Abstract
Over the last few decades, molecularly targeted agents have been used for the treatment of metastatic colorectal cancer. They have made remarkable contributions to prolonging the lives of patients. The emergence of several biomarkers and their introduction to the clinic have also aided in guiding such treatment. Recently, next-generation sequencing (NGS) has enabled clinicians to identify these biomarkers more easily and reliably. However, there is considerable uncertainty in interpreting and implementing the vast amount of information from NGS. The clinical relevance of biomarkers other than NGS are also subjects of debate. This review covers controversial issues and recent findings on such therapeutics and their molecular targets, including VEGF, EGFR, BRAF, HER2, RAS, actionable fusions, Wnt pathway and microsatellite instability for comprehensive understanding of obstacles on the road to precision oncology in metastatic colorectal cancer.
Keywords: colorectal neoplasms, biomarkers, antineoplastic agents
Introduction
Colorectal cancer (CRC) is the third most common cancer worldwide, comprising a considerable portion of the disease burden. About half of new cases and deaths occur in Asia,1 where awareness of CRC has risen in recent times. Approximately 20% of new cases present with distant metastases2 and 20%–25% of localised cases eventually experience recurrence.3 4 Systemic chemotherapy as well as multidisciplinary curative approaches have improved the survival of patients with metastatic CRC (mCRC).
Although the backbone of systemic therapy for mCRC still remains cytotoxic agents, new targets and therapeutics have emerged in the last few decades based on an improved understanding of the biology of CRC. Furthermore, the use of next-generation sequencing (NGS) tests to guide targeted therapy in mCRC has become increasingly prevalent in clinical settings due to falling costs. However, the NGS test results provide a bulk of information that requires careful interpretation. Besides, other biomarkers such as consensus molecular subtype (CMS), circulating tumour DNA (ctDNA), plasma proteins, microRNA and sidedness have been suggested to predict the efficacy of targeted agents. However, the clinical relevance of these markers remains controversial.
This review examines clinical and translational data concerning targeted agents for mCRC and deals with issues regarding the predictive value of various biomarkers.
Anti-angiogenic agents: still the universal answer to mCRC?
Several anti-angiogenic agents such as bevacizumab,5 aflibercept,6 ramucirumab7 and regorafenib,8 which are designed to bind to vascular endothelial growth factor (VEGF) or VEGF receptors (VEGFR), have been approved for the treatment of mCRC. Patient subgroups who benefit more from anti-VEGF treatment have not been clearly defined based on clinical characteristics or biomarkers. Several translational projects with randomised trials explored mutations in RAS,9–11BRAF10 or phosphatidylinositol 3-kinase catalytic alpha polypeptide (PIK3CA)11 and other biomarkers (table 1). Recently, microsatellite instability (MSI) status and CMS, a transcriptome-based molecular subtype identified by an international consortium study, have been suggested to predict the efficacy of bevacizumab. Briefly, CMS 1 and high MSI (MSI-H) were associated with a clinical benefit from bevacizumab, as opposed to cetuximab, in one of the largest translational sets analysed thus far, the CALGB/SWOG 80405 trial.12 13 However, these results were not reproduced in a similar FIRE-3 study, which conducted a head-to-head trial of bevacizumab versus cetuximab.14 In contrast, the AGITG MAX study showed better progression-free survival (PFS) in patients with CMS 2 or 3 (but not CMS 1) when they were administered bevacizumab along with chemotherapy (capecitabine or capecitabine plus mitomycin), as opposed to chemotherapy alone.15 These inconsistent results could be explained by the relatively small number of CMS 1 patients enrolled (less than 20% of the mCRC cases) and different platforms used for gene expression profiling (Almac Xcel array in FIRE-3 and AGITG MAX, and NanoString in CALGB) (table 2).
Table 1.
Biomarker studies from randomised trials comparing anti-VEGF or anti-EGFR antibodies to NO targeted therapies for mCRC
Study (author/year) |
Number of patients (analysed for biomarker/ randomised) | Design | Biomarker analysis platform | Analysed markers | Results |
AVF2107 (Hurwitz et al, 2009)9 |
230/813 | 1st line, Bev+IFL vs placebo+IFL | Direct sequencing | KRAS mutation | PFS-related benefit of Bev similar for KRAS-mt (HR 0.41, p=0.00008) and KRAS-wt (HR 0.44, p<0.0001) |
ORR better in Bev arm for KRAS-wt (60.0% vs 37.3%, p=0.006), but not for KRAS-mt (43.2% vs 41.2%, p=0.86) | |||||
VELOUR (Wirapati et al, 2017)10 |
482/1226 | 2nd line, aflibercept+FOLFIRI vs placebo+FOLFIRI | NGS | Extended RAS, BRAF mutation and transcriptome | Non-significant trend of OS-related benefit with aflibercept for BRAF mt (interaction p=0.08) and BRAF mt-like RNA signature (interaction p=0.2) |
Affymetrix gene chip | |||||
AGITG MAX (Price et al, 2015)11 |
280/471 | 1st line, bevacizumab+CTx vs CTx |
Pyrosequencing | RAS, PIK3CA mutation | None were prognostic or predictive of bevacizumab outcome |
AGITG MAX (Mooi et al, 2018)15 |
237/471 | 1st line, bevacizumab+CTx vs CTx | Almac Xcel microarray | CMS | Benefit of bevacizumab in terms of PFS in CMS2 (HR 0.44, 95% CI 0.29 to 0.68) and CMS3 (HR 0.35, 95% CI 0.14 to 0.86), interaction p=0.04 in multivariate analysis |
RAISE (Tabernero et al, 2018) |
894/1072 | 2nd line, ramucirumab+FOLFIRI vs placebo+FOLFIRI | Dual-monoclonal sandwich immunoassay | VEGF-C, VEGF-D, soluble VEGFR-1, soluble VEGFR2 and soluble VEGFR-3 | High VEGF-D level (≥115 pg/mL) predicted benefit from OS (HR 0.73, 95% CI 0.60 to 0.89, interaction p=0.0005) and PFS (HR 0.62, 95% CI 0.52 to 0.74, interaction p<0.0001) |
RAISE (Yoshino et al, 2019) |
912/1072 | 2nd line, ramucirumab+FOLFIRI vs placebo+FOLFIRI | Multiplex qPCR (Modaplex system, Qiagen) | RAS, BRAF mutation | No treatment-by-RAS/BRAF mutation status interaction (p=0.523 for OS, 0.655 for PFS), but numerically good OS in BRAF with ramucirumab (HR 0.54, p=0.103) |
CORRECT (Tabernero et al, 2015)119 |
503/760 (genetic biomarker) 611/760 (protein biomarker) |
3rd line, regorafenib vs placebo | BEAMing of plasma DNA, FoundationOne panel for tumour tissue, ELISA for 15 proteins of interest | KRAS, PIK3CA and BRAF mutation, plasma proteins including angiopoietin 2, interleukin 6, etc | None were predictive of PFS and OS-related benefit of regorafenib |
CRYSTAL+OPUS (Bokemeyer et al, 2012)30 |
800/1535 | 1st line, cetuximab+FOLFOX or FOLFIRI vs FOLFOX or FOLFIRI | PCR clamping and melting curve method | KRAS, BRAF mutation | Similar benefit of cetuximab in terms of ORR, PFS and OS in both BRAF-wt and BRAF-mt |
CO-17 (Karapetis et al, 2014)31 |
407/572 | 3rd line, cetuximab vs BSC | Nested PCR, IHC | PIK3CA, BRAF mutation and PTEN expression | None were predictive of PFS and OS-related benefit of cetuximab |
20100007 (Kim et al, 2018)120 |
270/377 | 3rd line, panitumumab vs BSC | Sanger sequencing | RAS, BRAF mutation | In BRAF mt (n=20), HR for OS favoured the panitumumab arm (HR 0.39, p=0.1597) and marginal benefit in terms of PFS was shown (HR 0.277, p=0.0502) |
Bev, bevacizumab; BSC, best supportive care; CMS, consensus molecular subtype; CTx, chemotherapy; EGFR, epidermal growth factor receptor; FOLFIRI, folinic acid, 5-fluorouracil and irinotecan; FOLFOX, folinic acid, 5-fluorouracil and oxaliplatin; IFL, irinotecan, 5-fluorouracil and leucovorin; IHC, immunohistochemical staining; mCRC, metastatic colorectal cancer; mt, mutant; NGS, next-generation sequencing; ORR, overall response rate; OS, overall survival; PFS, progression-free survival; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; wt, wild-type.
Table 2.
Biomarker study from randomised trials comparing anti-VEGF to anti-EGFR therapy for mCRC
Study—author, year | Number of patients (analysed for biomarker/ randomised) |
Design | Biomarker analysis platform | Analysed markers | Results | |
CALGB80405 | Lenz et al, 201912 |
663/1137 | 1st line, Bev+FOLFOX or FOLFIRI vs Cmab+FOLFOX or FOLFIRI |
Nanostring | CMS | Poorer OS (HR 2.34, p<0.001) and PFS (HR 2.28, p<0.001) with Cmab than with Bev in CMS1 (n=104) |
Better OS (HR 0.62, p=0.0046) with Cmab than with Bev in CMS2 (n=242) | ||||||
Innocenti et al, 201913 |
843/1137 | FoundationOne | 426 genes and 5 microsatellite markers | Better OS (HR 0.16, p<0.001) and PFS (HR 0.13, p<0.001) with Bev than with Cmab in MSI-H (n=52) | ||
Promega for MSI | ||||||
Nixon et al, 201621 | 715/1137 | ELISA | 23 plasma biomarkers | Low VEGF-D predicted PFS benefit from Bev (HR 1.70) rather than Cmab (HR 0.92) (interaction p=0.0097) Low PlGF predicted PFS benefit from Bev (HR 1.50) rather than Cmab (HR 0.94, interaction p=0.0298) |
||
FIRE3 | Stintzing et al, 201714 |
313/588 | 1st line, Bev+FOLFIRI vs Cmab+FOLFIRI |
Almac Xcel array | CMS | Better PFS (HR 0.63, p=0.031) and OS (HR 0.52, p=0.012) with Cmab than with Bev in CMS4 (n=104) in unadjusted analysis |
Laurent-Puig et al, 201946 |
340/592 | Taqman assay | miR-31-3p | Better PFS (HR 0.74, p=0.05), OS (HR 0.61, p<0.01), and objective response with Cmab than with Bev in low miR-31-3p expressers; no difference in high expressers | ||
Berger et al, 2017121 |
522/586 | PCR-based direct sequencing | SVCT1, SVCT2 and Glut1 gene polymorphism | SVCT1 CC genotype was associated poorer PFS and OS than any T genotype in Bev arm with KRAS mutation but not in Cmab arm in unadjusted analysis | ||
Heinemann et al, 2018122 |
373/592 | FoundationOne | 426 genes | No benefit with Bev in terms of OS (HR 1.17, p=0.82) in MSI-H (n=10); benefit of Cmab was marginally favourable (HR 0.75, p=0.08) in MAPK-wt (n=178); TMB or other markers could not be validated as prognostic or predictive | ||
Miller-Phillps et al, 2019123 | 333/592 | Almac Xcel array | miR-21 | Better ORR (80.0% vs 57.9%, p=0.005) and OS (HR 0.625, p=0.005) with Cmab than with Bev in low miR-21 subgroup (n=166) | ||
Stintzing et al, 2014124 | 299/592 | Direct sequencing | AREG SNP rs161511 | AREG A/G genotype was associated with poorer ORR (38% vs 79%, p=0.02), PFS (HR 3.46, p=0.001) and OS (HR 3.87, p=0.001) compared with G/G genotype in Cmab arm but not in Bev arm |
AREG, amphiregulin; Bev, bevacizumab; Cmab, cetuximab; CMS, consensus molecular subtype; EGFR, epidermal growth factor receptor; FOLFIRI, folinic acid, 5-fluorouracil and irinotecan; FOLFOX, folinic acid, 5-fluorouracil, and oxaliplatin; Glut, glucose transporter; mCRC, metastatic colorectal cancer; MSI, microsatellite instability; ORR, overall response rate; OS, overall survival; PFS, progression-free survival; PlGF, placental growth factor; SVCT, sodium-dependent vitamin C transporter; TMB, tumour mutational burden; VEGF, vascular endothelial growth factor.
Apart from genetic or transcriptomic profiles, biomarkers such as polymorphisms in VEGF-A16 or VEGFR-117 or changes in circulating angiogenic factors18 19 may be associated with benefits from bevacizumab in mCRC. However, most of these were tested in single-arm studies of bevacizumab, making their predictive impact difficult to assess. A recent study of second-line randomised trials with ramucirumab showed high plasma levels of VEGF-D, a ligand to VEGFR-2, predicted benefits with overall survival (OS) and PFS.20 These results are contrary to those from CALGB/SWOG 80405, which showed that low VEGFR-D level predicted benefits from bevacizumab+fluorouracil, leucovorin and oxaliplatin (FOLFOX).21 In the light of such confounding results, the development and validation of a reliable assay method for the plasma biomarkers is required (table 1).
Anti-epidermal growth factor receptor antibodies: an arena of diverse biomarkers
The anti-epidermal growth factor receptor (EGFR) antibodies, cetuximab and panitumumab, have been approved for front-line treatment of mCRC in combination with cytotoxic chemotherapy22 23 and later-line treatment as monotherapy or combination therapy.24 The clinical benefit from anti-EGFR antibodies is restricted to patients with wild-type RAS.23 25 Other genetic alterations in the EGFR signalling pathway such as PIK3CA mutation, phosphatase and tensin homolog (PTEN) loss, BRAF mutation and human epidermal growth factor receptor 2 (HER2) amplification have been associated with anti-EGFR resistance in retrospective series or single-arm phase II studies.26–29 Although these associations are highly plausible, there has been no statistically significant evidence from randomised trials showing that the magnitude of benefit from anti-EGFR antibodies is significantly jeopardised in such subgroups.30–32 This is probably because the incidence of these alterations is so rare that the subgroup analysis lacked sufficient power to prove their association with resistance or because the alterations are less potent than RAS mutations in terms of conferring resistance (table 2).
RAS and other genetic alterations that emerge during anti-EGFR treatment detected in tumour tissue as well as ctDNA have recently arisen as markers of acquired resistance. Up to 30%–40% of patients administered with anti-EGFR show RAS mutations in their plasma ctDNA at the time of disease progression.33–35 Mutations in the EGFR ectodomain (S492R) also confer resistance to anti-EGFR treatment, although the degree of resistance differs between cetuximab and panitumumab due to their different binding epitopes.36 Amplification of receptor tyrosine kinases (HER2 or MET) or BRAF mutations that emerge during anti-EGFR treatment have been suggested to be markers of acquired resistance.37
However, the clinical effectiveness of serial monitoring of ctDNA during anti-EGFR treatment has not been well established yet. According to a study that serially measured ctDNA during anti-EGFR treatment, RAS mutations appeared 3–4 months earlier than clinical progression, and the emergence of these mutations was not correlated with PFS.35 There has been no evidence that early switch of regimen in response to the emergence of ctDNA RAS mutation is more beneficial in terms of OS than conventional switch on clinical progression. However, once clinical progression occurs after anti-EGFR treatment, measurement of ctDNA might be helpful in guiding further treatment. A phase II study in patients previously treated with anti-EGFR proposed ctDNA RAS mutation as a predictors of response to anti-EGFR rechallenge.38 Knowing the dynamics of emergent RAS mutations after progression could help determine the optimal timing of anti-EGFR antibody rechallenge. A recent study showed that anti-EGFR resistant clones with RAS and EGFR mutations at progression after treatment with anti-EGFR antibodies decayed exponentially after anti-EGFR cessation with a cumulative half-life of 4.4 months.39
MSI-H has been associated with poor prognosis in patients treated with anti-EGFR antibodies, as compared with bevacizumab.13 40 Reduced EGFR ligand expression due to hypermethylation typically seen in MSI-H tumours could explain anti-EGFR resistance; however, not all MSI-H tumours exhibit a hypermethylation phenotype, especially in Asian countries.41 The precise mechanism of resistance in MSI-H tumours remains unknown.
MSI-H largely overlaps with right primary tumours, which are also adversely associated with anti-EGFR resistance. Subgroup analyses and systematic reviews of randomised trials have consistently revealed a lack of benefit from cetuximab or panitumumab in terms of PFS and OS in right-sided tumours in a front-line setting.42 43 Right-sided tumours more frequently harbour biomarkers associated with anti-EGFR resistance (RAS, BRAF, PIK3CA mutations and reduced EGFR ligand expression) than their left-sided counterparts. However, the CALGB study showed that sidedness was negatively associated with poor OS in cetuximab therapy as compared with bevacizumab therapy after adjusting for the aforementioned biomarker profiles.44 Several studies have shown more favourable tumour shrinkage with anti-EGFR therapy than with bevacizumab for right-sided tumours, suggesting that anti-EGFR antibodies could provide a means of achieving rapid control of tumour volume for certain classes of right-sided tumours.45
Transcriptional biomarkers have also been studied in association with cetuximab efficacy (table 2). Upregulation of a specific microRNA, miR-31-3p, plays a significant role in activating RAS signalling and was identified as a potential negative predictor of cetuximab efficacy in the FIRE-3 study.46 As seen with bevacizumab, the associations between CMS subtype and cetuximab efficacy are inconsistent between studies. A relative benefit of cetuximab as compared with bevacizumab was observed for CMS 4 in the FIRE-3 study but for CMS 2 in the CALGB dataset. The change in CMS has also been associated with acquired resistance; a paired biopsy study revealed that transcriptional change (switch of CMS from 2 to 4) with increased infiltration of cancer-associated fibroblasts was seen in tissues obtained after progression.47
From a clinical perspective, front-line anti-EGFR treatment generally produces better objective response rates (ORRs) and increased tumour shrinkage.48 Thus, this treatment is favoured over bevacizumab, especially for patients with borderline-resectable metastases or with high tumour burdens. However, anti-EGFR treatment usually causes skin toxicity and emotional stress, which could hinder the social lives of patients.49 Therefore, it is important to select the best-fit candidates for front-line anti-EGFR treatment based on predictive markers such as sidedness, RAS mutation or MSI. Comprehensive tumour profiling such as the PRESSING panel, a platform incorporating NGS, immunohistochemical staining (IHC), in situ hybridisation (ISH) and RNA sequencing,40 50 could help in optimising anti-EGFR treatment for mCRC.
Strategies targeting BRAF-mutant CRC
The poor prognosis of BRAF V600E mutant mCRC has been consistently seen in every clinical trial conducted so far, along with real-world data.51 52 Unlike BRAF-mutant melanoma, BRAF mutant CRC does not respond to BRAF inhibitor monotherapy due to parallel EGFR activation by negative downstream feedback.53 Several clinical trials have tested the strategy of blocking both of upstream (EGFR) and downstream (BRAF) elements of these pathways,54–56 which are active in BRAF-mutant mCRC (table 3). The SWOG1406 randomised phase II trial showed improved PFS with a combination of vemurafenib (BRAF inhibitor), cetuximab and irinotecan combination (VIC) as compared with just cetuximab and irinotecan.57 Recently, the BEACON randomised phase III trial showed that the triplet combination of encorafenib (BRAF inhibitor), binimetinib (mitogen-activated protein kinase kinase (MEK) inhibitor) and cetuximab showed improved OS when compared with the control arm (cetuximab+irinotecan-based chemotherapy). In this study, the doublet combination of encorafenib and cetuximab also showed improved OS as compared with the control arm.58 While this study did not permit cross-over between the arms, 48% of patients in the control arm of the SWOG trial did cross over to receive VIC, resulting in a slight, but statistically insignificant, improvement in OS.
Table 3.
Results of recent clinical trials on mCRC with BRAF V600E mutation
Study (author, year) |
Phase | N | Eligibility | Treatment | Results |
BEACON (Kopetz et al, 2019)58 |
III | 665 | PD after 1 or 2 prior treatments | Triplet: encorafenib+binimetinib+Cmab Doublet: encorafenib+Cmab Control: Cmab+irinotecan or Cmab+FOLFIRI |
Triplet vs doublet vs control: OS 9.0 m vs 8.4 m vs 5.4 m (p<0.001) PFS 4.3 m vs 4.2 m vs 1.5 m (p<0.0001) ORR 26% vs 20% vs 2% (p<0.001) |
SWOG S1406 (Kopetz et al, 2017)57 |
II | 106 | PD after 1 or 2 prior treatments | Vemurafenib+cetuximab+irinotecan vs cetuximab+irinotecan | PFS 4.3 m vs 2.0 m (p=0.001) OS 9.6 m vs 5.9 m (p=0.19) |
NCT01072175 (Corcoran et al, 2015)56 |
II | 43 | Any line | Dabrafenib+trametinib | ORR 12%, PFS 3.5 m |
NCT01750918 (Corcoran et al, 2018)125 |
I | 20 | Any line | Pmab+trametinib | ORR 0%, PFS 2.6 m |
91 | ≥1 prior treatment | Dabrafenib+Pmab+trametinib | ORR 21%, PFS 4.2 m | ||
20 | Dabrafenib+Pmab | ORR 10%, PFS 3.5 m | |||
NCT01750918 (Van Geel et al, 2017)126 |
I | 28 | Encorafenib+Cmab+alpelisib | ORR 17.9%, PFS 4.2 m | |
26 | Encorafenib+Cmab | ORR 19.2%, PFS 3.7 m |
Cmab, cetuximab; FOLFIRI, folinic acid, 5-fluorouracil and irinotecan; m, months; ORR, overall response rate; OS, overall survival; PD, progressive disease; PFS, progression-free survival; Pmab, panitumumab.
Given the high rate of grade 3 or 4 toxicity of the VIC regimen (nausea, diarrhoea and neutropenia in more than 20% of patients), encorafenib-based triplet or doublet combinations, which do not contain cytotoxic agents, appeared to be more feasible options for BRAF-mutant patients. The triplet regimen is now being tested for untreated populations in a phase II trial (NCT03693170). Until now, based on the results from subgroup analysis of the TRIBE trial, there has been a consensus that the optimal front-line treatment for BRAF mutant patients might be intensive chemotherapeutic regimens (bevacizumab+5-fluorouracil, oxaliplatin and irinotecan; FOLFOXIRI) to mitigate the aggressive biology.59 It would be worthwhile to evaluate if the targeted regimen (encorafenib and cetuximab with or without binimetinib) without cytotoxic agents could prove more effective than this intensive combination as a frontline treatment for BRAF V600-mutant mCRC.
Although the V600 mutation is the most common type of BRAF mutation, non-V600 BRAF mutations are being more frequently detected with NGS tests becoming more widely available. Non-V600 mutations account for 20%–40% of all BRAF mutations in mCRC60–62 and represent different clinical characteristics from those of V600 mutants. These differences manifest as fewer female patients, lower histological grades and greater incidence of left-sidedeness in the non-V600 mutants as compared with V600 mutants.60 63 Based on the degrees of RAS-dimer and RAF-dimer dependency in the signalling pathway, BRAF mutations are categorised as class 1 (V600 mutants: RAS-independent, dimer-independent and kinase-active), class 2 (RAS-independent, dimer-dependent and kinase-active) and class 3 (RAS-dependent, dimer-dependent and kinase-inactive). BRAF inhibitors show limited activity in class 2 and 3 mutants, which exhibit RAF dimer-dependent signalling.64 65 Class 3 BRAF mutations, which comprise more than half of non-V600 BRAF mutants, frequently overlap with RAS mutations. However, in the case of class 3 BRAF mutants with wild-type RAS, inhibiting the RAS signal with anti-EGFR antibodies could be a reasonable option59; moreover, anti-EGFR inhibitors when combined with MEK inhibitor can prevent feedback activation by BRAF inhibition and have been proposed as a more rational approach.64 Class 2 BRAF mutants are difficult to target due to RAS-independent kinase activity, although treatment options such as combinations of anti-EGFR, MEK and/or ERK inhibitors would be worth further exploration. For non-V600 BRAF mutant mCRC, the triplet regimen from the BEACON trial is currently being tested in a phase II trial (UMIN000031857).
HER2 blockades in CRC
HER2 amplification, observed in 2%–4% of mCRC cases, shows a predilection for the left colon or rectum and is mainly enriched in RAS and BRAF wild-type cancer; however, it has also been associated with anti-EGFR resistance.29 66 67 Unlike breast cancer, anti-HER2 antibody (trastuzumab) monotherapy has not been successful in treating HER2-amplified mCRC. This is likely due to delayed EGFR and HER3 activation following trastuzumab monotherapy may cause intrinsic resistance.66 68 Dual blockade targeting HER2 and EGFR/HER3 is therefore required for this disease subset.
Several trials have shown the clinical activity of a combination strategy for HER2 blockade (table 4). The HERACLES investigators defined certain CRC-specific criteria for IHC staining in HER2, which were concordant with ISH parameters, and screened more than 900 KRAS wild-type patients, of which 5% were HER2 positive. Trastuzumab and lapatinib showed promising activity in the heavily treated patients,69 and correlative biomarker analysis revealed the HER2 copy number in the tissue and ctDNA predicted the response to the treatment.70 The results of the MyPathway trial demonstrated the effectiveness of the combining pertuzumab with trastuzumab for this population, with a profound difference in outcome based on KRAS mutation status (table 4). DS-8201a, a novel HER2-targeted antibody–drug conjugate with trastuzumab and topoisomerase I inhibitor (deruxtecan) payload, also demonstrated clinical activity in HER2+mCRC in the dose expansion cohort of a phase I trial, thus warranting the currently ongoing phase II trial.25 Recently, a study from Japan showed that ctDNA could be used for negative selection of candidates for this combination; patients with ctDNA alterations of RAS, BRAF, PIK3CA and HER2 did not benefit from the dual HER2 blockade.71 Another dual combination comprising inhibitors of HER2, trastuzumab and tucatinib showed significant activity, with an ORR of 55% and median PFS of 6.2 months in 22 patients,72 while pertuzumab and trastuzumab-emtansine (T-DM1) produced an ORR of 10%, which did not meet the primary endpoint.73 With advancements in efficient HER2 blockades, including antibody–drug conjugates, bispecific antibodies, small molecule inhibitors or combinations with immunotherapy,74 more innovative therapeutics could emerge in this field.
Table 4.
Results of recent clinical trials of HER2-positive mCRC
Study (author/year) |
Phase | N | Eligibility | Treatment | Results |
HERACLES (Sartore-Bianchi et al, 2016)69 |
II | 27 | KRAS wt, progression after all standard treatments, HER2+ by HERACLES criteria | Trastuzumab+lapatinib | ORR 30% (95% CI 14 to 50) PFS 21 weeks (95% CI 16 to 32) |
Phase 1 dose expansion cohort of DS-8201a (Yoshino et al, 2018)127 |
I | 19 | HER2 IHC ≥1+ or HER2 amplified | DS-8201a | ORR 15.9% (3/19) DCR 82.4% (16/19) PFS 3.9 m (95% CI 2.1 to 8.3) |
MyPathway (Meric-Bernstam et al, 2019) |
II | 57 | ≥7 prior treatments, HER2+ by ISH, NGS or IHC | Trastuzumab+pertuzumab | ORR 32% (95% CI 20 to 45), PFS 2.9 m (95% CI 1.4 to 5.3) ORR 40%, PFS 5.3 m in KRAS wt (n=43) ORR 8%, PFS 1.4 m in KRAS mt (n=13) |
TRIUMPH (Nakamura et al, 2019)71 |
II | 18 | Tissue and/or ctDNA (Guardant360) confirmed RAS-wt and HER2-amplified mCRC | Trastuzumab+pertuzumab | Tissue-positive: ORR 35% (95% CI 14 to 62), PFS 4.0 m (95% CI 1.4 to 5.6) ctDNA-positive: ORR 33% (95% CI 12 to 62), PFS 4.0 m (95% CI 1.3 to 5.6) |
MOUNTAINEER (Strickler et al, 2019)72 |
II | 22 | HER2+ by NGS, ISH or IHC prior 5-FU, OXA, IRI, anti-VEGF | Trastuzumab+tucatinib | ORR 55% PFS 6.2 m (95% CI 3.5 to NE) |
HERACLES-B (Sartore-Bianchi et al, 2019)73 |
II | 30 | RAS/BRAF wt, HER2+ by HERACLES criteria, progression after 5-FU, OXA, IRI, anti-EGFR | Trastuzumab+T-DM1 | ORR 10% (95% CI 0 to 28) PFS 4.8 m (95% CI 3.6 to 5.8) |
ctDNA, circulating tumour DNA; EGFR, epidermal growth factor receptor; 5-FU, 5-fluorouracil; IHC, immunohistochemical staining; IRI, irinotecan; ISH, in situ hybridisation; m, months; mCRC, metastatic colorectal cancer; mt, mutant; NE, not estimated; NGS, next-generation sequencing; ORR, overall response rate; OXA, oxaliplatin; PFS, progression-free survival; VEGF, vascular endothelial growth factor; wt, wild-type.
RAS inhibitors: targeting the ‘undruggable’ genetic alterations
RAS mutations are notoriously undruggable targets due to their molecular structures with deeply seated hydrophobic pockets which are difficult to target using small molecules.75 Recent discoveries have allowed for the development of small-molecule inhibitors that selectively bind to a newly discovered allosteric regulatory site of the G12C mutant form of KRAS is underway.76 Preliminary data of a phase I study of AMG 510, the first-in-class KRAS G12C inhibitor, showed that approximately half of patients with non-small cell lung cancer achieved a partial response; however, an objective response was rare in the case of CRC.77 Because of the suboptimal activity in KRAS G12C mutant mCRC, which is a rare occurrence comprising approximately 4% of CRC cases,78 drug development in this field might be more challenging than expected. Recently, a preclinical study has suggested synergism between cetuximab and AMG 510 in KRAS G12C mutated CRC, implying that the combination could be explored as an alternative approach.79 Meanwhile, one can explore the upcoming results of different KRAS G12C inhibitors, as well as mutant-specific agents targeting more common variants such as KRAS G12D or G12V.80
Treating patients with rare genetic alterations
Other than BRAF or HER2, even rarer genetic alterations in mCRC are now being considered as actionable targets. ALK, ROS, NTRK, RET or FGFR2,3 fusions are rarely detected in mCRC, occurring in less than 1% of cases.81 82 For ALK, ROS1 and NTRK fusions, entrectinib may be useful as a tissue-agnostic therapeutic approach, although the response duration of patients with mCRC generally seems to be limited as compared with those in other disease subsets such as lung cancer or sarcoma.83 84 Larotrectinib, a selective TRK inhibitor, was also seen to be active in patients with NTRK fusion-positive solid tumours, 7% (4/55) of whom had mCRC.85 Therefore, patients with rare fusions can obtain clinical benefit from targeted agents, although the challenge lies in identifying these patients. DNA-level sequencing panels have limitations, especially for large genes such as NTRK2 or NTRK3, for which RNA-based sequencing assays are usually needed for reliable detection of fusions.86 However, it is not feasible in daily practice for all patients with mCRC to undergo RNA sequencing to detect rare genetic events. Although the clinical characteristics of patients with actionable fusions have been identified (elderly and female patients with right-sided and MSI-H tumours), it is uncertain whether limiting fusion testing of those patients would be an efficient method of screening.81 IHC could be a feasible alternative for fusion detection; these methods for detection of ALK and NTRK fusions showed varying rates of concordance with fluorescent ISH (FISH) and RNA-based sequencing in mCRC.87 88 However, we currently lack sufficient data on IHC for ROS1 or other fusions.
Ubiquitin ligase ring finger protein 43 (RNF43) is a negative regulator of the Wnt pathway. Somatic mutations in RNF43 occur in 6%–18% of CRC cases.89–91 Truncating mutations of RNF43 appear mutually exclusively with APC mutations, which is also associated with Wnt pathway activation. Fusions in RSPO2 or RSPO3 (secreted agonists of the Wnt-β-catenin pathway) are detected in approximately 10% of CRC cases and also avert APC mutations. These genetic alterations mainly overlap with MSI-H, which is a target for immune checkpoint inhibitors (ICIs). They may also be targeted by inhibiting porcupine, a protein involved in Wnt secretion.92 A high-throughput drug screening study using organoids showed that a colorectal tumour organoid with an RNF43 mutation was sensitive to IWP2, a small molecule porcupine inhibitor.93 Patient-derived xenografts of gastrointestinal cancer harbouring an RSPO2 fusion were also effectively treated by the porcupine inhibitor CGX1321.94 A recent phase I study of the first-in-class porcupine inhibitor WNT974 showed tumour regression in a case of appendiceal cancer with an RNF43 mutation.95 WNT974 is also being tested in patients with mCRC with BRAF V600 and RNF43 mutations or RSPO fusions, in combination with BRAF inhibitor and anti-EGFR to mitigate acquired resistance through the Wnt-β-catenin pathway96 in a phase II study (NCT02278133) and in combination with the ICI PDR001 (NCT01351103).
Immunotherapy for CRC: for and beyond MSI-H
MSI-H has been established as a reliable biomarker that predicts benefit from ICI in mCRC, as well as other types of cancers.97 Although MSI-H tumours comprises only 3%–5% of mCRC cases, they show high tumour mutational burden (TMB), high programmed-death ligand-1 (PD-L1) expression and high neoantigen load, making the tumour cells easily identifiable by immune system and sensitive to PD-1 or PD-L1 antagonists.98 Multicentre clinical trials with anti-PD-1 antibodies such as pembrolizumab and nivolumab have demonstrated favourable ORR (around 30%) and durable survival outcomes, with PFS at 12 months of 30%–50% and OS at 12 months of 70%–80% in pretreated patients with MSI-H mCRC.99 100 Phase III trials of these agents as compared with standard front-line regimens have been conducted and the results are awaited within the next few years: KEYNOTE 177 for pembrolizumab (NCT02563002) and CHECKMATE 8HW for nivolumab (NCT04008030). A combination of nivolumab and ipilimumab, an anti-cytotoxic T-lymphocyte–associated antigen (CTLA)−4 antibody, was also shown to be active in a pretreated population with MSI-H mCRC (ORR 55%, PFS at 12 months 71%)101 as well as untreated patients (ORR 60%, PFS at 12 months 77%).102 The activity of avelumab, an anti-PD-L1 antibody, is under investigation for MSI-H mCRC by our group, and the preliminary results have been promising in terms of ORR (30% in mCRC with MSI-H as defined by Bethesda panel or NGS).103 Recent translational studies have focused on the molecular heterogeneity within MSI-H tumours and its impact on clinical benefits from ICI treatment; higher TMB, especially insertion-deletion mutational load, has been known to be associated with the extent of response.104 105
Despite all the aforementioned developments, researchers are still struggling to identify valid immunotherapeutic options for microsatellite-stable (MSS) CRC, which shows no evidence of objective response to ICIs. A combination of MEK inhibitor with anti-PD-1 antibodies appeared to be active in preclinical studies106 and a phase I trial107; however, the IMblaze370 study, a phase III trial of atezolizumab and cobimetinib, showed no improvement in OS or PFS when compared with regorafenib.108 Another randomised trial of ICIs for mCRC, the MODUL study, compared bevacizumab, fluoropyrimidine+atezolizumab with bevacizumab and fluoropyrimidine. The study also failed to show benefits in terms of PFS, the primary endpoint, as well as overall response rate, disease control rate and duration of response.109 Although the TMB of MSS mCRC is much lower than that of MSI-H mCRC, the cause of intrinsic immune resistance of MSS mCRC cannot solely be explained by TMB because other types of cancers with similar TMB as MSS mCRC do respond to ICIs. Hence, the mechanism of de novo immune resistance of MSS CRC remains incompletely addressed. Recent translational studies have indicated that activated Wnt/β-cathenin signalling and transforming growth factor beta (TGF-β) signalling may cause T-cell exclusion and immune evasion in mCRC.110 111 In addition, a preclinical study showed that oncogenic KRAS mutations induce immunosuppression by downregulating interferon regulatory factor 2 (IRF2), resulting in activation of the CXCL3–CXCR2 axis and recruitment of myeloid-derived suppressive cells in CRC. This also suggests the possibility of combining ICI with CXCR2 inhibitor as a viable option to overcome the immunosuppressive microenvironment.112
Several clinical trials have suggested that combination strategies with ICI could work as therapeutics for MSS mCRC. The combination of ICI, tremelimumab (anti-CTLA-4 antibody) and durvalumab (anti-PD-L1) showed a slight improvement in OS (6.6 vs 4.1 months, stratified HR 0.72, 95% CI 0.54 to 097; p=0.07), but not PFS, in heavily treated patients with mCRC, mostly with MSS type.113 In addition, a phase I trial of regorafenib and nivolumab showed remarkable objective response (33%, 8/24) in patients with MSS mCRC, which spurred a phase III trial in refractory settings. Other early-phase studies of combination strategies, which showed anecdotal objective responses in MSS mCRC, are also intriguing and worth following for updated results, including BBI-608, a STAT3 inhibitor, combined with pembrolizumab114 as well as monalizumab, an anti-NKG2A (checkpoint of NK cell) antibody, with durvalumab+cetuximab.115
Conclusion
Even in the current era of precision medicine, there remain significant unmet needs for patients with mCRC. Most of the known actionable targets (BRAF, HER2, ALK, ROS1 and NTRK, as well as MSI-H for immunotherapy) are rarely present, and prevalent oncogenic genetic alterations such as APC, TP53 and RAS have been generally undruggable thus far. CMS has emerged as a prognostic or predictive marker of targeted therapy; however, substantial work is required for more robust classification of subtypes across different platforms and diverse clinical settings.116 Ongoing efforts to share and integrate clinical and genomic data could help in the discovery and validation of new actionable targets.117 Combinations of target blockades and ICIs could provide potential therapeutic opportunities for mCRC cases lacking druggable targets.118 Our ever-growing knowledge of tumour biology, including microenvironment and heterogeneity-related information, would also increase understanding of the precise mechanism of action and resistance of targeted agents and help refine the current strategies for mCRC treatment.
Acknowledgments
The authors thank the Scientific Publication Team at Asan Medical Center for their support for English editing.
Footnotes
Contributors: The first author drafted the manuscript under the guidance by the corresponding author, and the all authors revised and agreed on the final version of manuscript.
Funding: This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health and Welfare, Republic of Korea (grant no. HI18C2383).
Competing interests: None declared.
Patient consent for publication: Not required.
Provenance and peer review: Not commissioned; externally peer reviewed.
References
- 1.Wong MC, Ding H, Wang J, et al. Prevalence and risk factors of colorectal cancer in Asia. Intest Res 2019;17:317–29. 10.5217/ir.2019.00021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Siegel RL, Miller KD, Fedewa SA, et al. Colorectal cancer statistics, 2017. CA Cancer J Clin 2017;67:177–93. 10.3322/caac.21395 [DOI] [PubMed] [Google Scholar]
- 3.Primrose JN, Perera R, Gray A, et al. Effect of 3 to 5 years of scheduled CEA and CT follow-up to detect recurrence of colorectal cancer: the FACS randomized clinical trial. JAMA 2014;311:263–70. 10.1001/jama.2013.285718 [DOI] [PubMed] [Google Scholar]
- 4.Snyder RA, Hu C-Y, Cuddy A, et al. Association between intensity of posttreatment surveillance testing and detection of recurrence in patients with colorectal cancer. JAMA 2018;319:2104–15. 10.1001/jama.2018.5816 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004;350:2335–42. 10.1056/NEJMoa032691 [DOI] [PubMed] [Google Scholar]
- 6.Van Cutsem E, Tabernero J, Lakomy R, et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol 2012;30:3499–506. 10.1200/JCO.2012.42.8201 [DOI] [PubMed] [Google Scholar]
- 7.Tabernero J, Yoshino T, Cohn AL, 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 2015;16:499–508. 10.1016/S1470-2045(15)70127-0 [DOI] [PubMed] [Google Scholar]
- 8.Grothey A, Van Cutsem E, Sobrero A, et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 2013;381:303–12. 10.1016/S0140-6736(12)61900-X [DOI] [PubMed] [Google Scholar]
- 9.Hurwitz HI, Yi J, Ince W, et al. The clinical benefit of bevacizumab in metastatic colorectal cancer is independent of K-ras mutation status: analysis of a phase III study of bevacizumab with chemotherapy in previously untreated metastatic colorectal cancer. Oncologist 2009;14:22–8. 10.1634/theoncologist.2008-0213 [DOI] [PubMed] [Google Scholar]
- 10.Wirapati P, Pomella V, Vandenbosch B, et al. Velour trial biomarkers update: impact of RAS, BRAF, and sidedness on aflibercept activity. JCO 2017;35:3538–38. 10.1200/JCO.2017.35.15_suppl.3538 [DOI] [Google Scholar]
- 11.Price TJ, Bruhn MA, Lee CK, et al. Correlation of extended RAS and PIK3CA gene mutation status with outcomes from the phase III AGITG max study involving capecitabine alone or in combination with bevacizumab plus or minus mitomycin C in advanced colorectal cancer. Br J Cancer 2015;112:963–70. 10.1038/bjc.2015.37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lenz H-J, Ou F-S, Venook AP, et al. Impact of consensus molecular subtype on survival in patients with metastatic colorectal cancer: results from CALGB/SWOG 80405 (Alliance). J Clin Oncol 2019;37:1876–85. 10.1200/JCO.18.02258 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Innocenti F, Ou F-S, Qu X, et al. Mutational analysis of patients with colorectal cancer in CALGB/SWOG 80405 identifies new roles of microsatellite instability and tumor mutational burden for patient outcome. J Clin Oncol 2019;37:1217–27. 10.1200/JCO.18.01798 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Stintzing S, Wirapati P, Lenz H-J, et al. Consensus molecular subgroups (CMS) of colorectal cancer (CRC) and first-line efficacy of FOLFIRI plus cetuximab or bevacizumab in the FIRE3 (AIO KRK-0306) trial. JCO 2017;35:3510–10. 10.1200/JCO.2017.35.15_suppl.3510 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Mooi JK, Wirapati P, Asher R, et al. The prognostic impact of consensus molecular subtypes (CMS) and its predictive effects for bevacizumab benefit in metastatic colorectal cancer: molecular analysis of the AGITG max clinical trial. Ann Oncol 2018;29:2240–6. 10.1093/annonc/mdy410 [DOI] [PubMed] [Google Scholar]
- 16.Cui W, Li F, Yuan Q, et al. Role of VEGFA gene polymorphisms in colorectal cancer patients who treated with bevacizumab. Oncotarget 2017;8:105472–8. 10.18632/oncotarget.22295 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sibertin-Blanc C, Fabre A, Aparicio T, et al. Impact of genetic polymorphisms of VEGF pathway on the response to bevacizumab in metastatic colorectal cancer (mCRC): ancillary study of PRODIGE 9 trial. JCO 2016;34:3534–34. 10.1200/JCO.2016.34.15_suppl.3534 [DOI] [Google Scholar]
- 18.Cubillo A, Álvarez-Gallego R, Muñoz M, et al. Dynamic angiogenic switch as predictor of response to chemotherapy-bevacizumab in patients with metastatic colorectal cancer. Am J Clin Oncol 2019;42:56–9. 10.1097/COC.0000000000000474 [DOI] [PubMed] [Google Scholar]
- 19.Kopetz S, Hoff PM, Morris JS, et al. Phase II trial of infusional fluorouracil, irinotecan, and bevacizumab for metastatic colorectal cancer: efficacy and circulating angiogenic biomarkers associated with therapeutic resistance. J Clin Oncol 2010;28:453–9. 10.1200/JCO.2009.24.8252 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tabernero J, Hozak RR, Yoshino T, 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 2018;29:602–9. 10.1093/annonc/mdx767 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Nixon AB, Sibley A, Hatch AJ, et al. Blood-based biomarkers in patients (pts) with metastatic colorectal cancer (mCRC) treated with FOLFOX or FOLFIRI plus bevacizumab (Bev), cetuximab (Cetux), or Bev plus Cetux: results from CALGB 80405 (Alliance). JCO 2016;34:3597–97. 10.1200/JCO.2016.34.15_suppl.3597 [DOI] [Google Scholar]
- 22.Van Cutsem E, Köhne C-H, Hitre E, et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med 2009;360:1408–17. 10.1056/NEJMoa0805019 [DOI] [PubMed] [Google Scholar]
- 23.Douillard J-Y, Oliner KS, Siena S, et al. Panitumumab–FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med 2013;369:1023–34. 10.1056/NEJMoa1305275 [DOI] [PubMed] [Google Scholar]
- 24.Peeters M, Price TJ, Cervantes A, et al. Randomized phase III study of panitumumab with fluorouracil, leucovorin, and irinotecan (FOLFIRI) compared with FOLFIRI alone as second-line treatment in patients with metastatic colorectal cancer. J Clin Oncol 2010;28:4706–13. 10.1200/JCO.2009.27.6055 [DOI] [PubMed] [Google Scholar]
- 25.Zehir A, Benayed R, Shah RH, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med 2017;23:703–13. 10.1038/nm.4333 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.De Roock W, Claes B, Bernasconi D, et al. Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol 2010;11:753–62. 10.1016/S1470-2045(10)70130-3 [DOI] [PubMed] [Google Scholar]
- 27.Mao C, Yang ZY, Hu XF, et al. PIK3CA exon 20 mutations as a potential biomarker for resistance to anti-EGFR monoclonal antibodies in KRAS wild-type metastatic colorectal cancer: a systematic review and meta-analysis. Ann Oncol 2012;23:1518–25. 10.1093/annonc/mdr464 [DOI] [PubMed] [Google Scholar]
- 28.Sartore-Bianchi A, Martini M, Molinari F, et al. PIK3CA mutations in colorectal cancer are associated with clinical resistance to EGFR-targeted monoclonal antibodies. Cancer Res 2009;69:1851–7. 10.1158/0008-5472.CAN-08-2466 [DOI] [PubMed] [Google Scholar]
- 29.Jeong JH, Kim J, Hong YS, et al. HER2 amplification and cetuximab efficacy in patients with metastatic colorectal cancer harboring wild-type Ras and BRAF. Clin Colorectal Cancer 2017;16:e147–52. 10.1016/j.clcc.2017.01.005 [DOI] [PubMed] [Google Scholar]
- 30.Bokemeyer C, Van Cutsem E, Rougier P, 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 2012;48:1466–75. 10.1016/j.ejca.2012.02.057 [DOI] [PubMed] [Google Scholar]
- 31.Karapetis CS, Jonker D, Daneshmand M, et al. PIK3CA, BRAF, and PTEN status and benefit from cetuximab in the treatment of advanced colorectal cancer—results from NCIC CTG/AGITG CO.17. Clin Cancer Res 2014;20:744–53. 10.1158/1078-0432.CCR-13-0606 [DOI] [PubMed] [Google Scholar]
- 27.Van Cutsem E, Köhne C-H, Láng I, et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol 2011;29:2011–9. 10.1200/JCO.2010.33.5091 [DOI] [PubMed] [Google Scholar]
- 33.Kim TW, Peeters M, Thomas A, et al. Impact of emergent circulating tumor DNA RAS mutation in panitumumab-treated chemoresistant metastatic colorectal cancer. Clin Cancer Res 2018;24:5602–9. 10.1158/1078-0432.CCR-17-3377 [DOI] [PubMed] [Google Scholar]
- 34.Pietrantonio F, Vernieri C, Siravegna G, et al. Heterogeneity of acquired resistance to anti-EGFR monoclonal antibodies in patients with metastatic colorectal cancer. Clin Cancer Res 2017;23:2414–22. 10.1158/1078-0432.CCR-16-1863 [DOI] [PubMed] [Google Scholar]
- 35.Siena S, Sartore-Bianchi A, Garcia-Carbonero R, et al. Dynamic molecular analysis and clinical correlates of tumor evolution within a phase II trial of panitumumab-based therapy in metastatic colorectal cancer. Ann Oncol 2018;29:119–26. 10.1093/annonc/mdx504 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Arena S, Bellosillo B, Siravegna G, et al. Emergence of multiple EGFR extracellular mutations during cetuximab treatment in colorectal cancer. Clin Cancer Res 2015;21:2157–66. 10.1158/1078-0432.CCR-14-2821 [DOI] [PubMed] [Google Scholar]
- 37.Misale S, Di Nicolantonio F, Sartore-Bianchi A, et al. Resistance to anti-EGFR therapy in colorectal cancer: from heterogeneity to convergent evolution. Cancer Discov 2014;4:1269–80. 10.1158/2159-8290.CD-14-0462 [DOI] [PubMed] [Google Scholar]
- 122.Cremolini C, Rossini D, Dell'Aquila E, et al. Rechallenge for patients with RAS and BRAF wild-type metastatic colorectal cancer with acquired resistance to first-line cetuximab and irinotecan: a phase 2 single-arm clinical trial. JAMA Oncol 2019;5:343–50. 10.1001/jamaoncol.2018.5080 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Parseghian CM, Loree JM, Morris VK, et al. Anti-EGFR-resistant clones decay exponentially after progression: implications for anti-EGFR re-challenge. Ann Oncol 2019;30:243–9. 10.1093/annonc/mdy509 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Cremolini C, Morano F, Moretto R, et al. Negative hyper-selection of metastatic colorectal cancer patients for anti-EGFR monoclonal antibodies: the PRESSING case–control study. Ann Oncol 2017;28:3009–14. 10.1093/annonc/mdx546 [DOI] [PubMed] [Google Scholar]
- 41.Kim JH, Kang GH. Molecular and prognostic heterogeneity of microsatellite-unstable colorectal cancer. World J Gastroenterol 2014;20:4230–43. 10.3748/wjg.v20.i15.4230 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Arnold D, Lueza B, Douillard J-Y, 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 2017;28:1713–29. 10.1093/annonc/mdx175 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Tejpar S, Stintzing S, Ciardiello F, et al. Prognostic and predictive relevance of primary tumor location in patients with Ras wild-type metastatic colorectal cancer: retrospective analyses of the CRYSTAL and FIRE-3 trials. JAMA Oncol 2017;3:194–201. 10.1001/jamaoncol.2016.3797 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Venook AP, Ou F-S, Lenz H-J, et al. Primary (1°) tumor location as an independent prognostic marker from molecular features for overall survival (OS) in patients (pts) with metastatic colorectal cancer (mCRC): analysis of CALGB/SWOG 80405 (Alliance). JCO 2017;35:3503–03. 10.1200/JCO.2017.35.15_suppl.3503 [DOI] [Google Scholar]
- 45.Peeters M, Price T, Taieb J, et al. Relationships between tumour response and primary tumour location, and predictors of long-term survival, in patients with Ras wild-type metastatic colorectal cancer receiving first-line panitumumab therapy: retrospective analyses of the prime and peak clinical trials. Br J Cancer 2018;119:303–12. 10.1038/s41416-018-0165-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Laurent-Puig P, Grisoni M-L, Heinemann V, et al. Validation of miR-31-3p expression to predict cetuximab efficacy when used as first-line treatment in RAS wild-type metastatic colorectal cancer. Clin Cancer Res 2019;25:134–41. 10.1158/1078-0432.CCR-18-1324 [DOI] [PubMed] [Google Scholar]
- 47.Woolston A, Khan K, Spain G, et al. Genomic and transcriptomic determinants of therapy resistance and immune landscape evolution during anti-EGFR treatment in colorectal cancer. Cancer Cell 2019;36:35–50. 10.1016/j.ccell.2019.05.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Stintzing S, Modest DP, Rossius L, et al. FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab for metastatic colorectal cancer (FIRE-3): a post-hoc analysis of tumour dynamics in the final RAS wild-type subgroup of this randomised open-label phase 3 trial. Lancet Oncol 2016;17:1426–34. 10.1016/S1470-2045(16)30269-8 [DOI] [PubMed] [Google Scholar]
- 49.Naughton MJ, Schrag D, Venook AP, et al. Quality of life (QOL) and toxicity among patients in CALGB 80405. J Clin Oncol 2013:3611–11. [Google Scholar]
- 50.Morano F, Corallo S, Lonardi S, et al. Negative hyperselection of patients with RAS and BRAF wild-type metastatic colorectal cancer who received panitumumab-based maintenance therapy. J Clin Oncol 2019;37:Jco1901254:3099–110. 10.1200/JCO.19.01254 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kayhanian H, Goode E, Sclafani F, et al. Treatment and survival outcome of BRAF-mutated metastatic colorectal cancer: a retrospective matched case–control study. Clin Colorectal Cancer 2018;17:e69–76. 10.1016/j.clcc.2017.10.006 [DOI] [PubMed] [Google Scholar]
- 52.Venderbosch S, Nagtegaal ID, Maughan TS, 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 2014;20:5322–30. 10.1158/1078-0432.CCR-14-0332 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Prahallad A, Sun C, Huang S, et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 2012;483:100–3. 10.1038/nature10868 [DOI] [PubMed] [Google Scholar]
- 54.Tabernero J, Geel RV, Guren TK, 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). JCO 2016;34:3544 10.1200/JCO.2016.34.15_suppl.3544 [DOI] [Google Scholar]
- 55.Hong DS, Morris VK, El Osta B, et al. Phase Ib study of vemurafenib in combination with irinotecan and cetuximab in patients with metastatic colorectal cancer with BRAFV600E mutation. Cancer Discov 2016;6:1352–65. 10.1158/2159-8290.CD-16-0050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Corcoran RB, Atreya CE, Falchook GS, et al. Combined BRAF and MEK inhibition with dabrafenib and trametinib in BRAF V600-mutant colorectal cancer. J Clin Oncol 2015;33:4023–31. 10.1200/JCO.2015.63.2471 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kopetz S, McDonough SL, Lenz H-J, et al. Randomized trial of irinotecan and cetuximab with or without vemurafenib in BRAF-mutant metastatic colorectal cancer (SWOG S1406). JCO 2017;35:3505–05. 10.1200/JCO.2017.35.15_suppl.3505 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kopetz S, Grothey A, Yaeger R, et al. Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer. N Engl J Med 2019;381:1632–43. 10.1056/NEJMoa1908075 [DOI] [PubMed] [Google Scholar]
- 59.Yaeger R, Kotani D, Mondaca S, et al. Response to anti-EGFR therapy in patients with BRAF non-V600-mutant metastatic colorectal cancer. Clin Cancer Res 2019;25:7089–97. 10.1158/1078-0432.CCR-19-2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Jones JC, Renfro LA, Al-Shamsi HO, et al. Non-V600 BRAF mutations define a clinically distinct molecular subtype of metastatic colorectal cancerMutations Define a Clinically Distinct Molecular Subtype of Metastatic Colorectal Cancer. J Clin Oncol 2017;35:2624–30. 10.1200/JCO.2016.71.4394 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Yaeger R, Chatila WK, Lipsyc MD, et al. Clinical sequencing defines the genomic landscape of metastatic colorectal cancer. Cancer Cell 2018;33:125–36. 10.1016/j.ccell.2017.12.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Carter J, Tseng L-H, Zheng G, et al. Non-p.V600E BRAF mutations are common using a more sensitive and broad detection tool. Am J Clin Pathol 2015;144:620–8. 10.1309/AJCP85ATMJOZOUDJ [DOI] [PubMed] [Google Scholar]
- 63.Cremolini C, Di Bartolomeo M, Amatu A, et al. Braf codons 594 and 596 mutations identify a new molecular subtype of metastatic colorectal cancer at favorable prognosis. Ann Oncol 2015;26:2092–7. 10.1093/annonc/mdv290 [DOI] [PubMed] [Google Scholar]
- 64.Yao Z, Yaeger R, Rodrik-Outmezguine VS, et al. Tumours with class 3 BRAF mutants are sensitive to the inhibition of activated Ras. Nature 2017;548:234–8. 10.1038/nature23291 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Dankner M. Targeted therapy for colorectal cancers with Non-V600 BRAF mutations: perspectives for precision oncology. JCO Precis Oncol 2018;2:1–12. 10.1200/PO.18.00195 [DOI] [PubMed] [Google Scholar]
- 66.Bertotti A, Migliardi G, Galimi F, 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 2011;1:508–23. 10.1158/2159-8290.CD-11-0109 [DOI] [PubMed] [Google Scholar]
- 67.Richman SD, Southward K, Chambers P, 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 2016;238:562–70. 10.1002/path.4679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Leto SM, Sassi F, Catalano I, et al. Sustained inhibition of HER3 and EGFR is necessary to induce regression of HER2-amplified gastrointestinal carcinomas. Clin Cancer Res 2015;21:5519–31. 10.1158/1078-0432.CCR-14-3066 [DOI] [PubMed] [Google Scholar]
- 69.Sartore-Bianchi A, Trusolino L, Martino C, 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 2016;17:738–46. 10.1016/S1470-2045(16)00150-9 [DOI] [PubMed] [Google Scholar]
- 70.Siravegna G, Sartore-Bianchi A, Nagy RJ, et al. Plasma HER2 (ERBB2) copy number predicts response to HER2-targeted therapy in metastatic colorectal cancer. Clin Cancer Res 2019;25:3046–53. 10.1158/1078-0432.CCR-18-3389 [DOI] [PubMed] [Google Scholar]
- 71.Nakamura Y, Okamoto W, Kato T, et al. Triumph: primary efficacy of a phase II trial of trastuzumab (T) and pertuzumab (P) in patients (PTS) with metastatic colorectal cancer (mCRC) with HER2 (ErbB2) amplification (AMP) in tumour tissue or circulating tumour DNA (ctDNA): a GOZILA sub-study. Ann Oncol 2019;30:v199–200. 10.1093/annonc/mdz246.004 [DOI] [Google Scholar]
- 72.Strickler JH, Zemla T, Ou F-S, et al. Trastuzumab and tucatinib for the treatment of HER2 amplified metastatic colorectal cancer (mCRC): initial results from the MOUNTAINEER trial. Annal Oncol 2019;30:v200 10.1093/annonc/mdz246.005 [DOI] [Google Scholar]
- 73.Sartore-Bianchi A, Martino C, Lonardi S, et al. Phase II study of pertuzumab and trastuzumab-emtansine (T-DM1) in patients with HER2-positive metastatic colorectal cancer: the HERACLES-B (HER2 amplification for colo-rectaL cancer enhanced stratification, cohort B) trial. Annal Oncol 2019;30:v869–70. 10.1093/annonc/mdz394.024 [DOI] [Google Scholar]
- 74.Meric-Bernstam F, Johnson AM, Dumbrava EEI, et al. Advances in HER2-targeted therapy: novel agents and opportunities beyond breast and gastric cancer. Clin Cancer Res 2019;25:2033–41. 10.1158/1078-0432.CCR-18-2275 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Cox AD, Fesik SW, Kimmelman AC, et al. Drugging the undruggable RAS: mission possible? Nat Rev Drug Discov 2014;13:828–51. 10.1038/nrd4389 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ostrem JM, Peters U, Sos ML, et al. K-RAS(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature 2013;503:548–51. 10.1038/nature12796 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Govindan R, Fakih MG, Price TJ, et al. Phase I study of AMG 510, a novel molecule targeting KRAS G12C mutant solid tumours. Annal Oncol 2019;30:v163–4. 10.1093/annonc/mdz244.008 [DOI] [Google Scholar]
- 78.Imamura Y, Morikawa T, Liao X, et al. Specific mutations in KRAS codons 12 and 13, and patient prognosis in 1075 BRAF wild-type colorectal cancers. Clin Cancer Res 2012;18:4753–63. 10.1158/1078-0432.CCR-11-3210 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.McFall T, Trogdon M, Sisk-Hackworth L, et al. Inhibition of both mutant and wild-type Ras-GTP in KRAS G12C colorectal cancer through cotreatment with G12C and EGFR inhibitors. bioRxiv 2019;845263. [Google Scholar]
- 80.Porru M, Pompili L, Caruso C, et al. Targeting KRAS in metastatic colorectal cancer: current strategies and emerging opportunities. J Exp Clin Cancer Res 2018;37:57 10.1186/s13046-018-0719-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Pietrantonio F, Di Nicolantonio F, Schrock AB, et al. Alk, ROS1, and NTRK rearrangements in metastatic colorectal cancer. J Natl Cancer Inst 2017;109:1 10.1093/jnci/djx089 [DOI] [PubMed] [Google Scholar]
- 82.Clifton K, Raymond VM, Dasari A, et al. Actionable fusions in colorectal cancer using a cell-free circulating tumor DNA (ctDNA) assay. J Clin Oncol 2018;36:3507–07. 10.1200/JCO.2018.36.15_suppl.3507 [DOI] [Google Scholar]
- 83.Demetri GD, Paz-Ares L, Farago AF, et al. LBA4 efficacy and safety of entrectinib in patients with NTRK fusion-positive tumours: pooled analysis of STARTRK-2, STARTRK-1, and ALKA-372-001. Ann Oncol 2018;29 10.1093/annonc/mdy483.003 [DOI] [Google Scholar]
- 84.Siena S, Demetri G, Doebele R, et al. Entrectinib in NTRK-fusion positive gastrointestinal cancers: integrated analysis of patients enrolled in three trials (STARTRK-2, STARTRK-1, and ALKA-372-001). Ann Oncol 2019;30:iv134 10.1093/annonc/mdz154.023 [DOI] [Google Scholar]
- 85.Drilon A, Laetsch TW, Kummar S, et al. Efficacy of larotrectinib in Trk fusion-positive cancers in adults and children. N Engl J Med 2018;378:731–9. 10.1056/NEJMoa1714448 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Hsiao SJ, Zehir A, Sireci AN, et al. Detection of tumor NTRK gene fusions to identify patients who may benefit from tyrosine kinase (TRK) inhibitor therapy. J Mol Diagn 2019;21:553–71. 10.1016/j.jmoldx.2019.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Yakirevich E, Resnick MB, Mangray S, et al. Oncogenic ALK fusion in rare and aggressive subtype of colorectal adenocarcinoma as a potential therapeutic target. Clin Cancer Res 2016;22:3831–40. 10.1158/1078-0432.CCR-15-3000 [DOI] [PubMed] [Google Scholar]
- 88.Solomon JP, Linkov I, Rosado A, et al. NTRK fusion detection across multiple assays and 33,997 cases: diagnostic implications and pitfalls. Mod Pathol 2020;33:38-46 10.1038/s41379-019-0324-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Eto T, Miyake K, Nosho K, et al. Impact of loss-of-function mutations at the RNF43 locus on colorectal cancer development and progression. J Pathol 2018;245:445–55. 10.1002/path.5098 [DOI] [PubMed] [Google Scholar]
- 90.Giannakis M, Hodis E, Jasmine Mu X, et al. RNF43 is frequently mutated in colorectal and endometrial cancers. Nat Genet 2014;46:1264–6. 10.1038/ng.3127 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.AACR Project GENIE Consortium AACR Project GENIE: powering precision medicine through an international consortium. Cancer Discov 2017;7:818–31. 10.1158/2159-8290.CD-17-0151 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Dienstmann R, Vermeulen L, Guinney J, et al. Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer 2017;17:79–92. 10.1038/nrc.2016.126 [DOI] [PubMed] [Google Scholar]
- 93.van de Wetering M, Francies HE, Francis JM, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 2015;161:933–45. 10.1016/j.cell.2015.03.053 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Li C, Cao J, Zhang N, et al. Identification of RSPO2 fusion mutations and target therapy using a porcupine inhibitor. Sci Rep 2018;8:14244 10.1038/s41598-018-32652-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Janku F, Connolly R, LoRusso P, et al. Abstract C45: phase I study of WNT974, a first-in-class porcupine inhibitor, in advanced solid tumors. Molecular Cancer Therapeutics 2015;14:C45. [Google Scholar]
- 96.Chen G, Gao C, Gao X, et al. Wnt/β-catenin pathway activation mediates adaptive resistance to BRAF inhibition in colorectal cancer. Mol Cancer Ther 2018;17:806–13. 10.1158/1535-7163.MCT-17-0561 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409-413 10.1126/science.aan6733 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;372:2509–20. 10.1056/NEJMoa1500596 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Overman MJ, McDermott R, Leach JL, 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 2017;18:1182–91. 10.1016/S1470-2045(17)30422-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Le DT, Kim TW, Van Cutsem E, et al. Phase II open-label study of pembrolizumab in treatment-refractory, microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: KEYNOTE-164. J Clin Oncol 2020;38:Jco1902107 10.1200/JCO.19.02107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Overman MJ, Lonardi S, Wong KYM, et al. Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J Clin Oncol 2018;36:773–9. 10.1200/JCO.2017.76.9901 [DOI] [PubMed] [Google Scholar]
- 102.Lenz H-JJ, Van Cutsem E, Limon ML, et al. LBA18_PR 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 2018;29 10.1093/annonc/mdy424.019 [DOI] [Google Scholar]
- 103.A phase II study of avelumab monotherapy in patients with mismatch repair deficient or POLE mutated metastatic or unresectable colorectal cancer (KM-01, NCT03150706). 12th Annual Meeting of Korean Society of Medical Oncolgy & 2019 Interenational Conference 2019. Seoul.
- 104.Schrock AB, Ouyang C, Sandhu J, et al. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. Ann Oncol 2019;30:1096–103. 10.1093/annonc/mdz134 [DOI] [PubMed] [Google Scholar]
- 105.Mandal R, Samstein RM, Lee K-W, et al. Genetic diversity of tumors with mismatch repair deficiency influences anti–PD-1 immunotherapy response. Science 2019;364:485–91. 10.1126/science.aau0447 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ebert PJR, Cheung J, Yang Y, et al. MAP kinase inhibition promotes T cell and anti-tumor activity in combination with PD-L1 checkpoint blockade. Immunity 2016;44:609–21. 10.1016/j.immuni.2016.01.024 [DOI] [PubMed] [Google Scholar]
- 107.Hellmann MD, Kim T-W, Lee CB, et al. Phase Ib study of atezolizumab combined with cobimetinib in patients with solid tumors. Ann Oncol 2019;30:1134–42. 10.1093/annonc/mdz113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Eng C, Kim TW, Bendell J, 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 2019;20:849–61. 10.1016/S1470-2045(19)30027-0 [DOI] [PubMed] [Google Scholar]
- 109.Grothey A, Tabernero J, Arnold D, 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 2018;29:viii714–5. 10.1093/annonc/mdy424.020 [DOI] [Google Scholar]
- 110.Grasso CS, Giannakis M, Wells DK, et al. Genetic mechanisms of immune evasion in colorectal cancer. Cancer Discov 2018;8:730–49. 10.1158/2159-8290.CD-17-1327 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Tauriello DVF, Palomo-Ponce S, Stork D, et al. TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018;554:538–43. 10.1038/nature25492 [DOI] [PubMed] [Google Scholar]
- 112.Liao W, Overman MJ, Boutin AT, et al. KRAS-IRF2 axis drives immune suppression and immune therapy resistance in colorectal cancer. Cancer Cell 2019;35:559–72. 10.1016/j.ccell.2019.02.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Chen EX, Jonker DJ, Kennecke HF, et al. Cctg CO.26 trial: a phase II randomized study of durvalumab (D) plus tremelimumab (T) and best supportive care (BSC) versus BSC alone in patients (PTS) with advanced refractory colorectal carcinoma (rCRC). J Clin Oncol 2019;37:481–81. 10.1200/JCO.2019.37.4_suppl.48130620669 [DOI] [Google Scholar]
- 114.Shinozaki E, Kawazoe A, Kuboki Y, et al. Multicenter phase I/II trial of BBI608 and pembrolizumab combination in patients with metastatic colorectal cancer (SCOOP study): EPOC1503. J Clin Oncol 2018;36:3530–30. 10.1200/JCO.2018.36.15_suppl.3530 [DOI] [Google Scholar]
- 115.Segal NH, Naidoo J, Curigliano G, et al. First-in-human dose escalation of monalizumab plus durvalumab, with expansion in patients with metastatic microsatellite-stable colorectal cancer. J Clin Oncol 2018;36:3540–40. 10.1200/JCO.2018.36.15_suppl.3540 [DOI] [Google Scholar]
- 116.Fontana E, Eason K, Cervantes A, et al. Context matters—consensus molecular subtypes of colorectal cancer as biomarkers for clinical trials. Ann Oncol 2019;30:520–7. 10.1093/annonc/mdz052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.AACR Project Genie: powering precision medicine through an international consortium 2017. [DOI] [PMC free article] [PubMed]
- 118.Ciardiello D, Vitiello PP, Cardone C, et al. Immunotherapy of colorectal cancer: challenges for therapeutic efficacy. Cancer Treat Rev 2019;76:22–32. 10.1016/j.ctrv.2019.04.003 [DOI] [PubMed] [Google Scholar]
- 119.Tabernero J, Lenz H-J, Siena S, et al. Analysis of circulating DNA and protein biomarkers to predict the clinical activity of regorafenib and assess prognosis in patients with metastatic colorectal cancer: a retrospective, exploratory analysis of the CORRECT trial. Lancet Oncol 2015;16:937–48. 10.1016/S1470-2045(15)00138-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Kim TW, Elme A, Park JO, et al. Final analysis of outcomes and RAS/BRAF status in a randomized phase 3 study of panitumumab and best supportive care in chemorefractory wild type KRAS metastatic colorectal cancer. Clin Colorectal Cancer 2018;17:206–14. 10.1016/j.clcc.2018.03.008 [DOI] [PubMed] [Google Scholar]
- 121.Berger MD, Stintzing S, Yang D, et al. Genetic variations within the vitamin C transporter genes to predict outcome in metastatic colorectal cancer patients treated with first-line FOLFIRI and bevacizumab: data from FIRE-3 trial. JCO 2017;35:11507–07. 10.1200/JCO.2017.35.15_suppl.11507 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Heinemann V, Kraemer N, Buchner H, 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). JCO 2018;36:3591–91. 10.1200/JCO.2018.36.15_suppl.3591 [DOI] [Google Scholar]
- 123.Miller-Phillips L, Heinemann V, Stahler A, et al. Association of microRNA-21 with efficacy of cetuximab in RAS wild-type patients in the FIRE-3 study (AIO KRK-0306) and microRNA-21’s influence on gene expression in the EGFR signaling pathway. JCO 2019;37:3593–93. 10.1200/JCO.2019.37.15_suppl.3593 [DOI] [Google Scholar]
- 124.Stintzing S, Stremitzer S, Heinemann V, et al. Amphiregulin (AREG) SNP rs161511 to predict cetuximab efficacy independent of AREG mRNA levels: data from FIRE3 (AIO KRK-0306). JCO 2014;32:3521–21. 10.1200/jco.2014.32.15_suppl.3521 [DOI] [Google Scholar]
- 125.Corcoran RB, André T, Atreya CE, et al. Combined BRAF, EGFR, and MEK inhibition in patients with BRAFV600E-mutant colorectal cancer. Cancer Discov 2018;8:428–43. 10.1158/2159-8290.CD-17-1226 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.van Geel RMJM, Tabernero J, Elez E, et al. A phase Ib dose-escalation study of encorafenib and cetuximab with or without alpelisib in metastatic BRAF-mutant colorectal cancer. Cancer Discov 2017;7:610–9. 10.1158/2159-8290.CD-16-0795 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Yoshino T, Iwata H, Tamura K, et al. Updated results of phase I study of trastuzumab deruxtecan (DS-8201a) in HER2-expressing advanced colorectal cancer. Annal Oncol 2018;29:viii188–204. 10.1093/annonc/mdy281.109 [DOI] [Google Scholar]