0.67, 95% CI 0.58-0.7832) and DFS in retrospective analysis of patients with untreated stage II or III colon cancers in randomized trials (HR 0.51, P = 0.009)34. aLack of benefit from 5-FU-based chemotherapy (no effect of treatment on survival in retrospective analyses of patients with stage II or III CRCs in randomized trials; Table 2). Enriched for immunoscore-high tumours, and immunoscore may have superior prognostic value45,90.
Metastatic 3-5%92,93 bInferior OS (HR 1.35, 95% CI 1.13-1.61) and PFS (HR 1.33, 95% CI 1.12-1.57) in retrospective analysis of pooled data from patients treated with standard therapies in randomized trials92. aBenefit from ICIs (ORR 39% and DCR 75%; summarized from 125 patients treated with single-agent ICIs, mostly in prospective single-arm trials16,18,117; Table 3). Enriched for the drug targets BRAFV600Eand kinase fusions83,148. Hypermutated phenotype (including POLE mutations) may be a better predictive marker for ICIs (Table 3).
BRAFV600E Metastatic ~10%211 aMedian OS < ~1 year for patients treated with standard therapies164,165. Inferior OS also after metastasectomy (HR for OS > 2.7, P < 0.01)170,171. Retrospective analyses.. aBenefit from targeted combination therapies in prospective randomized trials with vemurafenib plus irinotecan plus cetuximab (ORR 16%)21, and with encorafenib plus binimetinib plus cetuxumab (ORR 48%)203. Prognostic value limited to MSS cancers in the primary setting150, possibly independent of MSI status in metastatic disease. Response to ICIs if MSI-H (ORR 55% with nivolumab plus ipilimumab122).
HER2 over-expression/ ERBB2 amplification Metastatic ~2%220 - bNo efficacy of anti-EGFR antibodies in retrospective analyses of RAS and BRAF wild-type cancers (HR for PFS ≥2.8, P < 0.001 in patients with amplification versus no amplification)228 ). Predictive value in RAS wild-type cancers.
bBenefit from dual HER2 inhibition in prospective trials with lapatinib plus trastuzumab (ORR 30% and DCR 59%)225, and with trastuzumab plus pertuzumab (ORR 38%)231.
Kinase fusions (ALK/NTRKs/RET/ROS1) Metastatic <2%88 bPoor prognosis on standard therapies in retrospective analyses of selected patient series (HR for OS ≥ 2.17, P < 0.001)248,249. bBenefit from tyrosine kinase inhibitors (responses have been reported in a few patients treated as part of prospective basket/umbrella trials249,253-257). Strongly enriched in sporadic MSI-H and RAS wild-type cancers. Response to ICI has been reported in one patient with MSI-H cancer248.
RAS mutations Metastatic ~40% (Supplementary Table 1) bInferior OS in pooled analysis of randomized trials of standard therapies (HR 1.35, P <0.001313), also after metastasectomy (HR 1.67, P <0.001; meta-analysis315) (Supplementary Table 1). aNo benefit from anti-EGFR antibodies in prospective randomized trials (HR for OS 0.72, P < 0.01 and HR for PFS 0.60, P <0.001 in retrospective analyses of pooled data for patients with RAS wild-type versus RAS-mutant cancers treated with anti-EGFR antibodies29) Mutually exclusive with mutations in other components of the MAPK signaling pathway which may confer resistance to anti-EGFR antibodies in RAS wild-type cancers29.
CMS4-mesenchymal/stromal gene expression subtype Primary and metastatic Primary: ~25%22; advanced stage: >25%345 bInferior OS (HR >1.5, P ≤0.021) in retrospective analyses of patients with primary CRC22,341,342. CMS1 might confer inferior OS compared with CMS4 in metastatic cancers (HR 2.9, P = 0.017)346. bPoor benefit from standard therapies in retrospective analyses of 5-FU337 and oxaliplatin352 in primary cancers, and anti-EGFR antibodies in KRAS wild-type metastatic cancers335,350. Mostly MSS22.
a

Biomarker recommended for clinical testing in this setting.

b

Biomarker currently not recommended for clinical testing in this setting (owing to conflicting data and/or small patient numbers).95% CI, 95% confidence interval; DCR, disease control rate; DFS, disease-free survival; ICI, immune checkpoint inhibitors; ORR, objective response rate; OS, overall survival; PFS, progression-free survival.

Biomarker complexity and MSI

Adjuvant chemotherapy in primary cancers

According to current guidelines, patients with stage II colon cancer with MSI-H and/or dMMR should not be offered adjuvant 5-FU-based chemotherapy27-29. This recommendation is based on a low risk of recurrence and a lack of treatment benefit. The first reports of a favorable prognostic association of MSI in CRC were published in 199330,31, preceding clinical implementation by approximately two decades. This association has now been confirmed for both OS and disease-free survival (DFS) in meta-analyses comparing data from over 1,200 patients with primary MSI-H CRCs to those with MSS CRCs32,33. This association also applies specifically to patients with stage II and III colon cancers who have not received adjuvant chemotherapy, as shown by retrospective analyses of data from randomized trials34,35. The biological basis of this prognostic association is likely the high tumour mutational burden (TMB) owing to MMR deficiency, and in particular the higher number of frameshift mutations in repetitive sequences36-40. This results in the generation of neoantigens capable of activating cytotoxic T cells, and intra-epithelial infiltration of activated lymphocytes causes a strong antitumour immune response41-45.

Approximately 15% of primary CRCs are MSI-H46. However, the prevalence differs according to clinicopathological factors and MSI is more common in older female patients (>70 years of age) with right-sided and poorly differentiated stage II colon cancers35,47-53. A corresponding association with clinicopathological features has also been reported for prognosis, with patients with proximal tumours having a greater relative benefit from MSI than those with distal tumours54. Various studies report stronger effects of MSI in stage II55,56, similar prognostic associations across stages II and III57,58, or even a stronger effect in stage III49 among patients with CRC. However, no statistically significant interactions with clinicopathological features, including tumour stage, were observed in a pooled analysis of data from >7,000 stage II or III colon cancers52, suggesting that the prognostic value of MSI is independent of disease stage.

The prognostic implications of MSI after adjuvant treatment might be confounded by a predictive value for 5-FU-based chemotherapy, thus illustrating prognostic–predictive biomarker complexity51. A loss of MMR function could result in a failure to recognize and respond to the incorporation of 5-FU into tumour DNA59. However, results have been inconsistent in the many retrospective analyses of the effects of 5-FU-based regimens in patients with MSI-H tumours51. Most studies comparing the effects of 5-FU-based chemotherapy versus no treatment have reported no significant improvements in OS or DFS in patients with MSI-H/dMMR tumours (TABLE 2), despite 5-FU-based chemotherapy significantly improving the outcomes of patients with MSS/MMR proficient colon cancer or CRC34,35,58,60,61. However, only two of the seven studies revealed a statistically significant interaction between MSI status and the response to chemotherapy62,63. A meta-analysis including data from almost 400 patients with MSI-H CRCs revealed a substantial degree of heterogeneity with respect to the effects of 5-FU-based chemotherapy, and a lack of benefit could not be definitively confirmed33.

Table 2.

Retrospective analyses of survival outcomes of patients with MSI-H/dMMR stage II or III CRCs receiving 5-FU-based chemotherapy

Study Study design Stage Treatment MSI-H/
dMMR
cancers
(n)
Treatment
benefit in
MSI-
H/dMMR
Interaction
(MSI/dMMR-
status and
treatment)
Ribic et al.63 Pooled analysis of 5 randomized phase III trials II and IIIa Randomization to 5-FU + LV/LEV versus no chemotherapy 95 Worse OS (non-significant) with treatment versus no treatment in MSI-H (HR = 2.14, P = 0.11) Significant (P = 0.01)
Carethers et al.60 Consecutive single-centre series II and III No randomization: 5-FU or no chemotherapy 36 No effect of treatment on OS in MSI-H (P = 0.52) NA (although treatment was associated with improved OS in non-MSI-H [P = 0.048])
Benatti et al.58 Patients from 3 centres II and III No randomization: 5-FU-based or no chemotherapy 206 No effect of treatment on DSS in MSI-H (HR = 0.55, 95% CI 0.20-1.69) NA (although treatment was associated with improved DSS in stage III MSS [P = 0.02], but not in stage II MSS [(P = 0.32])
Lanza et al.61 Consecutive single-centre series III No randomization: 5-FU + FA or no chemotherapy 41 No effect of treatment on DSS in dMMR (P = 0.91) NA (although treatment was non-significantly associated with improved DSS in non-dMMR [P = 0.08])
Jover et al.62 Patients from 10 centres II and III No randomization: 5-FU or no chemotherapy 60 No effect of treatment on OS (69.2% and 73.5% in treated and untreated, P = 0.8) or DFS (57.7% and 67.6% in treated and untreated, P = 0.6) in dMMR Significant (HR = 2 for OS and DFS, P = 0.0001)
Sargent et al.34 Pooled analysis of 5 randomized trials II and IIIa Randomization to 5-FU + LV/LEV versus no chemotherapy 70b No effect of treatment on DFS in dMRR (multivariable HR = 1.39, P = 0.56) Non-significant (P = 0.18; although treatment was associated with improved DFS in MMR proficient [HR = 0.67, P = 0.02])
Hutchins et al.35 Randomized phase III trial II and III (mainly II) No randomization: 5-FU + FA or no chemotherapy 218 No effect of treatment on risk of recurrence in dMMR (OR = 0.8, 95% CI 0.29-2.22) NA (although treatment was associated with lower risk of recurrence in MMR proficient [OR = 0.59, 95% CI 0.46-0.78])
a

Colon cancer only

b

Only patients independent from Ribic et al., N Eng J Med 2003 are included. 5-FU, 5-fluorouracil; 95% CI, 95% confidence interval; DFS, disease-free survival; DSS, disease-specific survival; FA, folinic acid; HR, hazard ratio; LEV, levamisole; LV, leucovorin; NA, not analyzed; OR, odds ratio; OS, overall survival.

MSI-status is not predictive of a lack of benefit from the combination chemotherapies commonly used in patients with CRC. The addition of oxaliplatin to 5-FU-based adjuvant chemotherapy regimens (FOLFOX/FLOX or CAPOX) has been shown to improve patient survival64, and this is also seen separately for patients with dMMR stage II or III colon cancers65,66. Furthermore, patients with dMMR tumors have improved survival outcomes compared to the MMR proficient subgroup after treatment with FOLFOX67,68 (dependent on tumour location in one study54,69), and this is consistent with the favorable prognostic effect of MSI. Irinotecan is also part of the chemotherapy regimens received by patients with metastatic CRC, although this agent is not used in the adjuvant setting for those with primary CRC70-72. Data from studies investigating the predictive value of MSI status are again conflicting, indicating both a survival benefit73 and a lack of benefit56 from the addition of irinotecan to 5-FU. Preclinical investigations of the effects of these agents are challenging, partly owing to differences in the in vitro and in vivo drug metabolism. These differences have been illustrated by discordant levels of 5-FU sensitivity in matched patient-derived organoid and xenograft (PDX) models74. In conclusion, the complex prognostic–predictive association between MSI and benefit from adjuvant chemotherapy in patients with primary CRC is not fully understood. Nonetheless, adjuvant chemotherapy is not recommended for patients with low-risk MSI-H/dMMR stage II CRCs owing to the generally good prognosis of such patients and a lack of treatment benefit27. In those with stage III disease, the risk of recurrence is higher, therefore, patients should receive standard chemotherapy irrespective of MSI status.

Heterogeneity and immunity

MSI-H CRCs have a distinct clinicopathological, biological and molecular profile51; nonetheless, these tumours are still heterogeneous. The hypermutated phenotype seen in MSI-H tumors (>10 mutations per megabase (mut/Mb)75) might augment this heterogeneity, and MSI-H CRCs are more heterogeneous in terms of point mutations and indels compared with MSS cancers, according to both intratumoral analyses76 and comparisons of biopsy material from metastatic cancers with matched circulating tumour DNA77. MMR deficiency in sporadic MSI-H tumours is primarily caused by promoter hypermethylation of MLH1, but MSI also occurs in a hereditary setting, resulting from germline mutations in the MMR genes and causing hereditary nonpolyposis CRC (HNPCC) syndrome78. Sporadic MSI can be further distinguished from HNPCC by the presence of the CpG island methylator phenotype (CIMP) and frequent BRAF mutations79. The implications of this distinction for patient outcomes and possible treatment benefit are uncertain57,58,66,80-83, and this information is not included in adjuvant chemotherapy guidelines84. Furthermore, loss of expression of the transcription factor CDX2 is common in MSI-H tumours and, in contrast to MSI, is a proposed biomarker of benefit from adjuvant chemotherapy85. Clinical data for this association are currently limited to retrospective analyses, but preclinical data indicate higher sensitivity of CRC cell lines harbouring a loss of CDX2 expression to several chemotherapies86, and further clinical investigation is warranted to determine the precise interpretation of the co-occurrence of MSI and loss of CDX2 expression in this setting. Sporadic MSI-H tumours also interact with other prominent CRC biomarkers and therapeutic targets, such as oncogenic kinase fusions, which are mutually exclusive to BRAF mutations. Kinase fusions are generally rare in patients with CRC (<1-2%), although they have been reported in 55% of BRAF and KRAS wildtype MSI-H tumours harbouring MLH1 methylation83,87,88.

Most, but not all, MSI-H tumours are of the immunogenic gene expression-based consensus molecular subtype CMS1-immune22. The level of immune cell infiltration in MSI tumours seems to differ according to these gene expression subtypes and to be particularly high in CMS1. This distinction is supported by a potentially favorable prognosis with MSI-H CMS1 tumours compared with MSI-H CMS2–489. Variations in the extent of tumour immunity among patients with MSI-H CRCs have also been clearly demonstrated by the immunoscore, a standardized immunohistochemistry-based scoring system that summarizes the density of tumour infiltrating T cells (CD3+ and CD8+) in the tumour centre and at the invasive margins. This approach provides a level of prognostic discriminatory power superior to that of MSI in primary CRC90. In 2018, an international multicentre consortium demonstrated the additive value of the immunoscore beyond that of clinicopathological prognostic factors and MSI status among 2,681 patients with stage I–III colon cancers45. This prompted the development of an attempt to integrate immunoscore with TNM staging to better guide adjuvant treatment decision-making. This approach seems particularly appealing considering the power of immunoscore in determining the prognosis of patients with stage II MSS CRCs who had not received adjuvant chemotherapy91.

Immunotherapy in metastatic cancers

Owing to associations with a more favourable prognosis, MSI is less common among patients with metastatic disease compared to those with primary CRCs and is typically reported in ≤5% of patients with metastatic CRC in clinical trials92-94. T cell infiltration is also a positive prognostic factor in metastatic disease, even after conventional chemotherapy and/or surgical resection95-98. However, heterogeneity in the density of immune-cell infiltration has been reported in comparisons of biopsy material from matched primary tumour and liver metastases99,100, as well as between liver metastases from individual patients96. Paradoxically, metastatic MSI-H tumours are aggressive and have been found to be associated with inferior PFS and OS outcomes relative to those of patients with metastatic MSS CRC in several studies92,101-103. While other reports indicate no prognostic associations and additional data are needed, the survival benefits associated with primary MSI-H CRCs seem to be lost in the metastatic setting104-106.

Several mechanisms have been proposed in an attempt to explain the paradoxical effects of MSI status on patient outcomes, including variations in patterns of metastatic spread between MSI-H and MSS cancers, with lower rates of liver metastases and higher rates of peritoneal metastases in MSI-H80,94,101,107. The paradoxical effect might also be driven by enrichment with BRAF mutations in MSI-H92,101,108. Curative surgical removal of metastases (metastasectomy) can improve the OS of patients with MSI-H CRCs109, but these patients might be less likely to undergo surgery, partly owing to a lack of benefit from conversion chemotherapy102. Whether MSI-H confers chemoresistance in patients with metastatic disease remains unclear11, although ‘tumour-sidedness’ has been identified as a prognostic factor in clinical trial cohorts receiving chemotherapy and/or targeted agents110-113: those with cancers originating from the right side of the colon have inferior outcomes. The prevalence of MSI-H, like multiple other molecular features, differs in a gradient-like fashion along the anatomical sections of the colorectum114, and although the independent prognostic contribution of each clinicopathological and biological factor is not clear, collectively, these data support inferior outcomes for patients with MSI-H metastatic disease. Metastasizing MSI-H cancer cells might also be particularly effective at immunoediting and subsequently to evade immune surveillance115. However, a dependency on immunoediting for continued growth could also render tumours more sensitive to treatments directed towards the immune microenvironment.

The first tissue-agnostic approval of a cancer therapy based on the presence of a molecular marker was granted to the anti-PD-1 antibody pembrolizumab in patients with advanced-stage MSI-H/dMMR solid tumours116. The introduction of this biomarker-driven approach was responsible for a jump in the use of molecularly-guided cancer therapies in 2017 (0.33% increase in number of patients eligible for genomically informed therapies)25. Three drugs are currently approved for patients with metastatic and chemotherapy-refractory MSI-H/dMMR CRCs based on data from phase II clinical trials16,18,117; pembrolizumab, another anti-PD-1 antibody, nivolumab, as well as the anti-CTLA4 antibody ipilimumab. Results from the totally 125 patients with MSI-H/dMMR treatment-refractory metastatic CRCs who received anti-PD-1 antibodies alone indicate an overall objective response rate (ORR) and disease control rate (DCR) of 39% and 75%, respectively (TABLE 3). Promisingly, the majority of responses were durable. The median PFS16,117, or median duration of response18, was not yet reached after a median follow-up of >36 weeks. Two randomized phase III trials evaluating the efficacy of immune-checkpoint inhibitors (ICIs) as a first-line treatment in patients with metastatic MSI-H/dMMR CRCs are currently ongoing. The KEYNOTE-177 study118 is designed to compare the efficacy of single-agent pembrolizumab with that of investigator’s choice of chemotherapy119. Similarly, COMMIT120 is designed to compare the efficacy of the anti-PD-L1 antibody atezolizumab, either as a single-agent or in combination with a standard treatment regimen (FOLFOX and a VEGF targeted therapy), to standard treatment alone. Additionally, the possibility of treatment benefit in the adjuvant setting is being evaluated in a randomized phase III trial comparing the efficacy of adjuvant FOLFOX with and without atezolizumab in patients with dMMR stage III colon cancers121.

Table 3.

Data from clinical studies of immune-checkpoint inhibition in hypermutated metastatic CRCs

Study Study design Agent Hypermutated
metastatic CRCs
(n)
ORRa DCRb
Monotherapies
Lipson et al. (2013)372 Single-arm phase I trial in patients with treatment-refractory solid tumours Nivolumab 1 MSI-H Durable complete response NA
Le et al. (2015)16 Single-arm phase II trial in patients with treatment-refractory metastatic MSI-H CRCs, MSS CRCs and MSI-H other Pembrolizumab 10 MSI-H (HNPCC and sporadic) 40% 90 %
Le et al. (2017)117 Single-arm phase II of treatment-refractory metastatic MSI-H/dMMR cancers from 12 cancer types Pembrolizumab 40 MSI-H/dMMR (HNPCC and sporadic) 52% 82 %
Overman et al. (2017)18 Reporting from monotherapy arm of a phase II study of treatment-refractory metastatic MSI-H/dMMR CRCsc Nivolumab 74 MSI-H/dMMR (ongoing) 31% 69 %
Gong et al. (2017)19 Case report of treatment-refractory MSS metastatic CRC Pembrolizumab 1 POLE-mutated Clinical response NA
Fabrizio et al.(2018)133 Case report of treatment-refractory MSS metastatic CRC Pembrolizumab 1 POLE-mutated Radiographic response NA
Combination therapies
Overman et al. (2018)122 Reporting from combination therapy arm of a phase II study of treatment-refractory metastatic MSI-H/dMMR CRCsc Nivolumab plus ipilimumab 119 MSI-H/dMMR (ongoing) 55% 80 %
Lenz et al. (2018)123 First-line treatmentc Nivolumab plus ipilimumab 45 MSI-H/dMMR (ongoing) 60% 84%
Hochster et al. (2017)373 Reporting from one treatment arm of a phase Ib study Atezolizumab plus bevacizumab 10 MSI-H 30% 90 %

CRC, colorectal cancer; DCR, disease control rate; HNPCC, hereditary nonpolyposis colorectal cancer; MSI, microsatellite instability; MSS, microsatellite stable; NA, not applicable; ORR, objective radiographic response rate

a

Complete or partial response according to RECIST1.1.

b

Complete/partial response or stable disease according to RECIST1.1.

c

Same clinical study.

Data published in 2018 show an apparent synergistic effect of ICIs when used in combination, including anti-PD-1/PD-L1 antibodies with anti-CTLA4 antibodies. Among 119 patients with chemotherapy-refractory MSI-H/dMMR metastatic CRCs receiving nivolumab and ipilimumab, 55% and 80% had objective responses and disease control, respectively (TABLE 3). Again, the responses were durable, lasting ≥6 months in 83% of patients122, and were found to be independent of poor prognostic factors such as the presence of BRAF and KRAS mutations. Initial results from 45 patients treated with this combination in the first-line revealed objective responses and disease control in 60% and 84%, respectively123. Promising preliminary data with the same combination have been presented also in the neoadjuvant setting in patients with early stage colon cancers, showing pathological responses in all seven patients with MMR deficiencies, including four complete responses124. A similar combination of durvalumab and tremelimumab is currently under investigation, irrespective of MSI-status125,126. Based on the successes and the level of interest with which ICIs are currently being investigated, the indications for use of these agents are likely to soon extend beyond patients with treatment refractory metastatic cancers with MSI-H/dMMR. However, primary resistance remains common and further research into the determinants of response and resistance in patients with MSI-H cancers is needed in order to improve patient selection for treatment.

Optimization of the use of immunotherapy.

The sensitivity of MSI-H tumours to ICIs is attributed to the hypermutated phenotype16. Mechanisms of response mirror those that explain the favourable prognosis of patients with primary MSI-H CRCs, involving the generation of cancer-specific neoantigens and subsequent activation of cytotoxic T cells76,127,128. The immunosuppression induced by cancers with a high TMB as a protective trait is, to a large extent, mediated by upregulated expression of PD-L1, as well as immunomodulatory receptors on T cells including CTLA4 and PD-1129,130. ICIs target this ligand–receptor interaction and reactivate T cell responses to tumour-associated antigens131. Consequently, treatment responses are expected also in cancers with a hypermutated phenotype that is unrelated to MSI. A prominent example is provided by MSS tumours with defective replication repair caused by mutations in the proofreading domain of the DNA polymerase POLE. Mutations in this enzyme often lead to an ‘ultra-hypermutator’ phenotype with a TMB >100 mut/Mb, exceeding that of MSI-H tumours (>10 mut/Mb)75, and with a similar extent of cytotoxic T cell infiltration132. Responses to pembrolizumab have been documented in two case reports describing patients with POLE-mutated MSS CRCs19,133 (TABLE 3), and additional clinical data are awaited from an ongoing study investigating the efficacy of the anti-PD-L1 antibody avelumab in patients with MSI-H or POLE-mutated CRC134. However, patients with non-metastatic POLE-mutated tumours also have a favourable prognosis compared to those with MSS CRCs132, and the prevalence of POLE mutations is <1% in patients with advanced-stage CRCs135. This observation suggests that this biomarker has only a modest level of potential to extend the indications for use of ICIs.

A high TMB (>10-20 nonsynonymous mut/Mb) is emerging as a separate positive predictive biomarker of benefit from ICIs in several cancer types136,137. The majority of hypermutated CRCs are identified by the presence of either MSI or POLE mutations, although an increased TMB has been found in 3% of MSS tumours, of which only 21% could be attributed to POLE133. A report showing a complete and durable response to nivolumab in one patient with POLE wild-type MSS cancer with an elevated TMB suggests the potential to refine the criteria for hypermutated CRCs in relation to treatment133. The criteria for hypermutation will need to be carefully adjusted based on additional clinical data, and the most appropriate TMB threshold is likely to vary across different cancer types, as well as according to the gene panel used for sequencing136. The presence of a pre-existing CD8+ cytotoxic T cell antitumour immune response might be a prerequisite for response to anti-PD-1 antibodies138 and tumour immunoreactivity provides another possible strategy for the refinement of patient eligibility criteria. Immunoscore enables the identification of cancers with an in situ adaptive immune response and is a potential biomarker. Up to 12% of patients with primary MSS CRCs that later recur might be classified as immunoscore high (>70% density of CD3+ and CD8+cells)139, although whether this is reflected in responsiveness to immune-checkpoint inhibition in metastatic lesions has yet to be shown.

Investigators attempting to optimize the clinical efficacy of ICIs in patients with CRC should have two immediate objectives (FIG. 2): identification of the mechanisms of innate resistance in patients with hypermutated and/or immunogenic cancers and; identification of the most effective combination strategies, in particular for patients with tumours of an immunologically ‘cold’, non-hypermutated phenotype140. Whether MSI is the result of either sporadic or hereditary (epi-)genetic alterations does not seem to affect the responses of patients to treatment117,122. However, a range of mechanisms of resistance have been proposed in patients with various cancer types, and several of these are likely to apply to patients with CRC. These can be broadly classified into one of four categories, including defects in the antigen presentation machinery (for example loss of expression of TAP, B2M and HLA molecules), insensitivity to cytotoxic T cells (for example owing to loss of IFNγ signalling), activation of inhibitory immune checkpoints (such as CTLA-4, PD-1 and others, including lymphocyte activation gene 3 protein (LAG-3) and V-type immunoglobulin domain-containing suppressor of T-cell activation (VISTA)), and infiltration of immunosuppressive cells (such as regulatory T cells and tumour-associated macrophages) into the tumour140.

Figure 2. Optimization of immunotherapy in CRC.

Figure 2.

Many of the genetic and/or clinical features that determine responsiveness to immune-checkpoint inhibitors (ICIs) (inside circle) are associated with genotypes and phenotypes (outside circle) that can be modulated. In patients with CRC, clinical (red and blue text) or pre-clinical (white text) data are available on a few biomarkers and/or mechanisms that might enable the modification of treatment responses. The best-described mechanisms of resistance in patients with hypermutated and/or immunogenic cancers include loss of IFNγ response owing to JAK1 mutations. The potential to promote immune-cell infiltration is strongest in tumours with an immunosuppressive phenotype, although limited clinical data are available on this possibility in patients with CRC. Experimental data suggest that chemotherapies, inhibition of TGFβ, as well as epigenetic modifiers that target MDSCs might all promote immune-cell infiltration. The expansion of this simplified model is an important task for the optimization of ICIs in the coming years. CMS, consensus molecular subtypes; CTL, CD8+ cytotoxic lymphocyte; IFNγ, interferon-gamma; MDSC, myeloid-derived suppressor cells; MSI-H, microsatellite instability-high; MSS, microsatellite stable; TGFβ-i, TGFβ inhibition; Th1, T helper 1 cell; T-reg, regulatory T cell; white arrow up/down; upregulation or increased levels/down-regulation or decreased levels.

The IFNγ signalling pathway has a key role in responsiveness to ICIs, and the loss of IFNγ signalling is one of few mechanisms of resistance that have been recognized to exist in patients with CRC. This mechanism arises from homozygous or hemizygous loss-of-function mutations in JAK1, eventually leading to a loss of PD-L1 expression141 (FIG. 2). Mutations in JAK1 occur in 20% of patients with primary MSI-H CRCs, and are associated with transcriptional profiles that are predictive of resistance to ICIs142. However, these alterations are also associated with a more favorable prognosis in the primary cancers, and are therefore likely to be less common among patients with metastatic disease89. Furthermore, anti-PD-1 antibodies have been shown to be effective in tumours harbouring loss-of-function mutations in one allele of JAK1, thus supporting the need for a ‘second hit’ and complete loss of JAK1 function to confer resistance. Preclinical data indicate that IFNγ secretion might be increased by treatment with an anti-LAG-3 antibody, and that this causes proliferation of isolated T cells, similarly to treatment with anti-PD-1 antibodies143. Evidence from patients with melanoma indicates that mutations in other genes involved in IFNγ signaling might also affect responsiveness to ICIs144. Another example is provided by mutations in STK11, which are associated with T cell depletion and primary resistance to anti-PD-1 antibodies in patients with KRAS-mutant lung cancer145. In summary, no robust biomarkers that might enable treatment stratification within the subgroup of patients with MSI-H CRCs are currently available. Moving forward, patient monitoring with repeated longitudinal sampling and analysis of immune signatures during treatment will likely be a successful strategy for identifying additional mechanisms of resistance as they occur146.

Targeting poor prognostic BRAF mutations

Guidelines for molecular testing in patients with metastatic CRC include assessment of BRAF mutation status for prognostic stratification29. The prognostic value of BRAFV600E is a prominent example of complexity caused by biomarker interactions147. The mutations are strongly enriched in, and occur in up to 60% of sporadic primary MSI-H CRCs, but in no more than 5–10% of MSS CRCs148-153. Consequently, the majority of epidemiological and clinicopathological associations are similar to those of the MSI subtype154, and in primary CRC, the poor prognosis associated with this alteration might be limited to those with MSS disease150. Retrospective analyses of clinical trial data show that the BRAFV600E confers inferior OS68,155,156 and DFS outcomes54,82,156 in patients with stage II and III MSS colon cancers, and the MSS-dependent poor prognostic value of BRAFV600E has also been shown in population-based series of CRCs157,158. The MSI–BRAF interaction is supported also at the level of gene expression, and BRAFV600E has a greater impact on mutation-associated gene expression patterns in MSS compared with MSI-H CRCs158. This effect might be related to the different tumorigenic roles of this mutation in the two subtypes. ‘Classical’ CRC tumorigenesis is initiated by activation of WNT signalling, eventually causing the development of MSS tumours that are chromosomally unstable159,160. In this model, BRAFV600E mutations are mutually exclusive to the more frequent KRAS mutations, which contribute to adenoma development but are not required for adenoma initiation161. However, in CIMP and MSI-H tumours arising from serrated polyps, BRAFV600E mutations are tumour-initiating events79,162,163. A role in the formation, rather than the development of MSI-H tumours provides a potential rationale for the weaker prognostic associations of BRAFV600E in this tumour subtype. Nonetheless, the size of the prognostic effect in MSS cancers has led to calls for stratification according to BRAF and MSI status in future trials involving adjuvant treatments29,156.

The association between BRAFV600E and survival in primary CRC might primarily reflect reduced OS after disease relapse, rather than an increased risk of relapse68,149, indicating a prognostic effect also in metastatic cancers. Furthermore, the dependence on MSI status seems to change in the metastatic setting, and here BRAFV600E also has prognostic relevance in patients with tumours of the MSI-H subtype109. This difference might reflect the more aggressive biology of MSI in metastatic disease. The median OS of patients with metastatic CRCs harbouring BRAFV600E is commonly reported to be <10 months164, but might reach 13 months depending on treatment165. The mutation affects both PFS and OS109,154,164-168 and survival after metastasectomy166,169-171. Consequently, the current level of benefit from standard therapies is inadequate29,172, and oxaliplatin-based and irinotecan-based therapies both seem to confer similar outcomes173,174. Improved survival and response rates might be obtained by the use of combination therapies in the first-line setting, including triplet or doublet chemotherapy (FOLFOXIRI or FOLFIRI) in combinations with VEGF targeted therapies (bevacizumab)165,175, although small cohort sizes preclude the statistical significance of this observation. Early indications suggested that BRAFV600E was predictive of a lack of response to EGFR targeted therapies (cetuximab or panitumumab) in the RAS wild type population176,177. This observation is consistent with the biological rationale that BRAF is a principal effector of KRAS signalling. However, as summarized in a review published in 2017172, several retrospective analyses of BRAFV600E in data from randomized trials exploring the efficacy of EGFR targeted therapies have been conducted94,178-185, and only one186 was able to confirm this association. Data from meta-analyses have either indicated a negative predictive value187, or concluded that insufficient evidence exists to support such an association188. The presence of BRAFV600E is not a contraindication for EGFR targeted therapy, according to guidelines published in 201729.

Considerable effort has been applied to the development of effective treatments for patients with BRAFV600E-mutant CRCs and the strongest clinical benefits have been achieved with agents targeting the mutated protein itself. Monotherapy with the BRAF inhibitor vemurafenib has not mirrored the encouraging initial response rates obtained in patients with melanoma189,190, although several targetable mechanisms of resistance have been identified, which has paved the way for more effective combination therapies. Re-activation of EGFR signalling is perhaps the most prominent mechanism of resistance191-193, and clinical studies evaluating the efficacy of different combinations of BRAF and EGFR targeted therapies have revealed disease regression in 52%190 and 67%194 of patients who have previously progressed on other treatments. However, formal criteria for a partial response were not met in all patients, and response rates according to RECIST have been lower and reported in the range of 4–22%, potentially related to selection of resistance190,194-197. Nevertheless, BRAF and EGFR targeted agents in combination with chemotherapy (vemurafenib, irinotecan and cetuximab)20 are now included in treatment guidelines for patients with treatment-refractory BRAFV600E-mutant metastatic CRC84. This recommendation is based on initial data from a randomized phase II trial that revealed superior PFS and an ORR of 16% among patients in the triple therapy arm, compared to 4% in those who did not receive vemurafenib21. Final results from this study are awaited.

Activation of the PI3K/AKT signalling pathway is another potential mechanism of resistance to BRAF inhibition193. A triplet including BRAF, EGFR and PI3K targeted agents (encorafinib, cetuximab and alpelisib) has been shown to prolong PFS in patients with treatment-refractory disease, relative to encorafinib plus cetuximab alone in an interim analysis of a randomized phase II trial197. However, the difference in survival was not statistically significant, and the triplet also increased the risk of adverse events (grade 3–4 adverse events occurred in 79% versus 58% of patients).

MEK is a downstream effector of BRAF, and responses to the combination of BRAF and MEK inhibition (with dabrafenib plus trametinib) have been reported in 12% of a cohort of 43 patients198. These outcomes do not parallel those of patients with melanoma199, although in a phase I study involving a triplet including an EGFR targeted agent (dabrafenib, panitumumab and trametinib), responses were achieved in 21% of patients, compared to 10% with dabrafenib plus panitumumab alone200,201. Results are awaited from the ongoing randomized phase III BEACON CRC study, in which the efficacy of encorafinib plus cetuximab, with or without the MEK inhibitor binimetinib will be evaluated in comparison with a control arm, in which patients are receiving chemotherapy plus cetuximab202. Early results from the safety lead-in phase of this trial show an ORR of 48% with good tolerability among the 29 patients receiving the triplet combination203. If similar outcomes are observed in the randomized phase, this combination will be confirmed as having unprecedented potential to improve the outcomes of patients with this aggressive CRC subtype. However, any improvements in PFS are likely to be limited to only a few months200,203, and an improved understanding of the biology of BRAFV600E-mutant CRC could enable the development of even more effective strategies.

Identifying additional targets

Tumours of the immunogenic CMS1-MSI/immune subtype are strongly enriched with BRAFV600E mutations22. This high prevalence is related not only to the co-occurrence of these mutations with MSI, but also to enrichment at a similar magnitude observed specifically among CMS1 MSS cancers, with a mutation frequency of 34%158. This observation suggests that inflammation and immunogenicity are defining characteristics of BRAF mutated CRCs. Interestingly, the combination of nivolumab plus ipilimumab resulted in an ORR of 55% in patients with BRAF-mutant MSI-H CRCs122. This response rate is the same as that of the overall MSI-H cohort, and is a very promising result for patients with BRAFV600E-mutant disease. If these data are confirmed in ongoing studies, the co-occurrence of MSI and BRAFV600E could become defined as a ‘double-target’ subgroup with a need for therapy prioritization between ICIs and BRAF targeted agents. In this setting, the longer duration of response achieved with ICIs compared to that obtained with BRAF inhibition will be an important consideration. Furthermore, improved characterization of the immune context of BRAFV600E metastatic CRCs is an important strategy for the potential to design combinations therapies including both ICIs and BRAF targeted agents.

Gene expression profiling has enabled the BRAFV600E-mutant phenotype to be extended to a subpopulation of BRAF wild type CRCs with a similarly poor prognosis204. Preclinical data suggest that the subgroup of tumours with this ‘BRAF-like’ gene expression pattern might be particularly vulnerable to silencing of the microtubule regulator RANBP2 and repurposing of the tubulin-binding agent vinorelbine205. However, in a phase II trial, in which patients with BRAFV600E metastatic CRC received vinorelbine, no clinical activity was reported and this treatment strategy has not been translated into clinical use206. Gene expression subtyping offers another potential method of stratifying patients with BRAFV600E-mutant disease who might benefit from combined BRAF plus MEK inhibition. BRAFV600E CRCs can be divided into two distinct gene expression subtypes: one subtype confers a poor prognosis and is characterized by activation of KRAS/AKT signalling and epithelial-to-mesenchymal transition (EMT), but with potentially greater sensitivity to BRAF plus MEK inhibition than the second subtype, which is characterized by cell-cycle dysregulation207. Clinical testing is required to evaluate these putative associations, which are currently derived from CRC cell lines.

Tumour heterogeneity is likely to have a role in acquired resistance to BRAF-targeted therapies. This association has been demonstrated in a case report describing a patient with expansion of a MET-amplified subclone during combination therapy with panitumumab and vemurafenib208. This case report revealed a new potential treatment approach following a successful switch to combination therapy with the ALK and MET inhibitor crizotinib plus vemurafenib, although the patient later also progressed on this drug combination owing to MET hyperamplification209. Acquired resistance to such regimens of two or three targeted therapies often leads to reactivation of the MAPK signalling pathway. Combined inhibition of EGFR, BRAF and ERK signalling therefore provides a new strategy that might circumvent the development of acquired resistance210. These studies clearly demonstrate that genomic monitoring of patients’ disease during treatment is a powerful approach that is likely to improve disease management, based on the early detection of treatment-resistant subclones.

Rare BRAF mutations

Approximately 2% of metastatic CRCs have BRAF mutations located outside of the hotspot in codon 600, most frequently in codon 594211,212. In contrast to BRAFV600E, some of these mutations might lead to inactivation of the kinase213, co-occur with RAS mutations, and are not enriched in MSI-H CRCs. Consequently, rare BRAF alterations have distinct clinical associations, including a propensity for left-sided or rectal primary locations and fewer peritoneal metastases, relative to BRAFV600E-mutant CRCs211,214. These tumours also confer a favorable prognostic association, at least in comparison with BRAFV600E-mutant CRCs (median OS approximately 60 months among patients with non-V600E BRAF-mutant metastatic CRCs)211,214. This association has also been observed in patients undergoing surgery for CRC liver metastases171. Insufficient data are available to determine whether non-V600E mutations confer an absence of poor prognostic associations, or also a better prognosis than wild-type BRAF. Differences in prognosis might be related to the specific codon affected215. Nonetheless, patients with these cancers might not need the same aggressive treatments as those with BRAFV600E-mutant disease, although clinical trials are required in order to determine the optimal treatment approach, and again this might depend on the specific mutated codon212. Patients with cancers harbouring certain non-V600E mutations in BRAF might benefit from EGFR inhibition, although currently available data are inconclusive216-218. Responses to approved BRAF inhibitors are unlikely, considering that these inhibitors bind to and inhibit monomeric BRAF, which is seen only with V600E mutations212.

Complexity among emerging biomarkers

HER2 overexpression

HER2 overexpression can be found in approximately 20% of breast cancers. This biomarker is associated with a poor prognosis following treatment with standard therapies, but improved outcomes with HER2-targeted agents219. In CRC, HER2 overexpression has a limited prevalence, occurring in approximately 2% of patients. This effect is caused by ERBB2 amplification in >90% of the patients220. The prognostic value of HER2 overexpression in patients with CRC remains uncertain220,221, although HER2 might have a dual predictive value relating to response to targeted therapies.

HER2 can activate the MAPK signalling pathway and preclinical data suggest that ERBB2 amplification is involved in both primary and acquired resistance to EGFR inhibition222,223. The prevalence of HER2 overexpression in this therapeutically relevant setting, among patients with RAS or BRAF wild type metastatic CRCs, is increased to approximately 5%220,224,225. However, clinical data are limited to retrospective analyses of a small number of patients. Data are available on the effects of ERBB2 amplifications among patients with KRAS wild type solid tumours treated with anti-EGFR monoclonal antibodies alone or in combination with chemotherapy223,226; from patients with RAS and/or BRAF wild type cancers receiving anti-EGFR antibodies after failure of first-line chemotherapy227,228; and from 15 patients with HER2 overexpressing cancers refractory to previous anti-EGFR therapies225. These data consistently confirmed a negative predictive value of HER2 overexpression, with a similar magnitude of effect on PFS after EGFR blockade to that of RAS mutations. However, prospective studies are needed in order to confirm any negative associations between HER2 overexpression and responses to anti-EGFR therapy.

The suggested dual predictive role of HER2 overexpression stems from the potential to directly target this protein. The first evidence of clinical responses to HER2 targeted therapies in patients with metastatic, HER2-overexpressing CRCs was obtained more than a decade ago, in combination with oxaliplatin-based229 or irinotecan-based chemotherapies230. However, both of these phase II trials were prematurely terminated owing to limited accrual of patients with the relevant biomarkers. Later preclinical studies suggested that dual inhibition improves the efficacy of HER2-targeted therapies. Long-lasting tumour regressions were achieved in cetuximab-resistant PDX models of liver metastases with wild type RAS, BRAF and PIK3CA harbouring ERBB2 amplifications following combined treatment with the dual HER2/EGFR inhibitor lapatinib and antibodies targeting either HER2 (pertuzumab) or EGFR (cetuximab), but not with monotherapies222. Encouraged by these preclinical data, the phase II HERACLES trial was conducted to evaluate the efficacy of dual HER2 inhibition with lapatinib plus trastuzumab in chemotherapy and anti-EGFR antibody refractory KRAS wild type, HER2-positive metastatic CRCs225. Objective responses were seen in 30% of the 27 eligible patients, with disease control in 59%. Furthermore, a combination of the two anti-HER2 antibodies, trastuzumab and pertuzumab, enabled objective responses in 38% of the 37 patients with HER2-positive metastatic CRCs enrolled in the MyPathway phase II basket study231. Confirmatory case-reports describing durable responses to trastuzumab monotherapy232 and in combination with chemotherapy, lapatinib or pertuzumab have also been published233,234. Therapies targeting HER2 in patients with HER2-positive metastatic CRC are currently considered investigational, although enrollment of such patients in clinical trials is encouraged84 and offers a therapeutic option for those with resistance to EGFR targeted therapies. The optimal timing of HER2 inhibition in relation to use of standard-of-care chemotherapies also needs to be addressed in future studies, in addition to the efficacy of this approach as an alternative to EGFR-targeted therapies in RAS wild type and HER2-overexpressing cancers.

Initial clinical investigations demonstrated that primary resistance to dual HER2 inhibition is a frequent occurrence in patients with CRC, and the development of resistance during treatment is almost inevitable. An initial report suggested that ERBB2, RAS and PIK3CA mutations are involved in the development of resistance235, although further genomic analyses of biopsy samples from relevant clinical cohorts is currently awaited. An antibody–drug conjugate (ADC) that combines trastuzumab with the cytotoxic agent emtansine has shown efficacy in patients with trastuzumab-resistant CRCs236,237. This agent is currently being evaluated in the HERACLES B and RESCUE trials, the latter including patients who progressed in the initial HERACLES study238. Preliminary data from an ongoing phase I trial239 combining another ADC, trastuzumab deruxtecan, which combines trastuzumab with a DNA topoisomerase I inhibitor show an ORR of 25% and a DCR of 83%, respectively, among 12 patients with HER2-expressing, KRAS wild type advanced-stage CRCs240. Translational studies designed to identify synergistic combination partners will be crucial to further improvements in the level of clinical benefit derived from HER2 inhibition, although such approaches are challenged by the limited prevalence of this biomarker.

In addition to amplifications, ERBB2 is also targeted by point mutations or indels in an additional 2% of metastatic CRCs233. These aberrations might be associated with an inferior prognosis241, and preclinical data indicate that activating mutations and amplifications have similar therapeutic implications. Resistance to cetuximab has been demonstrated in KRAS wild type cell lines and PDX models of CRCs harbouring activating ERBB2 mutations242,243. Monotherapy with HER2 inhibitors has no clinical efficacy in patients with these mutations244, although preclinical data suggest that dual inhibition of HER2 signalling might be more effective243. Accordingly, point mutations and indels add to the complexity of ERBB2/HER2 as a biomarker in CRC.

Oncogenic addiction to kinase fusions

In November 2018 the tropomyosin kinase (TRK) receptor inhibitor larotrectinib was granted accelerated FDA approval for patients with metastatic solid tumours harbouring NTRK1/2/3 (NTRK) fusions245. Of note, this was the second tissue-agnostic FDA approval of a cancer therapy. Gene fusions resulting in increased kinase activity and subsequent oncogene addiction are, arguably, among the most obvious targetable vulnerabilities. These kinase fusions are likely to be intimately associated with disease progression, although the limited prevalence of these alterations in CRCs creates a challenge for those attempting to analyze possible clinical associations. Kinase fusions have been reported in <1-2% of patients with CRC and primarily involve RET, NTRK, ALK or ROS1 (in addition to BRAF and FGFRs)88,246,247. Nonetheless, in an analysis of 27 metastatic CRCs harbouring ALK, NTRK, or ROS1 rearrangements identified from molecular screening programs, rearrangements were found to be most prevalent among patients with right-sided MSI-H RAS wild type tumours248. In comparison with a set of 319 fusion-negative cancers collected through similar means, the presence of these rearrangements also conferred an inferior prognosis including shorter median OS, independent of both MSI status and primary tumour location. Similar results were found by a study with a similar design in a cohort of 24 patients with metastatic CRCs harbouring RET fusions: shorter median OS was observed independent of MSI status and primary tumour location249.

Associations between kinase fusions and inferior outcomes might be augmented by poor responses to standard therapies. Preclinical analyses have shown that excessively high expression of ALK, NTRK and RET is associated with primary resistance to EGFR targeted therapy in RAS and BRAF wild type CRC cell lines250. Later studies have confirmed disease progression on such treatments in fusion-positive cancers248,249,251. Preclinical analyses have also suggested that selective tyrosine-kinase inhibitors (TKIs) might be effective250,252, and clinical efficacy has been confirmed in a few studies. These observations include disease regression in three of four patients with NTRK fusion-positive colon cancers who received larotrectinib as part of a basket trial253; responses to the ALK/ROS1/pan-TRK inhibitor entrectinib in two patients with treatment-refractory cancers harbouring either an ALK254 or an NTRK1 rearrangement255, as well as in two of three patients with NTRK-fusion positive CRCs from a pooled analysis of data from three tissue-agnostic phase I/II trials256; an exceptional 9-month response to the ALK inhibitor ceritinib in a patient with ALK fusion-positive CRC257; and a complete response to the broad-spectrum kinase inhibitor RXDX-105 in a patient with RET-rearranged CRC249. Additional clinical data are needed to support these anecdotal reports. In order to better identify eligible patients, understanding the likelihood of the co-occurrence of kinase fusions with other biomarkers might reveal a patient population with a frequency of such fusions that is appropriate for molecular screening258. For example, RET rearrangements can be found in two thirds of patients with right-sided MSI-H tumours that lack RAS or BRAF mutations249. Enrichment for NTRK fusions in MSI tumours has also been documented in clinically sequenced MSI-H metastatic CRC samples in a single-institution study, albeit with a prevalence of only 8% and 1% in MSI-H and MSS CRCs, respectively259.

Owing to associations with MSI, most patients with CRCs harbouring kinase fusions are also eligible for ICIs. This association provides an appealing possibility for combination therapies in patients with resistance to single-agent ICIs. Data supporting the use of TKIs in this setting are currently unavailable, although the combination of lorlatinib plus avelumab has shown clinical efficacy in patients with ALK-mutated non-small-cell lung cancers with no previous exposure to ICIs260. Of note, a durable response to single-agent nivolumab has been reported in one patient with MSI-H metastatic CRC harbouring an ALK rearrangement248. This observation suggests that this rearrangement is not predictive of innate resistance to ICIs. Again, the duration of response among patients with CRC who respond to ICIs is longer than that of patients who respond to TKIs: this will be an important consideration for therapy prioritization.

The R-spondins RSPO2/3 represent another class of potentially targetable oncogenic rearrangements in patients with CRC. These proteins are secreted proteins that regulate WNT signalling, and upon translocation to high-affinity promoter regions, can cause aberrant activation of WNT signalling in CRCs261. Virtually all CRCs of both the MSI and MSS subtypes have activating mutations in the WNT pathway, most commonly loss-of-function mutations in APC (in approximately 50% and 80% of MSI and MSS CRCs respectively)262. However, RSPO2/3 fusions are mutually exclusive to APC mutations, supporting an independent role of each alteration in the promotion of WNT signalling. These fusions are, in contrast to kinase fusions, primarily found in MSS CRCs and were initially reported to occur in 10% of MSS CRCs261. However, this initial report might be an overestimate and later studies suggest mutation frequencies of 0.35%88 and 4%263. RNF43 is another component in the same WNT regulatory complex264 and mutant forms of this tumour suppressor protein are also mutually exclusive to mutations in APC. Truncating alterations in RNF43 are most frequent in MSI-H CRCs and might occur in up to 80% of these tumours89,265. CRCs harbouring RSPO fusions or RNF43 mutations are, unlike tumours with many other types of WNT pathway alterations, dependent on secreted WNT ligands. This dependency offers an opportunity for targeted interventions, which is particularly relevant in light of the safety concerns associated with other therapeutic attempts to target this pathway266 and given that WNT signalling is essential for the homeostasis of nonmalignant adult tissues267,268. Indeed, preclinical data from an RNF43-mutated organoid model of CRC indicated sensitivity to PORCN inhibition, which blocks both the secretion and activity of WNTs23. Inhibition of PORCN signalling has been used to arrest tumour growth with on-target effects also reported in PTPRKRSPO3269 or RSPO2-fusion-positive270 PDX models, and the same effect was obtained with an anti-RSPO3 antibody in a PTPRKRSPO3 fusion positive PDX model271. A response to PORCN inhibition has been reported in one patient with an RNF43-mutated cancer who was treated in an ongoing phase I trial272; however, virtually no clinical data are currently available on the efficacy of RNF43 or RSPO-guided therapies in patients. Nonetheless, combination strategies based on translational and/or preclinical data are currently being investigated in early phase trials. These include the combination of agents that inhibit PORCN, BRAF and EGFR in patients with BRAFV600E-mutant metastatic CRCs harbouring either an RSPO fusion or an RNF43 mutation273. The combination of the porcupine inhibitor WNT974 with ICIs is also currently under investigation274, based on the association between activated WNT signalling and T cell exclusion observed in patients with CRC275.

Biomarkers of inferior responses

Several biomarkers are associated with a poor outcome in patients with CRC receiving standard-of-care therapies. In addition to the low-prevalence markers described previously, these include more common biomarkers that can be used to pinpoint particularly important target populations for the development of novel targeted therapies.

RAS mutations

Genetic testing for RAS mutations is recommended in clinical guidelines for the management of patients with metastatic CRC as a negative predictor of benefit from EGFR targeted therapies29,84. Associations between mutations in RAS and survival outcomes have also been intensively investigated in other therapeutic settings, although the prognostic value is less clear.

Hotspot mutations in KRAS or NRAS are mutually exclusive to mutations in BRAF and other recurrent mutations in the MAPK signalling pathway. KRAS mutations are the most frequent and occur in approximately 35% of stage I–IV primary CRCs (SUPPLEMENTARY TABLE 1), most commonly in right-sided tumours. Similar to BRAF, the prevalence of KRAS mutations decreases in a stepwise manner along the right-to-left axis of the large bowel. However, in contrast to BRAFV600E, KRAS mutations are twice as frequent in MSS compared with MSI-H colon cancers52,276. Furthermore, the potential size of the prognostic effect of these mutations is substantially smaller than that of BRAFV600E and remains debatable. However, the majority of studies of the prognostic implications of KRAS alterations revealed a negative effect on patient survival35,158,277-290 (SUPPLEMENTARY TABLE 1), including studies in which use of EGFR targeted therapies276,291 might have influenced the results.

Retrospective analyses of data from several randomized trials investigating the efficacy of adjuvant chemotherapies revealed no prognostic associations of KRAS mutations in patients with stage II or III CRCs68,292-294. However, a negative prognostic association was confirmed in an analysis of data from a pooled cohort of 7,326 patients with stage II and III colon cancers, which included some patients from the clinical trial cohorts52. Furthermore, the prognostic value of KRAS mutations in patients with primary CRCs might be limited to specific subgroups, including MSS cancers52,158,276,285,290, cancers with a distal primary tumour location295,296, or even to MSS cancers of the ‘epithelial-like’ CMS2/3 gene expression subtypes158. Accordingly, KRAS mutations have been suggested as a biomarker for the prognostic stratification of primary CRCs in trials in the adjuvant setting, in combination with both BRAF and MSI status29. Furthermore, KRAS mutations might be associated with inferior survival after relapse from stage III CRCs, although the result from this analysis was not statistically significant291. This association suggests that KRAS alterations have a stronger prognostic effect in patients with metastatic disease, and indeed, the majority of large-cohort studies have identified statistically significant negative prognostic associations (SUPPLEMENTARY TABLE 1)174,218,259,297-307, which possibly relates to the greater prevalence of MSS disease in this setting. Conflicting data on this association do exist308-312, although the prognostic effect of KRAS alterations is supported by a multivariable analysis of pooled data from 26 randomized trials, including 22,674 patients with metastatic CRC (HR for OS 1.35, 95% CI 1.30– 1.39; P <0.001)313.

NRAS mutations are less frequent and occur in only 3–4% of metastatic CRCs298,304,314. These mutations are, therefore, commonly grouped with KRAS alterations, with which they share some clinicopathological associations297,299,300,305,314. However, an independent association with a poor prognosis has also been demonstrated305, along with indications that mutant NRAS confers a somewhat worse prognosis than mutant KRAS298,314.

Importantly, RAS mutations seem to have prognostic value in patients who do not undergo metastasectomy, in patients undergoing partial liver resection300, and in patients with disease recurrence after partial liver resection297. An association with an inferior prognosis following hepatectomy has also been confirmed in a meta-analysis of data from 1,833 patients (HR for OS, 1.67, 95% CI 1.34–2.09; P <0.001)315. It has been suggested that surgical treatment might not be beneficial in some patients with RAS-mutated liver metastases 300; however, current guidelines for determining resectability do not consider the use of genetic testing for RAS or BRAF mutations84. Adding RAS mutations to clinical risk scores might help improve the risk:benefit ratio of therapeutic interventions in patients with resectable liver metastases, but prospective validation of such a personalized approach is needed316. In summary, although a substantial amount of evidence (SUPPLEMENTARY TABLE 1) supports a role for KRAS mutations in the prognostic stratification of patients with CRC, no appropriate clinical setting has been defined and the modest prognostic effect size limits clinical relevance.

The strongest associations between RAS mutations and outcome in patients with CRC relate to responsiveness to anti-EGFR therapies. Current expert consensus guidelines recommend extended pretreatment genetic testing for RAS alterations in patients with metastatic CRC, including KRAS and NRAS codons 12 and 13 (exon 2), 59 and 61 (exon 3), and 117 and 146 (exon 4)29. RAS mutations cause the aberrant activation of MAPK signalling downstream of EGFR, resulting in a poor response to EGFR inhibition218,317, and the recommendations for stratified treatment are based on large volumes of data from almost 75,000 patients. However, fewer than half of all patients with RAS wild type metastatic CRCs respond to anti-EGFR therapies218 and responsiveness might depend on the primary tumour location: patients with right-sided primary tumours derive no benefit from EGFR inhibition318. In future, the predictive biomarker panel for the detection of primary resistance might be further expanded to include ERBB2, KRAS and MET amplifications as well as mutations in the ectodomain of EGFR, MAP2K1, ERBB2, PIK3CA and possibly BRAF3. Indeed, a MAPK gene expression signature that enables tumours to be classified as either ‘wild-type like’ or ‘activated oncogenic’ has been shown to better predict treatment responsiveness than KRAS mutation status alone319. This example highlights the potential to improve the response rates of patients receiving targeted therapies using more comprehensive molecular screening approaches. Nevertheless, secondary resistance invariably develops, even among patients with responsive ‘multi-wild-type’ cancers, and the mechanisms largely converge on the same aberrations as in primary resistance. Again, tumour heterogeneity has a detrimental effect, and analyses of liquid biopsy samples have repeatedly demonstrated treatment-induced expansion of resistant subclones harbouring aberrations in the MAPK signalling pathway14,15,320-324. Importantly, rechallenge with anti-EGFR antibodies after therapy withdrawal might be possible owing to a decline in levels of the treatment-resistant subclone15,24. This phenomenon highlights the importance of molecular monitoring of patients during targeted therapy, both to identify mechanisms of resistance and to develop management strategies.

More than 40% of patients with metastatic CRC have RAS-mutant disease (SUPPLEMENTARY TABLE 1). This classification thus defines a large and heterogeneous patient population that routinely receives 5-FU-based chemotherapy with or without bevacizumab. A limited number of alternative options are available when treatment resistance develops, and RAS has for a long time been deemed undruggable325. Renewed optimism exists326 for example based on the in vitro effects of direct RAS inhibition by a selective KRAS-G12C inhibitor that locks the mutant protein in an inactive state327. However, KRAS-G12C constitutes only approximately 10% of KRAS mutations in CRC328 and data on clinical efficacy in patients are awaited. KRAS-targeted cellular immunotherapies might offer a new option, and tumor regressions have been demonstrated after infusion of autologous cytotoxic T cells with specific reactivity towards a KRAS-G12D neoantigen in a case report describing a patient with metastatic CRC329. Considering the genomic heterogeneity of RAS-mutant CRCs, different strategies are likely to be needed for different subsets of RAS-mutated cancers326. To this end, an international collaborative effort to subtype RAS-mutant metastatic CRCs according to their unique signalling dependencies is currently ongoing, and will hopefully provide a basis to design matched therapeutic interventions (https://www.colossusproject.eu/researchers/).

Combination strategies in CMS4 CRCs

The CMS4 gene expression subtype is found in approximately 25% of primary CRCs22, and the mesenchymal/stromal characteristics associated with this subtype have repeatedly been shown to confer a poor patient prognosis330-340. The poor prognostic value of CMS4 is independent of cancer stage and has been validated in an independent series of patients with stage I–IV CRCs341, as well as in a pooled analysis of tumours that partly overlaps with the original analysis cohort339. Adaptation of the CMS classification to the analysis of formalin-fixed paraffin-embedded tumour samples has further enabled the validation of the poor prognostic associations of CMS4 in almost 1,800 stage III colon cancer specimens from a randomized trial cohort342, and in a smaller multicentre series of patients with stage II cancers343. Bioinformatic modelling of intratumour heterogeneity in the former study suggests that both ‘pure’ CMS4 tumours and tumours in which CMS4 signals are heterogeneous and mixed with signals from any of the other subtypes confer a poor prognosis344.

The CMS classification of metastatic CRCs is complicated by several factors, including an expected enrichment with the poor-prognostic CMS4 group, dependence on the sample source used for gene expression profiling, and the effects of specific treatments prior to sample collection345. Analyses have largely been limited to profiling of the primary tumours, owing to effects of the tumour microenvironment on gene expression and the subsequent challenge of translating the classification to samples of metastatic tumours obtained from other organs. Accordingly, the extent of subtype heterogeneity in metastatic CRC is largely unknown, although a concordance in subtypes of 60% between primary tumours and their metastases have been reported (47% for the CMS4 subtype)346. The potential for CMS subtype-switching effects, as observed after neoadjuvant chemotherapy, increases the prevalence of a CMS4-like subtype in patients with pretreated metastatic tumours347. Furthermore, intratumour CMS heterogeneity in primary CRCs348, which is at least partly related to EMT in regions of tumour budding349, also suggests an increased prevalence of the CMS4 subtype in patients with metastatic disease. The poor prognostic value of CMS4 has been indicated relative to CMS2/3 in the metastatic setting, by a retrospective analysis of trial cohorts350. However, there is accumulating evidence that the CMS1 subtype is associated with inferior survival compared with CMS4 in patients with metastatic CRC346,351. This is likely related to the enrichment with MSI-H tumours in the CMS1 subtype. The treatments received by the patients might influence the prognostic analyses, and data on therapeutic outcomes associated with CMS4 are conflicting. CMS4-like subtypes have been suggested to be associated with limited benefit from standard-of-care therapies, including both 5-FU337 and oxaliplatin352 in the adjuvant setting in patients with stage II/III CRCs, as well as from EGFR targeted therapies in those with KRAS wild-type metastatic CRCs335,350. Associations with responsiveness to irinotecan-based chemotherapy334,353 and the broad-spectrum TKI regorafenib354 have also been reported in this disease subtype. However, chemotherapy resistance has been corroborated in preclinical models341,355 and overcoming treatment resistance in the CMS4 subtype is currently an important area of research. The potential to overcome resistance has been demonstrated using HSP90 inhibitors, which have synergistic antitumour effects when combined with 5-FU in PDX models of CMS4 CRC341.

Therapies designed to alter the tumour microenvironment of CMS4 CRCs are another area of considerable research interest. CMS4 cancers typically have robust activation of TGFβ-signalling, and inhibition of TGFβ signalling has been shown to inhibit crosstalk between cancer cells and cancer-associated fibroblasts, thus reducing the metastatic capacity of preclinical models338. CRCs of a mesenchymal phenotype are also immunosuppressive3,356, making the CMS4 group an attractive candidate for ‘immune-conversion’ strategies designed to render immunologically ‘cold’ tumours vulnerable to ICIs (FIG. 2). Several chemotherapies and molecularly targeted agents have been shown to temporarily promote antitumour immunity via various mechanisms140. Oxaliplatin has the potential to elicit immunogenic cell death357, and improvements in PFS observed with perioperative FOLFOX chemotherapy358 might be partly attributable to activation of a localized immune response97. The combination of FOLFOX with an anti-PD-1 antibody has a strong synergistic effect in mouse models of CRC359. However, these preclinical data have not been confirmed in patients, and a preliminary report indicates that the addition of atezolizumab to FOLFOX plus bevacizumab induction therapy does not improve the outcomes of patients with BRAF wild type metastatic CRCs360. Inhibition of TGFβ signalling also causes a cytotoxic T cell response and resensitizes PDX models of metastatic CRC to ICIs. This effect suggests that TGFβ-mediated activation of the tumour-associated stroma might be an important mechanism of immune evasion361. M7824, a bifunctional molecule simultaneously targeting PD-L1 and TGFβ confers long-term antitumour immunity and suppression of tumour growth362. Clinical benefit from this agent has been confirmed in one patient with MSS metastatic CRC of the CMS4 subtype in a phase I trial363. MEK inhibition also promotes the recruitment of cytotoxic T cells and synergizes with anti-PD-L1 antibodies in a mouse model of colon cancer364. Objective responses to this combination were observed in 10% of patients with metastatic MSS CRCs in a phase I trial365; however, the subsequent phase III study failed to demonstrate improvements in OS compared with regorafenib366. Finally, CMS4 tumours are characterized by a high proportion of myeloid-derived suppressor cells (MDSCs) in the tumour microenvironment367. MDSCs are known to prevent the activation of T cells during immune-checkpoint inhibition and might serve as negative predictors of treatment response in melanoma368,369. These cells can be targeted using epigenetic-modulating agents, which have synergistic effects when combined with ICIs in mouse models of moderately immunogenic colon cancers370.

In summary, the clinical translation of the CMS classification is currently premature, both regarding associations with therapeutic outcomes and the standardization of appropriate assays. However, the CMS classification does provide an improved biological taxonomy of CRCs and a new framework for patient stratification in biologically guided clinical trials.

Conclusions

Biomarker-guided treatment options for patients with primary CRCs remain limited. Upstaging of patients with high-risk primary CRCs to enable them to receive experimental therapies currently reserved for those with metastatic disease might offer the potential for cure, although this is currently not a well-explored approach. In this respect, data from the phase III trial in which patients with dMMR stage III colon cancers are receiving ICIs in the adjuvant setting are eagerly awaited121. In the metastatic setting, the accumulation of experimental data is broadening the applicability of established biomarkers. Associations with a poor prognosis in the context of standard-of-care therapies, albeit with the potential for improved outcomes by targeting the marker itself seem to be a common theme. Most established biomarkers have a low prevalence, although the number of biomarkers is increasing and CRC might, in this respect, eventually be considered an umbrella diagnosis encompassing numerous rare disease subtypes (FIG. 3). Beneath this umbrella, a growing level of biomarker complexity is emerging, primarily caused by interactions between different biomarkers. These interactions reinforce the importance of expanded genetic testing to enable improved treatment-related decision making. A rapid increase in the level of biomarker complexity is also expected to emerge from resources such as large-scale preclinical drug screens and comprehensive molecular profiles generated in translational studies. In this setting, artificial intelligence offers a new and intriguing opportunity to develop improved molecular prediction algorithms from the wealth of available data, and to develop synergistic drug combinations371.

Figure 3. Treatment options and biomarker interactions in metastatic CRCs.

Figure 3.

EGFR targeted therapies, guided by RAS mutation status remain the foundation of biomarker stratified medicine in patients with metastatic CRC, although treatment options are expanding, guided by several low-prevalence biomarkers and biomarker combinations. CRCs of the CMS4 subtype and/or those harbouring RAS mutations are important target populations for the development of new treatment strategies. BRAFi, BRAF inhibition; CMS, consensus molecular subtypes; MEKi, MEK inhibition; MSI, microsatellite instability; MSS, microsatellite stability; RAS, KRAS/NRAS.

Supplementary Material

Supplemental table

Key points.

  • The expanded use of biomarkers to guide the treatment of patients with colorectal cancer (CRC) has revealed a level of complexity arising from interactions between different biomarkers.

  • An improved understanding of the causes of primary resistance might improve response rates among patients receiving targeted therapies and enable more-effective drug combinations, exemplified by mutations in the MAPK signalling pathway for EGFR and/or BRAF targeted therapies.

  • Immune-checkpoint inhibition (ICI) has provided the largest contribution to the increased use of molecularly guided therapies, and biomarkers that complement patient stratification by MSI status are likely to provide further benefit.

  • Biomarkers that indicate a poor prognosis have motivated the search for more effective therapies for specific molecular subgroups; these biomarkers typically have a limited prevalence, but their accumulation could expand the eligibility for, and benefit from, targeted treatment.

  • Some CRCs harbour more than one molecular target and treatment sequencing both in relation to standard and targeted therapies is a growing challenge.

Acknowledgements

This work was supported by grants from The Norwegian Cancer Society (project numbers 6824048-2016 and 182759-2016), The Research Council of Norway (project number 250993), South-Eastern Norway Regional Health Authority (project numbers 2016123 and 2017102) and the NIH (project numbers R01 CA218230, R01 CA184843 and R01 CA187238).

Footnotes

Competing interests

A.S. and R.A.L. are co-inventors of a pending patent application (Attorney Docket Number: INVEN-35063/US-1/PRO) regarding the use of HSP90 inhibitors in relation to the consensus molecular subtypes of colorectal cancer. S.K. is a co-inventor of a pending patent application regarding a clinical classifier for the consensus molecular subtypes of colorectal cancer.

Additional information

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Linnekamp JF, Wang X, Medema JP & Vermeulen L Colorectal cancer heterogeneity and targeted therapy: a case for molecular disease subtypes. Cancer Res. 75, 245–249 (2015). [DOI] [PubMed] [Google Scholar]
  • 2.Dienstmann R, Salazar R & Tabernero J Personalizing colon cancer adjuvant therapy: selecting optimal treatments for individual patients. J. Clin. Oncol 33, 1787–1796 (2015). [DOI] [PubMed] [Google Scholar]
  • 3.Dienstmann R et al. Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat. Rev. Cancer 17, 79–92 (2017). [DOI] [PubMed] [Google Scholar]
  • 4.Schmoll HJ et al. ESMO consensus guidelines for management of patients with colon and rectal cancer. A personalized approach to clinical decision making. Ann. Oncol 23, 2479–2516 (2012). [DOI] [PubMed] [Google Scholar]
  • 5.Grothey A et al. Duration of adjuvant chemotherapy for stage III colon cancer. N. Engl. J. Med 378, 1177–1188 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Tabernero J et al. Ramucirumab versus placebo in combination with second-line FOLFIRI in patients with metastatic colorectal carcinoma that progressed during or after first-line therapy with bevacizumab, oxaliplatin, and a fluoropyrimidine (RAISE): a randomised, double-blind, multicentre, phase 3 study. Lancet Oncol. 16, 499–508 (2015). [DOI] [PubMed] [Google Scholar]
  • 7.Grothey A et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 381, 303–312 (2013). [DOI] [PubMed] [Google Scholar]
  • 8.Cremolini C et al. First-line chemotherapy for mCRC-a review and evidence-based algorithm. Nat. Rev. Clin. Oncol 12, 607–619 (2015). [DOI] [PubMed] [Google Scholar]
  • 9.Tabernero J et al. Analysis of angiogenesis biomarkers for ramucirumab efficacy in patients with metastatic colorectal cancer from RAISE, a global, randomized, double-blind, phase III study. Ann. Oncol 29, 602–609 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Toledo RA et al. Exome sequencing of plasma DNA portrays the mutation landscape of colorectal cancer and discovers mutated VEGFR2 receptors as modulators of anti-angiogenic therapies. Clin. Cancer Res 24, 3550–3559 (2018). [DOI] [PubMed] [Google Scholar]
  • 11.Van Cutsem E et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann. Oncol 27, 1386–1422 (2016). [DOI] [PubMed] [Google Scholar]
  • 12.Van Cutsem E et al. Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J. Clin. Oncol 33, 692–700 (2015). [DOI] [PubMed] [Google Scholar]
  • 13.Hammond WA, Swaika A & Mody K Pharmacologic resistance in colorectal cancer: a review. Ther. Adv. Med. Oncol 8, 57–84 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Misale S et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 486, 532–536 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Siravegna G et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat. Med 21, 795–801 (2015).Tracking of the evolution of resistance-bearing subclones in liquid biopsy samples during treatment with anti-EGFR antibodies provide a molecular explanation for the efficacy of therapy rechallenge.
  • 16.Le DT et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med 372, 2509–2520 (2015).Prospective trial showing that MMR status predicts clinical benefit from the ICI pembrolizumab in patients with treatment-refractory metastatic cancers.
  • 17.Diaz LA et al. Pembrolizumab therapy for microsatellite instability high (MSI-H) colorectal cancer (CRC) and non-CRC. J. Clin. Oncol 35, abstr. 3071 (2017). [Google Scholar]
  • 18.Overman MJ et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol. 18, 1182–1191 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gong J, Wang C, Lee PP, Chu P & Fakih M Response to PD-1 blockade in microsatellite stable metastastic colorectal cancer harboring a POLE mutation. J. Natl. Compr. Canc. Netw 15, 142–147 (2017). [DOI] [PubMed] [Google Scholar]
  • 20.Hong DS et al. Phase IB study of vemurafenib in combination with irinotecan and cetuximab in patients with metastatic colorectal cancer with BRAFV600E mutation. Cancer Discov. 6, 1352–1365 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kopetz S et al. Randomized trial of irinotecan and cetuximab with or without vemurafenib in BRAF-mutant metastatic colorectal cancer (SWOG 1406). J. Clin. Oncol 35, abstr. 520 (2017).Meeting abstract presenting initial data from a prospective randomized trial indicating prolonged PFS in patients with BRAFV600E mutated and RAS wild-type metastatic CRCs by addition of vemurafenib to the combination of irinotecan plus cetuximab.
  • 22.Guinney J et al. The consensus molecular subtypes of colorectal cancer. Nat. Med 21, 1350–1356 (2015).The international CRC subtyping consortium combined several gene expression-based classification frameworks for CRC into the four consensus molecular subtypes, based on analysis of almost 4,000 primary tumours.
  • 23.van de Wetering M et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 161, 933–945 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Siena S et al. Rechallenge with EGFR inhibitors in patients with metastatic colorectal cancer: effect on outcomes. Ann. Oncol 28, abstr. P-320 (2017). [Google Scholar]
  • 25.Marquart J, Chen EY & Prasad V Estimation of the percentage of US patients with cancer who benefit from genome-driven oncology. JAMA Oncol. 4, 1093–1098 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ballman KV Biomarker: Predictive or Prognostic? J. Clin. Oncol 33, 3968–3971 (2015). [DOI] [PubMed] [Google Scholar]
  • 27.Labianca R et al. Early colon cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol 24, vi64–vi72 (2013). [DOI] [PubMed] [Google Scholar]
  • 28.Duffy MJ et al. Tumor markers in colorectal cancer, gastric cancer and gastrointestinal stromal cancers: European group on tumor markers (EGTM) 2013 guidelines update. Int. J. Cancer 134, 2513–2522 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sepulveda AR et al. Molecular biomarkers for the evaluation of colorectal cancer: guideline from the American Society for Clinical Pathology, College of American Pathologists, Association for Molecular Pathology, and the American Society of Clinical Oncology. J. Clin. Oncol 35, 1453–1486 (2017). [DOI] [PubMed] [Google Scholar]
  • 30.Thibodeau SN, Bren G & Schaid D Microsatellite instability in cancer of the proximal colon. Science 260, 816–819 (1993). [DOI] [PubMed] [Google Scholar]
  • 31.Lothe RA et al. Genomic instability in colorectal cancer: Relationship to clinicopathological variables and family history. Cancer Res. 53, 5849–5852 (1993). [PubMed] [Google Scholar]
  • 32.Popat S, Hubner R & Houlston RS Systematic review of microsatellite instability and colorectal cancer prognosis. J. Clin. Oncol 23, 609–618 (2005). [DOI] [PubMed] [Google Scholar]
  • 33.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 46, 2788–2798 (2010). [DOI] [PubMed] [Google Scholar]
  • 34.Sargent DJ et al. Defective mismatch repair as a predictive marker for lack of efficacy of fluorouracil-based adjuvant therapy in colon cancer. J. Clin. Oncol 28, 3219–3226 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hutchins G et al. Value of mismatch repair, KRAS, and BRAF mutations in predicting recurrence and benefits from chemotherapy in colorectal cancer. J. Clin. Oncol 29, 1261–1270 (2011). [DOI] [PubMed] [Google Scholar]
  • 36.Myeroff LL et al. A transforming growth factor beta receptor type II gene mutation common in colon and gastric but rare in endometrial cancers with microsatellite instability. Cancer Res. 55, 5545–5547 (1995). [PubMed] [Google Scholar]
  • 37.Rampino N et al. Somatic frameshift mutations in the BAX gene in colon cancers of the microsatellite mutator phenotype. Science 275, 967–969 (1997). [DOI] [PubMed] [Google Scholar]
  • 38.Souza RF et al. Microsatellite instability in the insulin-like growth factor II receptor gene in gastrointestinal tumours. Nat. Genet 14, 255–257 (1996). [DOI] [PubMed] [Google Scholar]
  • 39.Thorstensen L et al. WNT1 inducible signaling pathway protein 3, WISP-3, a novel target gene in colorectal carcinomas with microsatellite instability. Gastroenterology 121, 1275–1280 (2001). [DOI] [PubMed] [Google Scholar]
  • 40.Røyrvik EC, Ahlquist T, Rognes T & Lothe RA Slip slidin' away: a duodecennial review of targeted genes in mismatch repair deficient colorectal cancer. Crit. Rev. Oncog 13, 229–257 (2007). [DOI] [PubMed] [Google Scholar]
  • 41.Dolcetti R et al. High prevalence of activated intraepithelial cytotoxic T lymphocytes and increased neoplastic cell apoptosis in colorectal carcinomas with microsatellite instability. Am. J. Pathol 154, 1805–1813 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jass JR et al. Morphology of sporadic colorectal cancer with DNA replication errors. Gut 42, 673–679 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Galon J et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 313, 1960–1964 (2006). [DOI] [PubMed] [Google Scholar]
  • 44.Pages F et al. In situ cytotoxic and memory T cells predict outcome in patients with early-stage colorectal cancer. J. Clin. Oncol 27, 5944–5951 (2009). [DOI] [PubMed] [Google Scholar]
  • 45.Pages F et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 391, 2128–2139 (2018).Retrospective international multi-center study showing that the immunoscore has prognostic value beyond that of clinicopathological prognostic factors and MSI status in patients with stage I–III colon cancers.
  • 46.Grady WM & Carethers JM Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology 135, 1079–1099 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ward R et al. Microsatellite instability and the clinicopathological features of sporadic colorectal cancer. Gut 48, 821–829 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gryfe R et al. Tumor microsatellite instability and clinical outcome in young patients with colorectal cancer. N. Engl. J. Med 342, 69–77 (2000). [DOI] [PubMed] [Google Scholar]
  • 49.Samowitz WS et al. Microsatellite instability in sporadic colon cancer is associated with an improved prognosis at the population level. Cancer Epidemiol. Biomarkers Prev 10, 917–923 (2001). [PubMed] [Google Scholar]
  • 50.Roth AD et al. Integrated analysis of molecular and clinical prognostic factors in stage II/III colon cancer. J. Natl. Cancer Inst 104, 1635–1646 (2012). [DOI] [PubMed] [Google Scholar]
  • 51.Vilar E & Gruber SB Microsatellite instability in colorectal cancer-the stable evidence. Nat. Rev. Clin. Oncol 7, 162 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Dienstmann R et al. Prediction of overall survival in stage II and III colon cancer beyond TNM system: a retrospective, pooled biomarker study. Ann. Oncol 28, 1023–1031 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Breivik J et al. Different genetic pathways to proximal and distal colorectal cancer influenced by sex-related factors. Int. J. Cancer 74, 664–669 (1997). [DOI] [PubMed] [Google Scholar]
  • 54.Sinicrope FA et al. Prognostic impact of deficient DNA mismatch repair in patients with stage III colon cancer from a randomized trial of FOLFOX-based adjuvant chemotherapy. J. Clin. Oncol 31, 3664–3672 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Merok MA et al. Microsatellite instability has a positive prognostic impact on stage II colorectal cancer after complete resection: Results from a large, consecutive Norwegian series. Ann. Oncol 24, 1274–1282 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Klingbiel D et al. Prognosis of stage II and III colon cancer treated with adjuvant 5-fluorouracil or FOLFIRI in relation to microsatellite status: results of the PETACC-3 trial. Ann. Oncol 26, 126–132 (2015). [DOI] [PubMed] [Google Scholar]
  • 57.Sinicrope FA 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 103, 863–875 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Benatti P et al. Microsatellite instability and colorectal cancer prognosis. Clin. Cancer Res 11, 8332–8340 (2005). [DOI] [PubMed] [Google Scholar]
  • 59.Jo WS & Carethers JM Chemotherapeutic implications in microsatellite unstable colorectal cancer. Cancer Biomark. 2, 51–60 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Carethers JM et al. Use of 5-fluorouracil and survival in patients with microsatellite-unstable colorectal cancer. Gastroenterology 126, 394–401 (2004). [DOI] [PubMed] [Google Scholar]
  • 61.Lanza G et al. Immunohistochemical test for MLH1 and MSH2 expression predicts clinical outcome in stage II and III colorectal cancer patients. J. Clin. Oncol 24, 2359–2367 (2006). [DOI] [PubMed] [Google Scholar]
  • 62.Jover R et al. The efficacy of adjuvant chemotherapy with 5-fluorouracil in colorectal cancer depends on the mismatch repair status. Eur. J. Cancer 45, 365–373 (2009). [DOI] [PubMed] [Google Scholar]
  • 63.Ribic CM et al. Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N. Engl. J. Med 349, 247–257 (2003).Retrospective analysis of pooled data from randomized trials with 5-FU-based adjuvant chemotherapies in patients with stage II or III colon cancer indicate a significant difference in the benefit of treatment in patients with MSI-H and MSS tumours.
  • 64.Andre T et al. Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N. Engl. J. Med 350, 2343–2351 (2004). [DOI] [PubMed] [Google Scholar]
  • 65.Flejou JF et al. Effect of adding oxaliplatin to adjuvant 5-fluorouracil/leucovorin (5FU/LV) in patients with defective mismatch repair (dMMR) colon cancer stage II and III included in the MOSIAC study. J. Clin. Oncol 31, abstract no. 3524 (2013). [Google Scholar]
  • 66.Tougeron D et al. Efficacy of adjuvant chemotherapy in colon cancer with microsatellite instability: a large multicenter AGEO study. J. Natl. Cancer Inst 108, djv438 (2016). [DOI] [PubMed] [Google Scholar]
  • 67.Zaanan A 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 17, 7470–7478 (2011). [DOI] [PubMed] [Google Scholar]
  • 68.Gavin PG et al. Mutation profiling and microsatellite instability in stage II and III colon cancer: an assessment of their prognostic and oxaliplatin predictive value. Clin. Cancer Res 18, 6531–6541 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Sinicrope F et al. 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, 3525–3525 (2014). [Google Scholar]
  • 70.Saltz LB et al. Irinotecan fluorouracil plus leucovorin is not superior to fluorouracil plus leucovorin alone as adjuvant treatment for stage III colon cancer: results of CALGB 89803. J. Clin. Oncol 25, 3456–3461 (2007). [DOI] [PubMed] [Google Scholar]
  • 71.Ychou M et al. A phase III randomised trial of LV5FU2 + irinotecan versus LV5FU2 alone in adjuvant high-risk colon cancer (FNCLCC Accord02/FFCD9802). Ann. Oncol 20, 674–680 (2009). [DOI] [PubMed] [Google Scholar]
  • 72.Van Cutsem E et al. Randomized phase III trial comparing biweekly infusional fluorouracil/leucovorin alone or with irinotecan in the adjuvant treatment of stage III colon cancer: PETACC-3. J. Clin. Oncol 27, 3117–3125 (2009). [DOI] [PubMed] [Google Scholar]
  • 73.Bertagnolli MM et al. Microsatellite instability predicts improved response to adjuvant therapy with irinotecan, fluorouracil, and leucovorin in stage III colon cancer: Cancer and Leukemia Group B Protocol 89803. J. Clin. Oncol 27, 1814–1821 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Schutte M et al. Molecular dissection of colorectal cancer in pre-clinical models identifies biomarkers predicting sensitivity to EGFR inhibitors. Nat. Commun 8, 14262 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Campbell BB et al. Comprehensive analysis of hypermutation in human cancer. Cell 171, 1042–1056.e1010 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Angelova M et al. Characterization of the immunophenotypes and antigenomes of colorectal cancers reveals distinct tumor escape mechanisms and novel targets for immunotherapy. Genome Biol. 16, 64 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Willis J et al. Impact of microsatellite instability (MSI) on tumor clonal evolution in metastatic colorectal cancer (mCRC). J. Clin. Oncol 36, abstr. 616 (2018). [Google Scholar]
  • 78.Peltomaki P & Vasen H Mutations associated with HNPCC predisposition -- Update of ICG-HNPCC/INSiGHT mutation database. Dis. Markers 20, 269–276 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Weisenberger DJ et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat. Genet 38, 787–793 (2006). [DOI] [PubMed] [Google Scholar]
  • 80.Cohen R et al. Clinical and molecular characterisation of hereditary and sporadic metastatic colorectal cancers harbouring microsatellite instability/DNA mismatch repair deficiency. Eur. J. Cancer 86, 266–274 (2017). [DOI] [PubMed] [Google Scholar]
  • 81.Haraldsdottir S et al. Patients with colorectal cancer associated with Lynch syndrome and MLH1 promoter hypermethylation have similar prognoses. Genet. Med 18, 863–868 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Sinicrope FA et al. Molecular markers identify subtypes of stage III colon cancer associated with patient outcomes. Gastroenterology 148, 88–99 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sato K et al. Fusion kinases identified by genomic analyses of sporadic microsatellite instability-high colorectal cancers. Clin. Cancer Res 25, 378–389 (2019). [DOI] [PubMed] [Google Scholar]
  • 84.Benson AB et al. NCCN Clinical Practice Guidelines in Oncology: Colon Cancer. Version 4.2018. National Comprehensive Cancer Network; https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf (2018). [DOI] [PubMed] [Google Scholar]
  • 85.Dalerba P et al. CDX2 as a prognostic biomarker in stage II and stage III colon cancer. N. Engl. J. Med 374, 211–222 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Bruun J et al. Prognostic, predictive and pharmacogenomic assessments of CDX2 refine stratification of colorectal cancer. Mol. Oncol 12, 1639–1655 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wang J et al. Prevalence of recurrent oncogenic fusion in mismatch repair-deficient colorectal carcinoma with hypermethylated MLH1 and wild-type BRAF and KRAS. Mod. Pathol, doi: 10.1038/s41379-41019-40212-41371 (2019). [DOI] [PubMed] [Google Scholar]
  • 88.Kloosterman WP et al. A systematic analysis of oncogenic gene fusions in primary colon cancer. Cancer Res. 77, 3814–3822 (2017). [DOI] [PubMed] [Google Scholar]
  • 89.Sveen A et al. Multilevel genomics of colorectal cancers with microsatellite instability – clinical impact of JAK1 mutations and consensus molecular subtype 1. Genome Med. 9, 46 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Mlecnik B et al. Integrative analyses of colorectal cancer show immunoscore is a stronger predictor of patient survival than microsatellite instability. Immunity 44, 698–711 (2016). [DOI] [PubMed] [Google Scholar]
  • 91.Galon J et al. Cancer classification using the Immunoscore: a worldwide task force. J. Transl. Med 10, 205 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Venderbosch S et al. 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 (2014).Retrospective analyses of pooled data from randomized clinical trials showing that dMMR cancers are associated with inferior PFS and OS compared with MMR proficient cancers in patients with metastatic CRC, and that this is likely driven by enrichment with BRAFV600E mutations.
  • 93.Koopman M et al. Deficient mismatch repair system in patients with sporadic advanced colorectal cancer. Br. J. Cancer 100, 266–273 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Smith CG et al. Somatic profiling of the epidermal growth factor receptor pathway in tumors from patients with advanced colorectal cancer treated with chemotherapy +/− cetuximab. Clin. Cancer Res 19, 4104–4113 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Halama N et al. Localization and density of immune cells in the invasive margin of human colorectal cancer liver metastases are prognostic for response to chemotherapy. Cancer Res. 71, 5670–5677 (2011). [DOI] [PubMed] [Google Scholar]
  • 96.Mlecnik B et al. Comprehensive intrametastatic immune quantification and major impact of immunoscore on survival. J. Natl. Cancer Inst 110, djx123 (2018). [DOI] [PubMed] [Google Scholar]
  • 97.Tanis E et al. Prognostic impact of immune response in resectable colorectal liver metastases treated by surgery alone or surgery with perioperative FOLFOX in the randomised EORTC study 40983. Eur. J. Cancer 51, 2708–2717 (2015). [DOI] [PubMed] [Google Scholar]
  • 98.Pugh SA, Harrison RJ, Primrose JN & Khakoo SI T cells but not NK cells are associated with a favourable outcome for resected colorectal liver metastases. BMC Cancer 14, 180 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Halama N et al. The localization and density of immune cells in primary tumors of human metastatic colorectal cancer shows an association with response to chemotherapy. Cancer Immun. 9, 1 (2009). [PMC free article] [PubMed] [Google Scholar]
  • 100.Halama N et al. Hepatic metastases of colorectal cancer are rather homogeneous but differ from primary lesions in terms of immune cell infiltration. Oncoimmunology 2, e24116 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Tran B et al. Impact of BRAF mutation and microsatellite instability on the pattern of metastatic spread and prognosis in metastatic colorectal cancer. Cancer 117, 4623–4632 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Kim CG et al. Effects of microsatellite instability on recurrence patterns and outcomes in colorectal cancers. Br. J. Cancer 115, 25–33 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Heinemann V et al. Somatic DNA mutations, tumor mutational burden (TMB), and MSI status: association with efficacy in patients (pts) with metastatic colorectal cancer (mCRC) of FIRE-3 (AIO KRK-0306). J. Clin. Oncol 36, abstr. 3591 (2018). [Google Scholar]
  • 104.Margonis GA et al. Microsatellite instability in resectable colorectal liver metastasis: an international multi-institutional analysis. J. Clin. Oncol 36, abstr. 220 (2018). [Google Scholar]
  • 105.Price TJ et al. Outcomes for metastatic colorectal cancer (mCRC) based on microsatellite instability. J. Clin. Oncol 36, abstr. 759 (2018). [Google Scholar]
  • 106.Jin Z et al. Outcome of mismatch repair-deficient metastatic colorectal cancer: the Mayo Clinic experience. Oncologist 23, 1083–1091 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Sjo OH et al. Peritoneal carcinomatosis of colon cancer origin: highest incidence in women and in patients with right-sided tumors. J. Surg. Oncol 104, 792–797 (2011). [DOI] [PubMed] [Google Scholar]
  • 108.Sorbye H et al. High BRAF mutation frequency and marked survival differences in subgroups according to KRAS/BRAF mutation atatus and tumor tissue availability in a prospective population-based metastatic colorectal cancer cohort. PLoS One 10, e0131046 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Goldstein J et al. Multicenter retrospective analysis of metastatic colorectal cancer (CRC) with high-level microsatellite instability (MSI-H). Ann. Oncol 25, 1032–1038 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Venook AP et al. Impact 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. Oncol 34, 3504–3504 (2016). [Google Scholar]
  • 111.Holch JW, Ricard I, Stintzing S, Modest DP & Heinemann V The relevance of primary tumoor location in patients with metastatic colorectal cancer: A meta-analysis of first-line clinical trials. Eur. J. Cancer 70, 87–98 (2017). [DOI] [PubMed] [Google Scholar]
  • 112.Loupakis F et al. Primary tumor location as a prognostic factor in metastatic colorectal cancer. J. Natl. Cancer Inst 107, dju427 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Cremolini C et al. Primary tumor sidedness and benefit from FOLFOXIRI plus bevacizumab as initial therapy for metastatic colorectal cancer. Retrospective analysis of the TRIBE trial by GONO. Ann. Oncol 29, 1528–1534 (2018). [DOI] [PubMed] [Google Scholar]
  • 114.Loree J et al. Classifying colorectal cancer by tumor location rather than sidedness highlights a continuum in mutation profiles and Consensus Molecular Subtypes. Clin. Cancer Res 24, 1062–1072 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Steinert G et al. Immune escape and survival mechanisms in circulating tumor cells of colorectal cancer. Cancer Res. 74, 1694–11704 (2014). [DOI] [PubMed] [Google Scholar]
  • 116.US Food and Drug Administration. FDA approves first cancer treatment for any solid tumor with a specific genetic feature, https://www.fda.gov/newsevents/newsroom/pressannouncements/ucm560167.htm (2017).
  • 117.Le DT et al. Mismatch-repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409–413 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.US National Library of Medicine. ClinicalTrials.gov, https://clinicaltrials.gov/ct2/show/NCT02563002 (2015). [DOI] [PubMed]
  • 119.Diaz LA et al. Phase 3, open-label, randomized study of first-line pembrolizumab (pembro) vs investigator-choice chemotherapy for mismatch repair-deficient (dMMR) or microsatellite instability-high (MSI-H) metastatic colorectal carcinoma (mCRC): KEYNOTE-177. J. Clin. Oncol 35, abstr. TPS3618 (2017). [Google Scholar]
  • 120.US National Library of Medicine. ClinicalTrials.gov, https://clinicaltrials.gov/ct2/show/NCT02997228 (2016). [DOI] [PubMed]
  • 121.US National Library of Medicine. ClinicalTrials.gov, https://clinicaltrials.gov/ct2/show/NCT02912559 (2016). [DOI] [PubMed]
  • 122.Overman MJ et al. Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J. Clin. Oncol 36, 773–779 (2018).Combination immune checkpoint inhibition with nivolumab plus ipilimumab achieved a high response rate in patients with MSI-H/dMMR metastatic CRCs (also in those with BRAFV600E mutations), and an indirect comparison of different trial cohorts suggest that this combination might improve the PFS and OS compared with single-agent ICI.
  • 123.Lenz HJJ et al. Durable clinical benefit with nivolumab (NIVO) plus low-dose ipilimumab (IPI) as first-line therapy in microsatellite instability-high/mismatch repair deficient (MSI-H/dMMR) metastatic colorectal cancer (mCRC). Ann. Oncol 29, abstract LBA18_PR (2018). [Google Scholar]
  • 124.Chalabi M et al. Neoadjuvant ipilimumab plus nivolumab in early stage colon cancer. Ann. Oncol 29, abstr. LBA37_PR (2018). [Google Scholar]
  • 125.Chen EX et al. The CCTG CO.26 trial: A phase II randomized study of durvalumab plustremelimumab and best supportive care (BSC) vs BSC alone in patients with advanced colorectal carcinoma (CRC) refractory to standard therapies. J. Clin. Oncol 35, abstr. TPS3621 (2017). [Google Scholar]
  • 126.US National Library of Medicine. ClinicalTrials.gov, https://clinicaltrials.gov/ct2/show/NCT02870920 (2016). [DOI] [PubMed]
  • 127.Giannakis M et al. Genomic correlates of immune-cell infiltrates in colorectal carcinoma. Cell Rep. 15, 857–865 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.van Rooij N et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J. Clin. Oncol 31, e439–442 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Pardoll DM The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12, 252–264 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Llosa NJ et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 5, 43–51 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Gubin MM et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515, 577–581 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Domingo E et al. Somatic POLE proofreading domain mutation, immune response, and prognosis in colorectal cancer: a retrospective, pooled biomarker study. Lancet Gastroenterol. Hepatol 1, 207–216 (2016). [DOI] [PubMed] [Google Scholar]
  • 133.Fabrizio DA et al. Beyond microsatellite testing: assessment of tumor mutational burden identifies subsets of colorectal cancer who may respond to immune checkpoint inhibition. J. Gastrointest. Oncol 9, 610–617 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.US National Library of Medicine. ClinicalTrials.gov, https://clinicaltrials.gov/ct2/show/NCT03150706 (2017). [DOI] [PubMed]
  • 135.Guerra J et al. POLE somatic mutations in advanced colorectal cancer. Cancer Med. 6, 2966–2971 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Chan TA et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann. Oncol 30, 44–56 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Goodman AM et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol. Cancer Ther 16, 2598–2608 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Kirilovsky A et al. Rational bases for the use of the Immunoscore in routine clinical settings as a prognostic and predictive biomarker in cancer patients. Int. Immunol 28, 373–382 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Galon J et al. MSI status plus immunoscore to select metastatic colorectal cancer patients for immunotherapies. Ann. Oncol 29, abstr. 12P (2018). [Google Scholar]
  • 140.Sharma P, Hu-Lieskovan S, Wargo J & Ribas A Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Shin DS et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov. 7, 188–201 (2017).Homozygous loss-of-function mutation in JAK1 identified as a likely mechanism of primary resistance to immune checkpoint inhibition in a metastatic colon cancer with a high TMB and no response to pembrolizumab.
  • 142.Hugo W et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 165, 35–44 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Zhou G et al. Blockade of LAG3 enhances responses of tumor-infiltrating T cells in mismatch repair-proficient liver metastases of colorectal cancer. Oncoimmunology 7, e1448332 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Zaretsky JM et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med 375, 819–829 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Skoulidis F et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov. 8, 822–835 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Chen PL et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 6, 827–837 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Hamilton SR BRAF mutation and microsatellite instability status in colonic and rectal carcinoma: context really does matter. J. Natl. Cancer Inst 105, 1075–1077 (2013). [DOI] [PubMed] [Google Scholar]
  • 148.Rajagopalan H et al. Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature 418, 934 (2002). [DOI] [PubMed] [Google Scholar]
  • 149.Lochhead P et al. Microsatellite instability and BRAF mutation testing in colorectal cancer prognostication. J. Natl. Cancer Inst 105, 1151–1156 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Samowitz WS et al. Poor survival associated with the BRAF V600E mutation in microsatellite-stable colon cancers. Cancer Res. 65, 6063–6069 (2005). [DOI] [PubMed] [Google Scholar]
  • 151.Gonsalves WI et al. Patient and tumor characteristics and BRAF and KRAS mutations in colon cancer, NCCTG/Alliance N0147. J. Natl. Cancer Inst 106, dju106 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Clarke CN & Kopetz ES BRAF mutant colorectal cancer as a distinct subset of colorectal cancer: clinical characteristics, clinical behaviour, and response to targeted therapies. J. Gastrointest. Oncol 6, 660–667 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Vedeld HM et al. CpG island methylator phenotype identifies high risk patients among microsatellite stable BRAF mutated colorectal cancers. Int. J. Cancer 141, 967–976 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Tie J et al. Optimizing targeted therapeutic development: analysis of a colorectal cancer population with the BRAFV600E mutation. Int. J. Cancer 128, 2075–2084 (2011). [DOI] [PubMed] [Google Scholar]
  • 155.Roth AD 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 28, 466–474 (2010). [DOI] [PubMed] [Google Scholar]
  • 156.Taieb J et al. Prognostic effect of BRAF and KRAS mutation in patients with stage III colon cancer treated with leucovorin, fluorouracil, and oxaliplatin with or without cetuximab: a post hoc analysis of the PETACC-8 trial. JAMA Oncol. 2, 643–653 (2016).Retrospective analyses of MSI-status, BRAFV600E and KRAS mutations in patients with stage III colon cancer included in the PETACC-8 clinical trial showed that mutations in both genes were independently associated with inferior outcomes in MSS cancers, suggesting that these markers need to be used for patient stratification in future trials involving adjuvant treatments.
  • 157.Seppala TT et al. Combination of microsatellite instability and BRAF mutation status for subtyping colorectal cancer. Br. J. Cancer 112, 1966–1975 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Smeby J et al. CMS-dependent prognostic impact of KRAS and BRAFV600E mutations in primary colorectal cancer. Ann. Oncol 29, 1227–1234 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Vogelstein B et al. Genetic alterations during colorectal-tumor development. N. Engl. J. Med 319, 525–532 (1988). [DOI] [PubMed] [Google Scholar]
  • 160.Fearon ER & Vogelstein B A genetic model for colorectal tumorigenesis. Cell 61, 759–767 (1990). [DOI] [PubMed] [Google Scholar]
  • 161.Fearon ER Molecular genetics of colorectal cancer. Annu.Rev.Pathol 6:479–507., 479-507 (2011). [DOI] [PubMed] [Google Scholar]
  • 162.Kambara T et al. BRAF mutation is associated with DNA methylation in serrated polyps and cancers of the colorectum. Gut 53, 1137–1144 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Rad R et al. A genetic progression model of BRAF(V600E)-induced intestinal tumorigenesis reveals targets for therapeutic intervention. Cancer Cell 24, 15–29 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Seligmann JF et al. Investigating the poor outcomes of BRAF-mutant advanced colorectal cancer: analysis from 2530 patients in randomised clinical trials. Ann. Oncol 28, 562–568 (2017). [DOI] [PubMed] [Google Scholar]
  • 165.Cremolini C et al. FOLFOXIRI plus bevacizumab versus FOLFIRI pluz 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. 16, 1306–1315 (2015). [DOI] [PubMed] [Google Scholar]
  • 166.Yaeger R et al. BRAF mutation predicts for poor outcomes after metastasectomy in patients with metastatic colorectal cancer. Cancer 120, 2316–2324 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Guren TK et al. Cetuximab in treatment of metastatic colorectal cancer: final survival analyses and extended RAS data from the NORDIC-VII study. Br. J. Cancer 116, 1271–1278 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Tol J, Nagtegaal ID & Punt CJ BRAF mutation in metastatic colorectal cancer. N. Eng. J. Med 361, 98–99 (2009). [DOI] [PubMed] [Google Scholar]
  • 169.Renaud S et al. KRAS and BRAF mutations are prognostic biomarkers in patients undergoing lung metastasectomy of colorectal cancer. Br. J. Cancer 112, 720–728 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Schirripa M et al. BRAF and RAS mutations as prognostic factors in metastatic colorectal cancer patients undergoing liver resection. Br. J. Cancer 112, 1921–1928 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Margonis GA et al. Association of BRAF mutations with survival and recurrence in surgically treated patients with metastatic colorectal liver cancer. JAMA Surg. 153, e180996 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Sanz-Garcia E, Argiles G, Elez E & Tabernero J BRAF mutant colorectal cancer: prognosis, treatment, and new perspectives. Ann. Oncol 28, 2648–2657 (2017). [DOI] [PubMed] [Google Scholar]
  • 173.Morris VK et al. Progression-free survival remains poor over sequential lines of systemic therapy in patients with BRAF-mutated colorectal cancer. Clin. Colorectal Cancer 13, 164–171 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Richman SD et al. KRAS 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 trial. J. Clin. Oncol 27, 5931–5937 (2009). [DOI] [PubMed] [Google Scholar]
  • 175.Loupakis F et al. FOLFOXIRI plus bevacizumab as first-line treatment in BRAF mutant metastatic colorectal cancer. Eur. J. Cancer 50, 57–63 (2014). [DOI] [PubMed] [Google Scholar]
  • 176.di Nicolantonio F et al. Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J. Clin. Oncol 26, 5705–5712 (2008). [DOI] [PubMed] [Google Scholar]
  • 177.Loupakis F 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 101, 715–721 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Maughan TS 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 377, 2103–2114 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Tveit KM et al. Phase III trial of cetuximab with continuous or intermittent fluorouracil, leucovorin, and oxaliplatin (Nordic FLOX) versus FLOX alone in first-line treatment of metastatic colorectal cancer: the NORDIC-VII study. J. Clin. Oncol 30, 1755–1762 (2012). [DOI] [PubMed] [Google Scholar]
  • 180.Bokemeyer C et al. Addition of cetuximab to chemotherapy as first-line treatment for KRAS wild-type metastatic colorectal cancer: pooled analysis of the CRYSTAL and OPUS randomised clinical trials. Eur. J. Cancer 48, 1466–1475 (2012). [DOI] [PubMed] [Google Scholar]
  • 181.Douillard JY et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N. Eng. J. Med 369, 1023–1034 (2013). [DOI] [PubMed] [Google Scholar]
  • 182.Stintzing S et al. 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, abstr. 445 (2014). [Google Scholar]
  • 183.Karapetis CS 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 20, 744–753 (2014). [DOI] [PubMed] [Google Scholar]
  • 184.Peeters M et al. Massively parallel tumor multigene sequencing to evaluate response to panitumumab in a randomized phase III study of metastatic colorectal cancer. Clin. Cancer Res 19, 1902–1912 (2013). [DOI] [PubMed] [Google Scholar]
  • 185.Peeters M et al. Updated analysis of KRAS/NRAS and BRAF mutations in study 20050181 of panitumumab (pmab) plus FOLFIRI for second-line treatment (tx) of metastatic colorectal cancer (mCRC). J. Clin. Oncol 32, abstr. 3568 (2014). [Google Scholar]
  • 186.Seymour M 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. 14, 749–759 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Pietrantonio F et al. Predictive role of BRAF mutations in patients with advanced colorectal cancer receiving cetuximab and panitumumab: a meta-analysis. Eur. J. Cancer 51, 587–594 (2015). [DOI] [PubMed] [Google Scholar]
  • 188.Rowland A et al. Meta-analysis of BRAF mutation as a predictive biomarker of benefit from anti-EGFR monoclonal antibody therapy for RAS wild-type metastatic colorectal cancer. Br. J. Cancer 112, 1888–1894 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Kopetz S et al. Phase II Pilot Study of Vemurafenib in Patients With Metastatic BRAF-Mutated Colorectal Cancer. J. Clin. Oncol 33, 4032–4038 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Hyman DM et al. Vermurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N. Eng. J. Med 373, 726–736 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Prahallad A et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 483, 100–103 (2012). [DOI] [PubMed] [Google Scholar]
  • 192.Corcoran RB et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2, 227–235 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Mao M et al. Resistance to BRAF inhibition in BRAF-mutant colon cancer can be overcome with PI3K inhibition or demethylating agents. Clin. Cancer Res 19, 657–667 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Yaeger R et al. Pilot trial of combined BRAF and EGFR inhibition in BRAF-mutant metastatic colorectal cancer patients. Clin. Cancer Res 21, 1313–1320 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Van Cutsem E et al. LBA-07 - Updated results 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). Ann. Oncol 26, iv119 (2015). [Google Scholar]
  • 196.van Geel RMJM et al. A phase Ib dose-escalation study of encorafenib and cetuximab with or without alpelisib in metastatic BRAF-mutant colorectal cancer. Cancer Discov. 7, 610–619 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Tabernero J et al. Phase 2 results: encorafenib (enco) and cetuximab (cetux) with or without alpelisib (alp) in patients with advanced BRAF-mutant colorectal cancer (BRAFm CRC). J. Clin. Oncol 34, abstr. 3544 (2016). [Google Scholar]
  • 198.Corcoran RB et al. Combined BRAF and MEK inhibition with dabrafenib and trametinib in BRAF V600-mutant colorectal cancer. J. Clin. Oncol 33, 4023–4031 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Flaherty KT et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N. Eng. J. Med 367, 1694–1703 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Corcoran RB et al. Combined BRAF, EGFR, and MEK Inhibition in Patients with BRAF(V600E)-Mutant Colorectal Cancer. Cancer Discov. 8, 428–443 (2018).Prospective phase I study demonstrating improved initial response rates to targeted combination therapy with dabrafenib plus panitumumab in patients with BRAFV600E-mutant metastatic CRC by adding the MEK inhibitor trametinib.
  • 201.US National Library of Medicine. ClinicalTrials.gov, https://clinicaltrials.gov/ct2/show/NCT01750918 (2012). [DOI] [PubMed]
  • 202.US National Library of Medicine. ClinicalTrials.gov, https://clinicaltrials.gov/ct2/show/NCT02928224 (2016). [DOI] [PubMed]
  • 203.Van Cutsem E et al. BEACON CRC study safety lead-in: Assessment of the BRAF inhibitor encorafenib + MEK inhibitor binimetinib + anti–epidermal growth factor receptor antibody cetuximab for BRAFV600E metastatic colorectal cancer. Ann. Oncol 29, abstr. O-027 (2018). [Google Scholar]
  • 204.Popovici V et al. Identification of a poor-prognosis BRAF-mutant-like population of patients with colon cancer. J. Clin. Oncol 30, 1288–1295 (2012). [DOI] [PubMed] [Google Scholar]
  • 205.Vecchione L et al. A vulnerability of a subset of colon cancers with potential clinical utility. Cell 165, 317–330 (2016). [DOI] [PubMed] [Google Scholar]
  • 206.Cremolini C et al. Vinorelbine in BRAF V600E mutated metastatic colorectal cancer: a prospective multicentre phase II clinical study. ESMO Open 2, e000241 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Barras D et al. BRAF V600E mutant colorectal cancer subtypes based on gene expression. Clin. Cancer Res 23, 104–115 (2017). [DOI] [PubMed] [Google Scholar]
  • 208.Pietrantonio F et al. MET-driven resistance to dual EGFR and BRAF blockade may be overcome by switching from EGFR to MET inhibition in BRAF-mutated colorectal cancer. Cancer Discov. 6, 963–971 (2016). [DOI] [PubMed] [Google Scholar]
  • 209.Oddo D et al. Emergence of MET hyper-amplification at progression to MET and BRAF inhibition in colorectal cancer. Br. J. Cancer 117, 347–352 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Hazar-Rethinam M et al. Convergent therapeutic strategies to overcome the heterogeneity of acquired resistance in BRAF(V600E) colorectal cancer. Cancer Discov. 8, 417–427 (2018).Analyses of liquid biopsy samples from patients treated with BRAFV600E targeted combination therapies show that development of aquired resistance is driven by multiple alterations in the MAPK pathway, and clonal outgrowth in preclinical models is abrogated by ERK inhibition.
  • 211.Jones JC et al. Non-V600 BRAF mutations define a clinically distinct molecular subtype of metastatic colorectal cancer. J. Clin. Oncol 35, 2624–2630 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Dankner M Targeted therapy for colorectal cancers with non-V600 BRAF mutations: perspectives for precision oncology. JCO Precision Oncology doi: 10.1200/PO.18.00195(2018). [DOI] [PubMed] [Google Scholar]
  • 213.Wan PT et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 116, 855–867 (2004). [DOI] [PubMed] [Google Scholar]
  • 214.Cremolini C et al. BRAF codons 594 and 596 mutations identify a new molecular subtype of metastatic colorectal cancer at favorable prognosis. Ann. Oncol 26, 2092–2097 (2015). [DOI] [PubMed] [Google Scholar]
  • 215.Schirripa M et al. Clinico-pathological and molecular characterisation of BRAF mutant metastatic colorectal cancer (mCRC): Are all mutations created equal? J. Clin. Oncol 36, abstr. no 3590 (2018). [Google Scholar]
  • 216.Shimada Y et al. Clinical significance of BRAF non-V600E mutations in colorectal cancer: a retrospective study of two institutions. J. Surg. Res 232, 72–81 (2018). [DOI] [PubMed] [Google Scholar]
  • 217.Shinozaki E et al. Clinical significance of BRAF non-V600E mutations on the therapeutic effects of anti-EGFR monoclonal antibody treatment in patients with pretreated metastatic colorectal cancer: the Biomarker Research for anti-EGFR monoclonal Antibodies by Comprehensive Cancer genomics (BREAC) study. Br. J. Cancer 117, 1450–1458 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.De Roock W 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. 11, 753–762 (2010). [DOI] [PubMed] [Google Scholar]
  • 219.Ross JS et al. The HER-2 receptor and breast cancer: ten years of targeted anti-HER-2 therapy and personalized medicine. Oncologist 14, 320–368 (2009). [DOI] [PubMed] [Google Scholar]
  • 220.Richman SD et al. HER2 overexpression and amplification as a potential therapeutic target in colorectal cancer: analysis of 3256 patients enrolled in the QUASAR, FOCUS and PICCOLO colorectal cancer trials. J. Pathol 238, 562–570 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Siena S et al. Targeting the human epidermal growth factor receptor 2 (HER2) oncogene in colorectal cancer. Ann. Oncol 29, 1108–1119 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Bertotti A et al. A molecularly annotated platform of patient-derived xenografts ("xenopatients") identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. Cancer Discov. 1, 508–523 (2011). [DOI] [PubMed] [Google Scholar]
  • 223.Yonesaka K et al. Activation of ERBB2 signaling causes resistance to the EGFR-directed therapeutic antibody cetuximab. Sci. Transl. Med 3, 99ra86 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Valtorta E et al. Assessment of a HER2 scoring system for colorectal cancer: results from a validation study. Mod. Pathol 28, 1481–1491 (2015). [DOI] [PubMed] [Google Scholar]
  • 225.Sartore-Bianchi A et al. Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol. 17, 738–746 (2016).Prospective trial showing an ORR of 30% to dual HER2 inhibition with trastuzumab plus lapatinib in patients with HER2-positive KRAS wild-type treatment refractory metastatic CRCs.
  • 226.Martin V et al. HER2 gene copy number status may influence clinical efficacy to anti-EGFR monoclonal antibodies in metastatic colorectal cancer patients. Br. J. Cancer 108, 668–675 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Jeong JH et al. HER2 amplification and cetuximab efficacy in patients with metastatic colorectal cancer harboring wild-type RAS and BRAF. Clin. Colorectal Cancer 16, e147–e152 (2017). [DOI] [PubMed] [Google Scholar]
  • 228.Raghav K et al. Validation of HER2 amplification as a predictive biomarker for anti–epidermal growth factor receptor antibody therapy in metastatic colorectal cancer. JCO Precision Oncol. DOI: 10.1200/PO.18.00226, 1–13 (2019). [DOI] [PubMed] [Google Scholar]
  • 229.Clark JW, Niedzwiecki D, Hollis D & Mayer R Phase-II trial of 5-fluororuacil (5-FU), leucovorin (LV), oxaliplatin (Ox), and trastuzumab (T) for patients with metastatic colorectal cancer (CRC) refractory to initial therapy. Onkologie 26, 46 (2003). [Google Scholar]
  • 230.Ramanathan RK et al. Low overexpression of HER-2/neu in advanced colorectal cancer limits the usefulness of trastuzumab (Herceptin) and irinotecan as therapy. A phase II trial. Cancer Invest. 22, 858–865 (2004). [DOI] [PubMed] [Google Scholar]
  • 231.Hainsworth JD et al. Targeted therapy for advanced solid tumors on the basis of molecular profiles: results from MyPathway, an open-label, phase IIa multiple basket study. J. Clin. Oncol 36, 536–542 (2018). [DOI] [PubMed] [Google Scholar]
  • 232.Gluck WL, Martin JC, Edenfield W,J, Chung KY & Arguello D Prolonged response of widely metastatic HER2-positive colon cancer to trastuzumab therapy. Colorect. Cancer 6, 57–61 (2017). [Google Scholar]
  • 233.Ross JS et al. Targeting HER2 in colorectal cancer: The landscape of amplification and short variant mutations in ERBB2 and ERBB3. Cancer 124, 1358–1373 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Martinelli E et al. Sequential HER2 blockade as effective therapy in chemorefractory, HER2 gene-amplified, RAS wild-type, metastatic colorectal cancer: learning from a clinical case. ESMO Open 3, e000299 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Siravegna G et al. Radiologic and genomic evolution of individual metastases during HER2 blockade in colorectal cancer. Cancer Cell 34, 148–162.e147 (2018).Analyses of liquid biopsy samples from patients with HER2-positive metastatic CRC during treatment with HER2 targeted therapy identified emerging alterations in ERBB2, RAS and PIK3CA to be associated with aquired resistance.
  • 236.Haslem DS, Ji HP, Ford JM & Nadauld LD Precision oncology strategy in trastuzumab-resistant human epidermal growth factor receptor 2–positive colon cancer: case report of durable response to ado-trastuzumab emtansine. JCO Precision Oncol., doi: 10.1200/PO.1216.00055 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Parikh A, Atreya C, Korn WM & Venook AP Prolonged response to HER2-directed therapy in a patient with HER2-amplified, rapidly progressive metastatic colorectal cancer. J. Natl. Compr. Canc. Netw 15, 3–8 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Siena S et al. HER2 amplification as a ‘molecular bait’ for trastuzumab-emtansine (T-DM1) precision chemotherapy to overcome anti-HER2 resistance in HER2 positive metastatic colorectal cancer: The HERACLES-RESCUE trial. J. Clin. Oncol 34, abstr. TPS774 (2016). [Google Scholar]
  • 239.US National Library of Medicine. ClinicalTrials.gov, https://clinicaltrials.gov/ct2/show/NCT02564900 (2015). [DOI] [PubMed]
  • 240.Yoshino T et al. Updated results of phase I study of trastuzumab deruxtecan (DS-8201a) in HER2-expressing advanced colorectal cancer. Ann. Oncol 29, abstr. 563P (2018). [Google Scholar]
  • 241.Loree JM et al. Molecular landscape of ERBB2/ERBB3 mutated colorectal cancer. J. Natl. Cancer Inst 110, 1409–1417 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Bertotti A et al. The genomic landscape of response to EGFR blockade in colorectal cancer. Nature 526, 263–267 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Kavuri SM et al. HER2 activating mutations are targets for colorectal cancer treatment. Cancer Discov. 5, 832–841 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Hyman DM et al. HER kinase inhibition in patients with HER2- and HER3-mutant cancers. Nature 554, 189–194 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.US Food and Drug Association. FDA approves larotrectinib for solid tumors with NTRK gene fusions, https://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm626720.htm (2018).
  • 246.Stransky N, Cerami E, Schalm S, Kim JL & Lengauer C The landscape of kinase fusions in cancer. Nat Commun. 5, 4846 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Madison R et al. Kinase fusions in colorectal cancers: a unique biologic subset. Ann. Oncol 29, abstr. 457PD (2018). [Google Scholar]
  • 248.Pietrantonio F et al. ALK, ROS1, and NTRK rearrangements in metastatic colorectal cancer. J. Natl. Cancer Inst 109, djx089 (2017). [DOI] [PubMed] [Google Scholar]
  • 249.Pietrantonio F et al. RET fusions in a small subset of advanced colorectal cancers at risk of being neglected. Ann. Oncol 29, 1394–1401 (2018).A case series of metastatic CRCs with RET rearrangements shows enrichment with MSI-H, RAS/BRAF wild-type tumors and inferior OS compared with RET negative cancers.
  • 250.Medico E et al. The molecular landscape of colorectal cancer cell lines unveils clinically actionable kinase targets. Nat. Commun 6, 7002 (2015). [DOI] [PubMed] [Google Scholar]
  • 251.Cremolini C et al. Negative hyper-selection of metastatic colorectal cancer patients for anti-EGFR monoclonal antibodies: the PRESSING case-control study. Ann. Oncol 28, 3009–3014 (2017). [DOI] [PubMed] [Google Scholar]
  • 252.Lee SJ et al. NTRK1 rearrangement in colorectal cancer patients: evidence for actionable target using patient-derived tumor cell line. Oncotarget 6, 39028–39035 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Drilon A et al. Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children. N. Engl. J. Med 378, 731–739 (2018).Prospective basket trial showing an ORR of 75% to the selective TRK inhibitor larotrectinib in adults and children with treatment refractory TRK fusion-positive cancers, including disease regression in three of four patients with colon cancer.
  • 254.Amatu A et al. Novel CAD-ALK gene rearrangement is drugable by entrectinib in colorectal cancer. Br. J. Cancer 113, 1730–1734 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Sartore-Bianchi A et al. Sensitivity to entrectinib associated with a novel LMNA-NTRK1 gene fusion in metastatic colorectal cancer. J. Natl. Cancer Inst 108, pii: djv306 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Demetri GD et al. Efficacy and safety of entrectinib in patients with NTRK fusion-positive (NTRK-fp) tumors: pooled analysis of STARTRK-2, STARTRK-1 and ALKA-372-001. Ann. Oncol 29, abstr. LBA17 (2018). [Google Scholar]
  • 257.Yakirevich E et al. Oncogenic ALK fusion in rare and aggressive subtype of colorectal adenocarcinoma as a potential therapeutic target. Clin. Cancer Res 22, 3831–3840 (2016). [DOI] [PubMed] [Google Scholar]
  • 258.Santos C, Sanz-Pamplona R & Salazar R RET-fusions: a novel paradigm in colorectal cancer. Ann. Oncol 29, 1340–1343 (2018). [DOI] [PubMed] [Google Scholar]
  • 259.Yaeger R et al. Clinical sequencing defines the genomic landscape of metastatic colorectal cancer. Cancer Cell 33, 125–136.e123 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Shaw AT et al. Avelumab (anti–PD-L1) in combination with crizotinib or lorlatinib in patients with previously treated advanced NSCLC: Phase 1b results from JAVELIN Lung 101. J. Clin. Oncol 36, abstr. 9008 (2018). [Google Scholar]
  • 261.Seshagiri S et al. Recurrent R-spondin fusions in colon cancer. Nature 488, 660–664 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.The Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Shinmura K et al. RSPO fusion transcripts in colorectal cancer in Japanese population. Mol. Biol. Rep 41, 5375–5384 (2014). [DOI] [PubMed] [Google Scholar]
  • 264.de Lau W, Peng WC, Gros P & Clevers H The R-spondin/Lgr5/Rnf43 module: regulator of Wnt signal strength. Genes Dev. 28, 305–316 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Giannakis M et al. RNF43 is frequently mutated in colorectal and endometrial cancers. Nat. Genet 46, 1264–1266 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Kahn M Can we safely target the WNT pathway? Nat. Rev. Drug. Discov 13, 513–532 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Fevr T, Robine S, Louvard D & Huelsken J Wnt/beta-catenin is essential for intestinal homeostasis and maintenance of intestinal stem cells. Mol. Cell. Biol 27, 7551–7559 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Reya T et al. A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature 423, 409–414 (2003). [DOI] [PubMed] [Google Scholar]
  • 269.Madan B et al. Wnt addiction of genetically defined cancers reversed by PORCN inhibition. Oncogene 35, 2197–2207 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Li C et al. Identification of RSPO2 fusion mutations and target therapy using a porcupine inhibitor. Sci. Rep 8, 14244 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Storm EE et al. Targeting PTPRK-RSPO3 colon tumours promotes differentiation and loss of stem-cell function. Nature 529, 97–100 (2016). [DOI] [PubMed] [Google Scholar]
  • 272.Janku F et al. Phase I study of WNT974, a first-in-class Porcupine inhibitor, in advanced solid tumors. Molecular Cancer Therapeutics 14, Abstr C45 (2015). [Google Scholar]
  • 273.US National Library of Medicine. ClinicalTrials.gov, https://clinicaltrials.gov/ct2/show/NCT02278133 (2017). [DOI] [PubMed]
  • 274.Rodon J et al. Biomarker analyses from a phase I study of WNT974, a first-in-class Porcupine inhibitor, in patients (pts) with advanced solid tumors. Cancer Res. 78, Abstr CT175 (2018). [Google Scholar]
  • 275.Grasso CS et al. Genetic mechanisms of immune evasion in colorectal cancer. Cancer Discov. 8, 730–749 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Taieb J et al. Prognostic Value of BRAF and KRAS Mutations in MSI and MSS Stage III Colon Cancer. J. Natl. Cancer Inst 109, djw272 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Hu J et al. Coexistence of MSI with KRAS mutation is associated with worse prognosis in colorectal cancer. Medicine (Baltimore) 95, e5649 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Siraj AK et al. A very low incidence of BRAF mutations in Middle Eastern colorectal carcinoma. Mol. Cancer 13, 168 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Lin CC et al. The prognostic role of microsatellite instability, codon-specific KRAS, and BRAF mutations in colon cancer. J. Surg. Oncol 110, 451–457 (2014). [DOI] [PubMed] [Google Scholar]
  • 280.Imamura Y et al. Analyses of clinicopathological, molecular, and prognostic associations of KRAS codon 61 and codon 146 mutations in colorectal cancer: cohort study and literature review. Mol. Cancer 13, 135 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Ogura T et al. Clinicopathological characteristics and prognostic impact of colorectal cancers with NRAS mutations. Oncol. Rep 32, 50–56 (2014). [DOI] [PubMed] [Google Scholar]
  • 282.Wangefjord S et al. Sex differences in the prognostic significance of KRAS codons 12 and 13, and BRAF mutations in colorectal cancer: a cohort study. Biol. Sex Differ 4, 17 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Samadder NJ et al. Associations between colorectal cancer molecular markers and pathways with clinicopathologic features in older women. Gastroenterology 145, 348–356.e341-342 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Eklof V et al. The prognostic role of KRAS, BRAF, PIK3CA and PTEN in colorectal cancer. Br. J. Cancer 108, 2153–2163 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Phipps AI et al. KRAS-mutation status in relation to colorectal cancer survival: the joint impact of correlated tumour markers. Br. J. Cancer 108, 1757–1764 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Nash GM et al. KRAS mutation and microsatellite instability: two genetic markers of early tumor development that influence the prognosis of colorectal cancer. Ann. Surg. Oncol 17, 416–424 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Samowitz WS et al. Microsatellite instability and survival in rectal cancer. Cancer Causes Control 20, 1763–1768 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Barault L et al. Hypermethylator phenotype in sporadic colon cancer: study on a population-based series of 582 cases. Cancer Res. 68, 8541–8546 (2008). [DOI] [PubMed] [Google Scholar]
  • 289.Andreyev HJ et al. Kirsten ras mutations in patients with colorectal cancer: the 'RASCAL II' study. Br. J. Cancer 85, 692–696 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Kadowaki S et al. Prognostic value of KRAS and BRAF mutations in curatively resected colorectal cancer. World J. Gastroenterol 21, 1275–1283 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Sinicrope FA et al. Association of DNA mismatch repair and mutations in BRAF and KRAS with survival after recurrence in stage III colon cancers : a secondary analysis of 2 randomized clinical trials. JAMA Oncol. 3, 472–480 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Ogino S et al. KRAS mutation in stage III colon cancer and clinical outcome following intergroup trial CALGB 89803. Clin. Cancer Res 15, 7322–7329 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Popovici V et al. Context-dependent interpretation of the prognostic value of BRAF and KRAS mutations in colorectal cancer. BMC Cancer. 13, 439 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Mouradov D et al. Survival in stage II/III colorectal cancer is independently predicted by chromosomal and microsatellite instability, but not by specific driver mutations. Am. J. Gastroenterol 108, 1785–1793 (2013). [DOI] [PubMed] [Google Scholar]
  • 295.Blons H et al. Prognostic value of KRAS mutations in stage III colon cancer: post hoc analysis of the PETACC8 phase III trial dataset. Ann. Oncol 25, 2378–2385 (2014). [DOI] [PubMed] [Google Scholar]
  • 296.Sinicrope FA et al. Analysis of molecular markers by anatomic tumor site in stage III colon carcinomas from adjuvant chemotherapy trial NCCTG N0147 (Alliance). Clin. Cancer Res. 21, 5294–5304 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Okuno M et al. RAS mutation is associated with unsalvageable recurrence following hepatectomy for colorectal cancer liver metastases. Ann. Surg. Oncol 25, 2457–2466 (2018). [DOI] [PubMed] [Google Scholar]
  • 298.Cercek A et al. Clinical features and outcomes of patients with colorectal cancers harboring NRAS mutations. Clin. Cancer Res 23, 4753–4760 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Dienstmann R et al. Analysis of mutant allele fractions in driver genes in colorectal cancer - biological and clinical insights. Mol. Oncol 11, 1263–1272 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Passot G et al. Is hepatectomy justified for patients with RAS mutant colorectal liver metastases? An analysis of 524 patients undergoing curative liver resection. Surgery 161, 332–340 (2017). [DOI] [PubMed] [Google Scholar]
  • 301.Summers MG et al. BRAF and NRAS locus-specific variants have different outcomes on survival to colorectal cancer. Clin. Cancer Res 23, 2742–2749 (2017). [DOI] [PubMed] [Google Scholar]
  • 302.Adenis A et al. Survival, safety, and prognostic factors for outcome with Regorafenib in patients with metastatic colorectal cancer refractory to standard therapies: results from a multicenter study (REBECCA) nested within a compassionate use program. BMC Cancer 16, 412 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Margonis GA et al. Codon 13 KRAS mutation predicts patterns of recurrence in patients undergoing hepatectomy for colorectal liver metastases. Cancer 122, 2698–2707 (2016). [DOI] [PubMed] [Google Scholar]
  • 304.Modest DP et al. Outcome according to KRAS-, NRAS- and BRAF-mutation as well as KRAS mutation variants: pooled analysis of five randomized trials in metastatic colorectal cancer by the AIO colorectal cancer study group. Ann. Oncol 27, 1746–1753 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Schirripa M et al. Role of NRAS mutations as prognostic and predictive markers in metastatic colorectal cancer. Int. J. Cancer 136, 83–90 (2015). [DOI] [PubMed] [Google Scholar]
  • 306.Petrelli F, Coinu A, Cabiddu M, Ghilardi M & Barni S KRAS as prognostic biomarker in metastatic colorectal cancer patients treated with bevacizumab: a pooled analysis of 12 published trials. Med. Oncol 30, 650 (2013). [DOI] [PubMed] [Google Scholar]
  • 307.Smith JC et al. KRAS mutations are associated with inferior clinical outcome in patients with metastatic colorectal cancer, but are not predictive for benefit with cediranib. Eur. J. Cancer 49, 2424–2432 (2013). [DOI] [PubMed] [Google Scholar]
  • 308.Tejpar S et al. 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 30, 3570–3577 (2012). [DOI] [PubMed] [Google Scholar]
  • 309.Tol J et al. Chemotherapy, bevacizumab, and cetuximab in metastatic colorectal cancer. N. Engl. J. Med 360, 563–572 (2009). [DOI] [PubMed] [Google Scholar]
  • 310.Palomba G et al. Prognostic impact of KRAS, NRAS, BRAF, and PIK3CA mutations in primary colorectal carcinomas: a population-based study. J. Transl. Med 14, 292 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Bencsikova B et al. Efficacy of bevacizumab and chemotherapy in the first-line treatment of metastatic colorectal cancer: broadening KRAS-focused clinical view. BMC Gastroenterol. 15, 37 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Saridaki Z et al. BRAFV600E mutation analysis in patients with metastatic colorectal cancer (mCRC) in daily clinical practice: correlations with clinical characteristics, and its impact on patients' outcome. PLoS One 8, e84604 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Sjoquist KM et al. Personalizing survival predictions in advanced colorectal cancer: the ARCAD nomogram project. J. Natl. Cancer Inst 110, 638–648 (2017).Analysis of pooled data from 26 randomized trials and including 22,674 patients with metastatic CRC confirm that both KRAS mutations (HR for OS 1.35, P < 0.001) and BRAF mutation (HR = 2.21, P < 0.001) are assoociated with inferior OS and PFS in multivariable models.
  • 314.Wang Y et al. Distinct impacts of KRAS, NRAS and BRAF mutations on survival of patients with metastatic colorectal cancer. J. Clin. Oncol 36, abstr. 3513 (2018). [Google Scholar]
  • 315.Tosi F et al. Effect of KRAS and BRAF mutations on survival of metastatic colorectal cancer after liver resection: a systematic review and meta-analysis. Clin. Colorectal Cancer 16, e153–e163 (2017). [DOI] [PubMed] [Google Scholar]
  • 316.Brudvik KW et al. RAS mutation clinical risk score to predict survival after resection of colorectal liver metastases. Ann. Surg 269, 120–126 (2019). [DOI] [PubMed] [Google Scholar]
  • 317.Karapetis CS et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N. Engl. J. Med 359, 1757–1765 (2008). [DOI] [PubMed] [Google Scholar]
  • 318.Arnold D et al. Prognostic and predictive value of primary tumour side in patients with RAS wild-type metastatic colorectal cancer treated with chemotherapy and EGFR directed antibodies in six randomized trials. Ann. Oncol 28, 1713–1729 (2017).Retrospective analyses of pooled data from six randomized trials with standard therapy with our without anti-EGFR antibodies in patients with RAS wild-type metastatic CRCs show that a right-sided primary tumour location might be a negative predictive factor for benefit from EGFR inhibition.
  • 319.Tian S et al. A combined oncogenic pathway signature of BRAF, KRAS and PI3KCA mutation improves colorectal cancer classification and cetuximab treatment prediction. Gut 62, 540–549 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320.Bettegowda C et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med 6, 224ra224 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321.Morelli MP et al. Characterizing the patterns of clonal selection in circulating tumor DNA from patients with colorectal cancer refractory to anti-EGFR treatment. Ann. Oncol 26, 731–736 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Russo M et al. Tumor heterogeneity and lesion-specific response to targeted therapy in colorectal cancer. Cancer Discov. 6, 147–153 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Mohan S et al. Changes in colorectal carcinoma genomes under anti-EGFR therapy identified by whole-genome plasma DNA sequencing. PLoS Genet. 10, e1004271 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Pietrantonio F et al. Heterogeneity of Acquired Resistance to Anti-EGFR Monoclonal Antibodies in Patients with Metastatic Colorectal Cancer. Clin. Cancer Res 23, 2414–2422 (2017). [DOI] [PubMed] [Google Scholar]
  • 325.Cox AD, Fesik SW, Kimmelman AC, Luo J & Der CJ Drugging the undruggable RAS: Mission possible? Nat. Rev. Drug Discov 13, 828–851 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326.Papke B & Der CJ Drugging RAS: Know the enemy. Science 355, 1158–1163 (2017). [DOI] [PubMed] [Google Scholar]
  • 327.Patricelli MP et al. Selective inhibition of oncogenic KRAS output with small molecules targeting the inactive state. Cancer Discov. 6, 316–329 (2016). [DOI] [PubMed] [Google Scholar]
  • 328.Wiesweg M et al. Impact of RAS mutation subtype on clinical outcome-a cross-entity comparison of patients with advanced non-small cell lung cancer and colorectal cancer. Oncogene 38, 2953–2966 (2019). [DOI] [PubMed] [Google Scholar]
  • 329.Tran E et al. T-cell transfer therapy targeting mutant KRAS in cancer. N. Engl. J. Med 375, 2255–2262 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Loboda A et al. EMT is the dominant program in human colon cancer. BMC Med. Genomics 4, 9 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331.Perez-Villamil B et al. Colon cancer molecular subtypes identified by expression profiling and associated to stroma, mucinous type and different clinical behavior. BMC Cancer. 12, 260 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.Schlicker A et al. Subtypes of primary colorectal tumors correlate with response to targeted treatment in colorectal cell lines. BMC Med. Genomics 5, 66 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 333.Budinska E et al. Gene expression patterns unveil a new level of molecular heterogeneity in colorectal cancer. J. Pathol 231, 63–76 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334.Sadanandam A et al. A colorectal cancer classification system that associates cellular phenotype and responses to therapy. Nat. Med 19, 619–625 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335.De Sousa E. Melo F et al. Poor-prognosis colon cancer is defined by a molecularly distinct subtype and develops from serrated precursor lesions. Nat. Med 19, 614–618 (2013). [DOI] [PubMed] [Google Scholar]
  • 336.Marisa L et al. Gene expression classification of colon cancer into molecular subtypes: characterization, validation, and prognostic value. PLoS Med. 10, e1001453 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 337.Roepman P et al. Colorectal cancer intrinsic subtypes predict chemotherapy benefit, deficient mismatch repair and epithelial-to-mesenchymal transition. Int. J. Cancer 134, 552–562 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338.Calon A et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat. Genet 47, 320–329 (2015).TGF-β signalling enhances the stimulating effect of cancer-associated fibroblasts on tumour-initiating cells, and disease progression in preclinical models can be halted by inhibiting the crosstalk between cancer cells and cancer-associated fibroblasts by inhibition of TGF-β signalling.
  • 339.Bramsen JB et al. Molecular-subtype-specific biomarkers improve prediction of prognosis in colorectal cancer. Cell Rep. 19, 1268–1280 (2017). [DOI] [PubMed] [Google Scholar]
  • 340.Isella C et al. Selective analysis of cancer-cell intrinsic transcriptional traits defines novel clinically relevant subtypes of colorectal cancer. Nat Commun. 8, 15107 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341.Sveen A et al. Colorectal cancer Consensus Molecular Subtypes translated to preclinical models uncover potentially targetable cancer-cell dependencies. Clin. Cancer Res 24, 794–806 (2018). [DOI] [PubMed] [Google Scholar]
  • 342.Marisa L et al. Clinical utility of colon cancer molecular subtypes: Validation of two main colorectal molecular classifications on the PETACC-8 phase III trial cohort. J. Clin. Oncol 35, abstr. 3509 (2017). [Google Scholar]
  • 343.Yamaguchi S et al. A validation study of stratification by the 55-gene classifier for assessing recurrence risk in stage II colon cancer: The 55 STAR study (UMIN23879). J. Clin. Oncol 36, abstr. 3526 (2018). [Google Scholar]
  • 344.Laurent-Puig P et al. Colon cancer molecular subtype intratumoral heterogeneity and its prognostic impact: An extensive molecular analysis of the PETACC-8. Ann. Oncol 29, abstr. 60PD (2018). [Google Scholar]
  • 345.Fontana E, Eason K, Cervantes A, Salazar R & Sadanandam A Context matters - consensus molecular subtypes of colorectal cancer as biomarkers for clinical trials. Ann. Oncol, doi: 10.1093/annonc/mdz1052 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Piskol R et al. A clinical applicable gene expression classifier reveals intrinsic and extrinsic contributions to consensus molecular subtypes in primary and metastatic colon cancer. Clin. Cancer Res doi: 10.1158/1078-0432.CCR-18-3032(2019). [DOI] [PubMed] [Google Scholar]
  • 347.Trumpi K et al. Neoadjuvant chemotherapy affects molecular classification of colorectal tumors. Oncogenesis 6, e357 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.Dunne PD et al. Cancer-cell intrinsic gene expression signatures overcome intratumoural heterogeneity bias in colorectal cancer patient classification. Nat Commun 8, 15657 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.De Smedt L et al. Expression profiling of budding cells in colorectal cancer reveals an EMT-like phenotype and molecular subtype switching. Br. J. Cancer 116, 58–65 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350.Trinh A et al. Practical and robust identification of molecular subtypes in colorectal cancer by immunohistochemistry. Clin. Cancer Res 23, 387–398 (2016). [DOI] [PubMed] [Google Scholar]
  • 351.Lenz HJ et al. Impact of consensus molecular subtype on survival in patients with metastatic colorectal cancer: results from CALGB/SWOG 80405 (Alliance). J. Clin. Oncol doi: 10.1200/JCO.18.02258(2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Song N et al. Clinical outcome from oxaliplatin treatment in stage II/III colon cancer according to intrinsic subtypes: secondary analysis of NSABP C-07/NRG Oncology randomized clinical trial. JAMA Oncol. 2, 1162–1169 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Okita A et al. Consensus molecular subtypes classification of colorectal cancer as a predictive factor for chemotherapeutic efficacy against metastatic colorectal cancer. Oncotarget 9, 18698–18711 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354.Teufel M et al. Molecular subtypes and outcomes in regorafenib-treated patients with metastatic colorectal cancer (mCRC) enrolled in the CORRECT trial. J. Clin. Oncol 33, abstr. 3558 (2015). [Google Scholar]
  • 355.Linnekamp JF et al. Consensus molecular subtypes of colorectal cancer are recapitulated in in vitro and in vivo models. Cell Death Differ. 25, 616–633 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 356.Le DT et al. A blueprint to advance colorectal cancer immunotherapies. Cancer Immunol. Res 5, 942–949 (2017). [DOI] [PubMed] [Google Scholar]
  • 357.Garg AD et al. Trial watch: immunogenic cell death induction by anticancer chemotherapeutics. Oncoimmunology 6, e1386829 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 358.Nordlinger B et al. Perioperative FOLFOX4 chemotherapy and surgery versus surgery alone for resectable liver metastases from colorectal cancer (EORTC 40983): long-term results of a randomised, controlled, phase 3 trial. Lancet Oncol. 14, 1208–1215 (2013). [DOI] [PubMed] [Google Scholar]
  • 359.Dosset M et al. PD-1/PD-L1 pathway: an adaptive immune resistance mechanism to immunogenic chemotherapy in colorectal cancer. Oncoimmunology 7, e1433981 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 360.Grothey A et al. Fluoropyrimidine (FP) + bevacizumab (BEV) + atezolizumab vs FP/BEV in BRAFwt metastatic colorectal cancer (mCRC): Findings from Cohort 2 of MODUL – a multicentre, randomized trial of biomarker-driven maintenance treatment following first-line induction therapy. Ann. Oncol 29, abstr. LBA19 (2018). [Google Scholar]
  • 361.Tauriello DVF et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 554, 538–543 (2018).Expression of TGF-β in the tumour microenvironment is a mechanism of immune evasion, and inhibition of TGF-β in mice with CRC liver metastases promotes an anti-tumour cytotoxic T cell response and renders the tumours sensitive to ICIs.
  • 362.Lan Y et al. Enhanced preclinical antitumor activity of M7824, a bifunctional fusion protein simultaneously targeting PD-L1 and TGF-beta. Sci. Transl. Med 10, eaan5488 (2018). [DOI] [PubMed] [Google Scholar]
  • 363.Kopetz S et al. M7824 (MSB0011359C), a bifunctional fusion protein targeting PD-L1 and TGF-β, in patients with heavily pretreated CRC: preliminary results from a phase I trial. J. Clin. Oncol 36, abstr. 764 (2018). [Google Scholar]
  • 364.Ebert PJR et al. MAP kinase inhibition promotes T cell and anti-tumor activity in combination with PD-L1 checkpoint blockade. Immunity 44, 609–621 (2016). [DOI] [PubMed] [Google Scholar]
  • 365.Bendell JC et al. A phase Ib study of safety and clinical activity of atezolizumab (A) and cobimetinib (C) in patients (pts) with metastatic colorectal cancer (mCRC). J. Clin. Oncol 36, abstr. 560 (2018). [Google Scholar]
  • 366.Eng C et al. Atezolizumab with or without cobimetinib versus regorafenib in previously treated metastatic colorectal cancer (IMblaze370): a multicentre, open-label, phase 3, randomised, controlled trial. Lancet Oncol. doi: 10.1016/S1470-2045(19)30027-0(2019). [DOI] [PubMed] [Google Scholar]
  • 367.Becht E et al. Immune and stromal classification of colorectal cancer is associated with molecular subtypes and relevant for precision immunotherapy. Clin. Cancer Res 22, 4057–4066 (2016). [DOI] [PubMed] [Google Scholar]
  • 368.Meyer C et al. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol. Immunother 63, 247–257 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369.Weber R et al. Myeloid-derived suppressor cells hinder the anti-cancer activity of immune checkpoint inhibitors. Front. Immunol 9, 1310 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 370.Kim K et al. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc. Natl. Acad. Sci. U. S. A 111, 11774–11779 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 371.Azuaje F Artificial intelligence for precision oncology: beyond patient stratification. NPJ Precis. Oncol 3, 6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Lipson E et al. Durable cancer regression off-treatment and effective re-induction therapy with an anti-PD-1 antibody. Clin. Cancer Res 19, 462–468 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 373.Hochster HS et al. Efficacy and safety of atezolizumab (atezo) and bevacizumab (bev) in a phase Ib study of microsatellite instability (MSI)-high metastatic colorectal cancer (mCRC). J. Clin. Oncol 35, 673–673 (2017). [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental table

RESOURCES