IJGII Inernational Journal of Gastrointestinal Intervention

pISSN 2636-0004 eISSN 2636-0012
ESCI
scopus

Article

Review Article

Int J Gastrointest Interv 2024; 13(3): 74-81

Published online July 31, 2024 https://doi.org/10.18528/ijgii240030

Copyright © International Journal of Gastrointestinal Intervention.

Immune check point inhibitors in BRAF mutated metastatic colorectal cancer: A review

Alfredo Colombo1,* , Stefano Cordio2, and Concetta Maria Porretto1

1Oncology Unit, Casa di Cura Macchiarella S.p.A., Palermo, Italy
2Department of Oncology, Ospedale Maria Paternò Arezzo di Ragusa, Ragusa, Italy

Correspondence to:*Oncology Unit, Casa di Cura Macchiarella S.p.A., Viale Regina Margherita 25, Palermo 90138, Italy.
E-mail address: alfredocolombo63@gmail.com (A. Colombo).

Received: May 27, 2024; Accepted: June 8, 2024

This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/bync/4.0) which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Metastatic colorectal cancers with BRAF mutation are a class of tumors that have a poor prognosis compared to wild type patients. Even if this group has found some hope in targeted therapy with multi-targeted blockade of the mitogen-activated protein kinase (MAPK) signalling pathway, more work must be done to increase treatment efficacy, particularly for the microsatellite stability/DNA proficient mismatch repair (MSS/pMMR) subtype. Patients with BRAF mutant colorectal cancer and high microsatellite instability/DNA deficient mismatch repair (MSI-H/dMMR) are considered to have a high tumor mutation load and a lot of neoantigen, which makes immunotherapy likely to be effective. It is generally accepted that MSS/pMMR colorectal cancer is an immunologically “cold” tumor that is resistant to immunotherapy. Then, patients with BRAF mutant colorectal cancer appear to find hope with targeted therapy paired with immune checkpoint blockade therapy. The clinical effectiveness and developing new approaches of immune checkpoint blockade therapy for MSI-H/dMMR and MSS/pMMR BRAF mutant metastatic colorectal cancer are reviewed in this paper. We also address possible biomarkers in the tumor immune microenvironment for predicting immunotherapeutic response in BRAF mutant colorectal cancer.

Keywords: Colon, Immunotherapy, Mutation, Neoplasms, Proto-oncogene proteins B-raf

Colorectal cancer (CRC) is third most prevalent malignant tumor and second most deadly cancer worldwide.1 Economic growth and the diagnosis of CRC in younger may be attributed to hereditary, lifestyle, food, and environmental variables.1 Even if screening programmes successfully minimise the death and morbidity from CRC,2 between 20 and 35 percent of individuals with CRC have distant metastases at the time of diagnosis.3,4 Five years of survival is around 13% for patients with metastatic CRC (mCRC).5 The National Comprehensive Cancer Network (NCCN) Guidelines for colon cancer promote molecular analysis, including oncogenic alterations for KRAS/NRAS/BRAF and mismatch repair (MMR) status, for mCRC before beginning a treatment.6 Only 4%–8% of mCRC have BRAF mutation V600E7 which is a punctiform substitution, from valine to glutamine in codon 600.8 The median overall survival (mOS) of BRAF V600E mutant mCRC is fewer than 15 months.9 The milestone of treatment for CRC is chemotherapy with or without targeted agents. The standard first-line chemotherapy for patients with BRAF mutations mCRC is fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI) with or without bevacizumab in patients fit or doublet of chemotherapy in association with bevacizumab, even if it was observed a poor effectiveness with progression free survival (PFS) and overall survival (OS) very low.10 Opposite to melanoma, patients with BRAF mutant CRC have not shown therapeutic benefit from BRAF inhibitors alone8 because the activation of alternate signalling pathways—such as PI3K/Akt,1114 and Wnt/β15—plays a driving role. In the last years many clinical trials with multi-target combination have been conducted. In previously treated BRAF mutant mCRC, combination targeted therapies involving BRAF inhibitor and Epidermal Growht factor receptore (EGFR) inhibitor produced positive results.1618 Moreover, a combined targeted therapy (encorafenib, binimetinib, and plus cetuximab) is being studied in the first line BRAF V600E mut. mCRC.19 Immune checkpoint inhibitors (ICIs) have been evaluated in CRC.20 ICIs generate a substantial immune system activation and an effective anticancer immune response.21 The most used are monoclonal antibodies against cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death 1 (PD-1), that have been approved for many cancer types in clinical practice.22 Studies already demonstrated that in mCRC with high microsatellite instability (MSI-H) which have a higher tumor mutation burden, they respond better to ICIs.23 Advanced CRC commonly exhibits BRAF mutations associated to MSI-H/DNA deficient MMR (dMMR).24 Thirty percent of BRAF mutant mCRC, are MSI-H. Treatment with ICIs has recently shown to be very helpful for achieving a long response in MSI-H/dMMR mCRC.25 Instead, it is believed that ICIs monotherapy is not very effective in microsatellite instability (MSS) mCRC with BRAF mutation.26 For this group of patients, ICIs are therefore being investigated in combination with other anticancer drugs, including BRAF inhibitors.27 We address in this study the immunological environment of BRAF mutant CRC.

Using the terms colon, cancer, mutations, immunotherapy, BRAF, we searched PubMed (www.ncbi.nlm.nih.gov/pubmed) for full-text papers from 2017 to May 31, 2023.

Each of the found full-text articles was thoroughly read. We also looked over every abstract that was presented at international conferences between January 2020 and January 2024.

Two of the multiple different mechanisms that lead to CRC are the serrated and traditional adenoma-carcinoma pathways.28 Many serrated premalignant lesions result from BRAF mutations, which show as an early event in the serrated pathway.29,30 BRAF is essential to cell proliferation, differentiation, angiogenesis, apoptosis, and metastasis.31 Because BRAF kinase is constitutively phosphorylated when mutations occur, tumor growth is caused by the prolonged stimulation of MAPK pathway signalling.11,32

Molecular characteristics and signalling pathways allowed the differentiation of three BRAF mutant types.32 The serrated pathway is closely related to class 1, which has the highest incidence but the worst prognosis among these. By contrast, class 3 CRCs are more likely to have a similar prognosis to wild-type CRCs.33

Chromosomal instability (CIN), the CpG pathway of methylation phenotype (CIMP), and microsatellite instability (MSI) are at least three separate molecular pathways that drive CRC.34,35 Advanced CRC is frequently associated with BRAF mutations, particularly the BRAF V600E mutation,32,36,37 which may be linked to hypermethylation of the MLH1 promoter and the CpG island methylator phenotype.38 The microsatellites belong to a large group of DNA motifs which have a length of 1–6 bp, harbouring high levels of sequence variation, common to all genomes.39 Mostly observed at microsatellites, the status of dMMR causes many mutations or gene silencing.40,41 Less than 10% of MSS CRC, include BRAF mutations.24,37,42 Classification of BRAF mutated mCRC would be useful in predicting which treatment regimens to choose to improve patient outcomes.43 Consensus molecular subtypes (CMS) state that BRAF mutant CRC mostly (up to 70%) belongs to CMS1 (MSI Immune), which has higher immune infiltration and improved OS.44,45 Moreover, based on gene expression in CRC, subtypes of BRAF V600E mutation were proposed, independent of MSI status, PI3K mutation, gender, and sidedness. Through pathways including stimulation of IL2/STAT5, tumor necrosis factorα signalling, IL6/JAK/STAT3, and allograft rejection, BRAF V600E Mutation 1 (BM1) had an overall stronger immunological profile than BRAF V600E Mutation 2 (BM2). Better response rates, median progression-free survival (mPFS), and mOS were shown for BM1 in patients receiving EGFR inhibitors (dabrafenib, trametinib, and panitumumab) in addition to BRAF plus mitogen-activated protein kinase kinase (MEK).46,47

MSI-H/dMMR subgroup

Recently there has been considerable progress in the use of ICIs in MSI-H CRC. Pembrolizumab, a PD-1 inhibitor, was tested in the phase II Keynote 164 trial (NCT02460198) in MSI-H/dMMR mCRC patients who had previously received systemic therapy. Pembrolizumab monotherapy showed response in five (55%) and one (25%) BRAF mutant patients in cohorts A (patients pre-treated with at least two standard lines of chemotherapy) and B (patients pre-treated with one line of systemic chemotherapy).48 To treat MSI-H/dMMR mCRC, nivolumab, a PD-1 inhibitor, is being investigated in the phase II, multicenter, open-label CheckMate 142 trial (NCT02060188) either alone or in combination with Ipilimumab. Comparing the investigator-assessed objective response rate (ORR) of 31.1% (95% confidence interval [CI], 20.8%–42.9%) in 74 patients with MSI-H mCRC who had undergone at least one prior line of therapy the ORR, in BRAF mutated subgroup, was 25%.49 Also, was studied in CheckMate 142, nivolumab with low-dose ipilimumab, a CTLA-4 inhibitor. The treatment gave an ORR of 65% (95% CI, 55%–73%) and a disease control rate (DCR) of 81% (95% CI, 72%–87%) in the group of previously treated patients.50 In subgroups with BRAF V600E mutation, the ORR was 70%,50 the safety profile was handleable. Complementing the previously reported findings, a meta-analysis revealed that there was no statistically significant difference in the ORR between BRAF wild-type mCRC and BRAF mutant mCRC with MSI-H/dMMR (odds ratio [OR] 1.04; 95% CI, 0.48–2.25).9 In the later-line therapy of MSI-H/dMMR mCRC with BRAF mutation, ICIs may offer a potential clinical response advantage. A significant response was also shown in the first-line cohort in CheckMate 142 by nivolumab in combination with a low-dose ipilimumab regimen. Patients with BRAF mutations clearly benefited from sustained response with a 24-month PFS rate of 76.5% and ORR (76% per investigator; 82% per blinded indipedent central review).51 Pembrolizumab monotherapy vs. chemotherapy in treatment-naïve MSI-H/dMMR mCRC patients is the focus of the phase III, multicenter, open-label, international trial Keynote 177.25,52 While a lack of benefit in mOS between the study arms (not available [NA] vs. 36.7 months, respectively, pembrolizumab monotherapy vs. chemotherapy, hazard ratio [HR] 0.74; 95% CI, 0.53–1.03, P = 0.036) was found due to falling short of the preset statistical boundary, a final analysis reported a significant improvement in mPFS (16.5 months vs. 8.2 months, respectively, pembrolizumab monotherapy vs. chemotherapy, HR 0.59; 95% CI, 0.45–0.79, P = 0.0002). In a subgroup analysis in tumors characterised by BRAF V600E mutation, no OS benefit was seen (HR 0.72; 95% CI, 0.35–1.47) even though data analysis revealed a statistically significant benefit in PFS (HR 0.48; 95% CI, 0.27–0.86). With 60% of patients getting anti-PD-1 or anti-program death ligand 1 (PD-L1) therapy following first-line chemotherapy, the crossover design may be responsible for the lack of appreciable OS benefit. Moreover, the health-related quality of life improved and there were fewer adverse events with pembrolizumab monotherapy.52,53 In the MSI-H/dMMR mCRC population, the clinical benefit of the BRAF mutant subgroup did not differ significantly from that of the non-BRAF mutation population, according to the scant clinical trial data that are now available, in first-line or later-line treatment. The effectiveness of ICIs with conventional therapies must be compared in more head-to-head trials. The BRAF V600E mutation was, however, recently linked to a deleterious impact on the ICIs response in MSI-H mCRC patients in a real-world retrospective investigation.54 Patients with BRAF V600E mutation showed lower ORR (44.4% vs. 74.2% respectively; P = 0.120) but no statistically significant, and the 12- and 24-month PFS rates were much lower than those of BRAF wild-type patients (12-months: 40.0% vs. 73.3%, P < 0.001; 24-months: 26.7% vs. 73.3%, P < 0.001). Still, there were not many BRAF V600E individuals in this subgroup analysis, and the treatment timing was not clear. Large-scale trials in the subgroup with BRAF mutant mCRC are therefore still required to confirm the effectiveness of immunotherapy alone. A fraction of MSI-H/dMMR metastatic patients respond poorly to ICIs, which may be because of acquired or intrinsic resistance. There is still needed to investigate new biomarkers for screening appropriate populations and workable treatment plans. As such, combined therapy approaches have been suggested to overcome immunotherapy resistance. In the first-line treatment of MSI-H/dMMR mCRC with BRAF mutation, the open-label, multicenter SEAMARK study is already underway and compares the efficacy of pembrolizumab alone or with cetuximab (an EGFR inhibitor) and encorafenib (a BRAF inhibitor).

In conclusion, immunotherapy-treated BRAF-mutated MSI-H patients may also have a good prognosis and a long-lasting response, albeit some of them show treatment resistance.

MSS subgroup

While Keynote 016 showed a significant advancement in developing an immunotherapeutic approach for MSI-H/dMMR mCRC, treating MSS/pMMR mCRC with immune checkpoint inhibition alone showed less benefit.5,26,55,56 Stated otherwise, intrinsic resistance by many mechanisms renders MSS/pMMR mCRC patients resistant to ICIs monotherapy.57 Nevertheless, logical combinations based on ICIs still need to be investigated further for MSS/pMMR mCRC by choosing appropriate populations with predictive biomarkers and getting over intrinsic resistance, particularly for patients with BRAF mutation who have a bad prognosis and no efficient treatment plan.

Combining ICIs and anti VEGF

A step in growth of a lot of solid tumors, is angiogenesis; proangiogenic factors are markedly increased in the BRAF V600E mutant tissue.58 Significantly involved in vasculogenesis and angiogenesis, vascular endothelial growth factor (VEGF) causes aberrant and leaky blood vessels in cancer.59 Preclinical studies revealed that VEGF might directly mediate immune cells, including regulatory T cells, dendritic cells, tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs), thereby exerting an immunosuppressive impact.60,61 Hence, by normalising the tumor vasculature and changing the immunological milieu, blocking the VEGF signalling pathway may improve the effectiveness of ICIs.

A precedent for the combination therapy for MSS/pMMR mCRC was established with the REGONIVO trial, a phase Ib/II study, investigate the safety and efficacy of nivolumab in combination with multikinase inhibitor regorafenib (a VEGF receptor [VEGFR]2 inhibitor) in patients with MSS gastrointestinal cancers.62 In the mCRC cohort, 25 patients, the mPFS was 7.5 months and the ORR was 36%. Study data on patients with BRAF mutant mCRC are currently scarce, nonetheless. Preliminary findings from the NIVACOR trial, a phase II trial assessing the effectiveness of FOLFOXIRI in combination with bevacizumab and nivolumab in advanced CRC with a mutation in RAS/BRAF, were published.63 The ORR in 52 MSS patients was 78.9%, and the median DOR was 7.59 (95% CI, 6.21–11.43) months. Within the MSS patient subgroup, the mPFS was 9.8 (95% CI, 8.18–15.24) months and the DCR was 96.2%. It is still unknown how the results from advanced CRC with BRAF mutations will turn out. In the meanwhile, a case study revealed that, with a combination therapy of nivolumab and VEGFR inhibitor (bevacizumab, an anti-angiogenic agent), an MSS/pMMR mCRC patient with BRAF V600E mutation had a PFS of more than 17 months.64

ICIs with blockade of the MAPK signalling pathway

Growing research indicates that via altering the tumor microenvironment (TME) and anti-tumor immunomodulation, inhibiting the MAPK pathway may work together with immunotherapy.65 Clinical improvement is obtained in BRAF mutant melanoma by ICI therapy combined with MAPK pathway inhibition.6669 Studies on the simultaneous inhibition of the immune checkpoint and MAPK pathway have also been conducted in mCRCs with BRAF mutations. A phase II trial assessed patients with BRAF V600E mutated mCRC treated with dabrafenib, a BRAF inhibitor, trametinib, a MEK inhibitor, and PDR001, also known as spartalizumab.70,71 Seventy-three BRAF V600E mutated mCRCs were enrolled; the ORR and DCR were 24.3% and 70.3%, respectively. In 28 MSS mCRC patients who had never treatment with ICIs or BRAF inhibitors, the mPFS was 5.6 months, and the ORR and DCR were 25% (7/28) and 75% (21/28), respectively. Comparing the regimen of dabrafenib plus trametinib to the combination of the PD-1 inhibitor, a good effectiveness was shown.72 Compared to pre-treatment biopsies, T cell and other immune cell populations infiltrated tumor biopsies more after therapy.70 In MSS, BRAF V600E mutant, unresectable or mCRC patients, the phase I/II trial was conducted for evaluate the dosage, safety, and efficacy of encorafenib (a BRAF inhibitor) combination with cetuximab (an EGFR inhibitor) and nivolumab.73 Results indicated a strong efficacy with ORRs of 96% (95% CI, 78%–100%) and 48% (95% CI, 27%–69%). Of those who respond, the median response time is 7.7 months (95% CI, 4.5–NA). Noteworthy, mPFS and mOS were 7.4 months (95% CI, 7.7–NA) and 15.1 months (95% CI, 5.6–NA), respectively, demonstrating a better efficacy than targeted treatment. Regarding safety profile, patients did well with the combination; 19% (5/26) of them had grade 3 or 4 adverse events. Currently underway, a second randomised phase II trial investigated the benefits of adding nivolumab, in previously treated MSS mCRC patients with BRAF V600E mutation, with encorafenib and cetuximab (NCT05308446). Ultimately, the small clinical trial suggests that patients with BRAF-mutated MSS/pMMR mCRC may benefit from a combination of MAPK inhibition and ICIs. ICI-containing combination therapy has been tested in a number of studies recently in BRAF mutant mCRC (Table 1).

Table 1 . Clinical Trials Exploring Combination Therapies Based on ICIs in BRAF Mutant Metastatic Colorectal Cancer.

NCT numberTitlePhaseStatusInterventionICIs related armsCondition
NCT05019534A Phase I study on tolerance and safety of vemurafenib film-coated tablets, cetuximab solution for infusion and camrelizumab protocol (VCC) in the after line therapy of BRAF V600E mutation/MSS metastatic colorectal cancerPhase IRecrutingDrug: vemurafenib; cetuximab; camrelizumabSingle arm: vemurafenib, cetuximab combined with camrelizumabMicrosatellite stable, BRAF V600E-mutated, metastatic colorectal cancer
NCT05308446Randomized Phase II trial of encorafenib and cetuximab with or without nivolumab (NSC #748,726) for patients with previously treated, microsatellite stable, BRAF V600E metastatic and/or unresectable colorectal cancerPhase IIRecrutingDrug: cetuximab; encorafenib; nivolumabExperimental (arm I): encorafenib, cetuximab, nivolumab; Active comparator (arm II): encorafenib, cetuximabMicrosatellite stable, BRAF V600E metastatic and/or unresectable colorectal cancer
NCT04044430Phase I/II trial of encorafenib, binimetinib, and nivolumab in microsatellite stable BRAF V600E metastatic colorectal cancerPhase I/IIActive, not recrutingDrug: binimetinib; encorafenib; nivolumabSingle arm: encorafenib, binimetinib, nivolumabMicrosatellite stable, BRAF V600E metastatic colorectal cancer
NCT04653480Surufatinib and toripalimab combined with chemotherapy for second-line treatment of advanced RAS/BRAF mutant and microsatellite stable colorectal cancerPhase IIRecruitingDrug: surufatinib; toripalimab; chemotherapySingle arm: surufatinib, toripalimab, and chemotherapyMicrosatellite stable, RAS/BRAF mutant advanced colorectal cancer
NCT04294160A Phase Ib, multicenter, open-label dose escalation and expansion platform study of select drug combinations in adult patients with advanced or metastatic BRAF V600 colorectal cancerPhase IbRecruitingDrug: dabrafenib; LTT462; spartalizumab; tislelizumabBackbone arm 1: dabrafenib + LTT462; triplet arm 4: dabrafenib + LTT462 + spartalizumab; triplet arm 6: dabrafenib + LTT462 + tislelizumabAdvanced or metastatic BRAF V600 colorectal cancer

NCT, National Clinical Trial; ICIs, immune checkpoint inhibitors; MSS, microsatellite instability..


Cells of the immune system

A dynamic system of cellular including immune cells, fibroblasts, stromal cells and extracellular matrix is known as TME, a crucial component that influences the effectiveness of immunotherapy.74,75 The immune microenvironment of tumor (TIME) is composed of immune cells (CD8+T cells, CD4+T cells), MDSCs, anti-inflammatory macrophages, natural killer (NK) cells and associated non-cellular components and is essential to the development of tumors and their evasion of immune therapy.76 Improved ICI efficacy in BRAF mutant mCRC patients depends on understanding the properties of TIME and managing the role of immunosuppressive variables in the TME.

Tumor-infiltrating lymphocytes (TILs)

TILs include CD8+ T cells, CD4+ helper T cells, regulatory T cells, B cells, and are essential for host immune response to tumor cells.77,78 It is proposed that the density of TILs may perform as a marker to some degree for predicting prognosis in a range of malignancies and response to ICI therapy.77,79 Unfortunately, there are currently no data on the predictive utility of TIL density in evaluating the effectiveness of immunotherapy in BRAF mutant CRC; yet a higher density of TILs was shown to be linked with a positive prognosis independent of BRAF mutation status.80,81 We addressed the limited information and contradicting findings about the density of TILs in BRAF mutant CRC. BRAF mutation status was not significantly connected with the density of CD8+ intratumor cell-infiltrating lymphocytes in a study included 24 patients with BRAF mutant CRC (P = 0.090).82 In another work, CD45RO+ T cells were greater in 114 BRAF mutant CRCs (P = 0.0006), but the density of CD8+ and CD3+, as well as FOXP3+ T cells, was not significantly correlated with BRAF mutation.83 TIL density in the periphery of tumor infiltration has been linked, in the context of anti-PD1 therapy, to an anti-PD1 response more closely than in those with central infiltration. Infiltrative margins of BRAF-mutated advanced colorectal tumors had a significantly higher density of FOXP3+ T cells (P < 0.001), but the centre of these tumors had lower densities of CD4+ and FOXP3+ T cells (P = 0.011 and P < 0.001, respectively).84

A comprehensively assessment of immunological environment in CRC BRAF mutated, was conducted by Cen et al.85 The expression of CD8+ T cells was considerably higher in BRAF mutant colon cancer patients than in wild-type ones (P < 0.001) in this study included 43 BRAF mutant colon cancer patients.85 The fraction of CD8+ T cells was considerably higher in BRAF-mutated colon cancer tissues than in BRAF wild-type colon cancer tissues (P < 0.01) according to data from The Cancer Genome Atlas (TCGA) datasets, which included 59 BRAF-mutated and 337 BRAF wild-type colon cancer patients.85 By contrast, there were no appreciable variations in the percentage of CD8+ T cells between groups according to the Gene Expression Omnibus (GEO) datasets (51 BRAF-mutated, 441 BRAF wild-type).85 Furthermore, exhibiting this paradox are CD4+ T cells. The researchers further found that colon tumors harbouring a BRAF mutation had reduced tumor purity (P = 0.0003), a greater stromal score (P = 0.02), an immune score (P < 0.0001), and an Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) score (P = 0.0001). The TILs level may not be sufficient to completely characterise the complicated mechanism that is TIME of BRAF mutant CRC. Thus, this problem must be considered together with other elements like CMS classification and MSI status. Digiacomo et al86 assessed the TILs in 22 MSI and 37 MSS BRAF-mutated mCRC and found variations in TILs between distinct subtypes of the disease. Results indicated that in MSI compared to MSS tumors, CD8+ and CD3+ lymphocytes, both intratumoral lymphatic invasion (ILI) and peritumoral lymphatic invasion (PLI), were more prevalent (CD8, P = 0.0001 and P < 0.0001; CD3, P = 0.003 and P = 0.0003; ILI and PLI, respectively). A greater CD3+ and CD8+ lymphocyte infiltration was found in class 2 BRAF-mutated mCRC cases than in class 3 cases (P = 0.033) by Schirripa et al.33

Tumor-associated macrophages

TAMs are essential elements of TME that, by direct and indirect interactions with other immune cells, contribute to tumor immunosuppression, development, invasion, metastasis, angiogenesis, and drug tolerance.87 While M2 macrophages are believed to influence immunosuppression and the course of cancer, M1 macrophages are pro-inflammatory and anti-tumor.88 First identified as M2-like macrophages, TAMs were poor indicators for various cancer prognoses.88 On BRAF mutant CRC, however, not many studies have been reported. One study found that whereas CD68+ M1 macrophages showed no difference (mean ± standard deviation, 18.43 ± 13.53 vs. 20.96 ± 15.34, respectively, P = 0.664), CD163+ M2 macrophages were significantly increased in BRAF V600E mutant CRC tumor compared to a wild-type tumor (mean ± 5.93 vs. 3.67 ± 3.02, respectively, P = 0.040).89 M1 macrophages (P < 0.001, in both datasets) were higher in BRAF mutated patients than that in wild-type patients; no significant differences of M2 macrophages between BRAF mutant patients and BRAF wild-type patients (P > 0.05, in both datasets) were found in a second study combining 110 BRAF-mutated and 798 wild-type colon cancer patients from TCGA datasets (59 BRAF-mutated, 337 BRAF wild-type) and GEO datasets (51 BRAF-mutated, 441 BRAF wild-type).85 High tumor infiltration CD68+ macrophages had no predictive value for BRAF mutant mCRC in another study of a subgroup of patients with unresectable mCRC based on BRAF status (data not shown).88 In BRAF wild type mCRC with higher tumor infiltration CD68+ macrophages, however, a better mOS (high tumor infiltration CD68+ macrophages vs. low tumor infiltration CD68+ macrophages patients, 26 vs. 15 months, P = 0.002) was observed.88 Explanation of the features of TAMs in this subgroup of CRC for prospective immunotherapy and prognosis still needs more research.

Expression of PD-L1

Transmembrane protein PD-L1, sometimes referred as CD274 or B7 homolog 1, is produced on cancer cells and, by binding to PD-1 in T cells, suppresses the immune system. Now well acknowledged, immune checkpoint blockade (ICB) against PD-L1 or PD-1 at used to treat non-small cell lung cancer, melanoma, esophageal cancer, breast cancer, renal cancer, and gastric cancer.90,91 One of predictive indicators of response to ICIs is PD-L1 expression.92 Anti-PD-1/PD-L1 therapy is apparently ineffective for a significant number of CRC patients.93 To the best of our knowledge, MSI-H is a biomarker to predict how well anti-PD-1/PD-L1 therapy will work in CRC.55 Remarkably, MSI colon cancer had higher PD-L1 expression levels than the MSS cohort.94,95 It is therefore now a crucial clinical topic if PD-L1 expression could function as another useful biomarker to predict the effectiveness of ICB therapy in CRC. Specifically, 9%–15% of CRC patients express PD-L1.96

Poorly differentiated and solid/medullary histology, MSI-H or dMMR, and BRAF mutations signatures of the serrated neoplasia pathway of colorectal adenocarcinomas, are linked to activation of PD-L1 in CRC cells.95,97,98 Cen et al85 discovered, in tissue samples from 43 patients with colon cancer, that BRAF mutant colon cancer had significantly increased expression of PD-L1, PD-1, CTLA4, lymphocyte-activation gene 3, and TIM3, which is similar with results from 396 colon cancers from TCGA datasets. Furthermore, the connection between BRAF mutation and PD-L1 expression has been investigated recently.99,100 BRAF V600E produced a significant level of PD-L1 expression in MSS CRC cell lines,101 raising the possibility that PD-L1 expression is not limited to MSI-H BRAF-mutated CRC. Although no data are available for the subset of BRAF mutations, there is no clear correlation between PD-L1 expression and the effectiveness of anti-PD-1/PD-L1 immunotherapy in CRC.49,50 Other immune-independent functions of PD-L1 expression in BRAF mutant CRC were found. Feng et al102 shown that in colon tumors, oncogenic BRAF V600E can transcriptionally upregulate intrinsic PD-L1 expression, which by increasing BIM and BIK proteins, improves chemotherapy-induced apoptosis. Feng et al101 demonstrated how c-JUN and YAP promote PD-L1 expression in colon tumors with BRAF V600E.

Tertiary lymphoid structures (TLS)

Mostly developing in infectious diseases, inflammatory disorders, autoimmune syndromes, and tumors are TLS.103 As a part of the TME, TLS becomes implicated in tumor growth and metastasis.103 Moreover, TLS may function as a prognostic biomarker in human malignancies and as a predictive biomarker for ICIs.103,104 A higher degree of TLS density was associated with colorectal tumors with MSI-H and/or BRAF mutations (median: 0.61 vs. 0.45, rank-sum P = 0.03), according to Posch et al105 TLS mostly exhibited in the peripheral region of CRC tumor tissues (97%).

Patients with BRAF mutant CRC progresses quickly, has a bad prognosis, and responds poorly to conventional treatment. While the BEACON study shown that mCRC patients with BRAF V600E mutations can benefit clinically from a BRAF inhibitor plus an EGFR inhibitor ± MEK inhibitor, the proven ORR is only 20%.16 Elez et al106 discovered that improved ORR and PFS were achieved by BRAF mutant mCRC with MSS and ring finger protein 43 mutation. New treatment possibilities must be investigated, for BRAF mutant mCRC patients who are not benefited by targeted combination therapy. Durable responses have been obtained by immunological checkpoint inhibitor therapy in certain MSI-H/dMMR CRC patients. Subgroup analysis of clinical trials supports the advantage for MSI-H patients with BRAF-mutated mCRC; more research in this population is required to confirm. Not all MSI-H/dMMR patients, nevertheless, react effectively to immunological checkpoint inhibitor therapy. The investigation of new predictive biomarkers for screening immunotherapy-sensitive populations other than MSI-H should continue since BRAF mutant CRC is a diverse tumor subtype with varying efficacy for ICB therapy. Patients with MSS/pMMR CRC are usually known to not react to ICIs. Combination therapy with ICIs may be another approach to enhance prognosis for mCRC patients with MSS/pMMR and MSI-H/dMMR who do not react to ICIs. Blocking the MAPK pathway works in concert with immunotherapy. Promising results have been seen in mCRC patients with MSS/pMMR BRAF mutations with immunotherapy and MAPK pathway inhibition. To be sure of it, nevertheless, extensive prospective phase III clinical trials are still required. Furthermore, more combination treatments, like ICIs with radiation, chemotherapy, and tumor vaccines, are still needed to boost the host immune system and get over the immunotherapy insensitivity of particular patients.

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209-49.
    Pubmed CrossRef
  2. Brenner H, Stock C, Hoffmeister M. Effect of screening sigmoidoscopy and screening colonoscopy on colorectal cancer incidence and mortality: systematic review and meta-analysis of randomised controlled trials and observational studies. BMJ. 2014;348:g2467.
    Pubmed KoreaMed CrossRef
  3. Jin H, Amonkar M, Aguiar-Ibáñez R, Thosar M, Chase M, Keeping S. Systematic literature review and network meta-analysis of pembrolizumab versus other interventions for previously untreated, unresectable or metastatic, MSI-high or MMR-deficient CRC. Future Oncol. 2022;18:2155-71.
    Pubmed CrossRef
  4. Piawah S, Venook AP. Targeted therapy for colorectal cancer metastases: a review of current methods of molecularly targeted therapy and the use of tumor biomarkers in the treatment of metastatic colorectal cancer. Cancer. 2019;125:4139-47.
    Pubmed CrossRef
  5. O'Neil BH, Wallmark JM, Lorente D, Elez E, Raimbourg J, Gomez-Roca C, et al. Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced colorectal carcinoma. PLoS One. 2017;12:e0189848.
    Pubmed KoreaMed CrossRef
  6. National Comprehensive Cancer Network (NCCN). NCCN clinical practice guidelines in oncology, colon cancer (version 1.2022). NCCN; 2022.
  7. Yukimoto R, Nishida N, Hata T, Fujino S, Ogino T, Miyoshi N, et al. Specific activation of glycolytic enzyme enolase 2 in BRAF V600E-mutated colorectal cancer. Cancer Sci. 2021;112:2884-94.
    Pubmed KoreaMed CrossRef
  8. Kopetz S, Desai J, Chan E, Hecht JR, O'Dwyer PJ, Maru D, et al. Phase II pilot study of vemurafenib in patients with metastatic BRAF-mutated colorectal cancer. J Clin Oncol. 2015;33:4032-8.
    Pubmed KoreaMed CrossRef
  9. Park R, Lopes L, Lee S, Riano I, Saeed A. The prognostic and predictive impact of BRAF mutations in deficient mismatch repair/microsatellite instability-high colorectal cancer: systematic review/meta-analysis. Future Oncol. 2021;17:4221-31.
    Pubmed CrossRef
  10. Cremolini C, Loupakis F, Antoniotti C, Lupi C, Sensi E, Lonardi S, et al. FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab as first-line treatment of patients with metastatic colorectal cancer: updated overall survival and molecular subgroup analyses of the open-label, phase 3 TRIBE study. Lancet Oncol. 2015;16:1306-15.
    Pubmed CrossRef
  11. Corcoran RB, Ebi H, Turke AB, Coffee EM, Nishino M, Cogdill AP, et al. EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2012;2:227-35.
    Pubmed KoreaMed CrossRef
  12. Coffee EM, Faber AC, Roper J, Sinnamon MJ, Goel G, Keung L, et al. Concomitant BRAF and PI3K/mTOR blockade is required for effective treatment of BRAF(V600E) colorectal cancer. Clin Cancer Res. 2013;19:2688-98.
    Pubmed KoreaMed CrossRef
  13. Mao M, Tian F, Mariadason JM, Tsao CC, Lemos R Jr, Dayyani F, et al. Resistance to BRAF inhibition in BRAF-mutant colon cancer can be overcome with PI3K inhibition or demethylating agents. Clin Cancer Res. 2013;19:657-67.
    Pubmed KoreaMed CrossRef
  14. Su F, Bradley WD, Wang Q, Yang H, Xu L, Higgins B, et al. Resistance to selective BRAF inhibition can be mediated by modest upstream pathway activation. Cancer Res. 2012;72:969-78.
    Pubmed CrossRef
  15. Chen G, Gao C, Gao X, Zhang DH, Kuan SF, Burns TF, et al. Wnt/β-catenin pathway activation mediates adaptive resistance to BRAF inhibition in colorectal cancer. Mol Cancer Ther. 2018;17:806-13.
    Pubmed KoreaMed CrossRef
  16. Tabernero J, Grothey A, Van Cutsem E, Yaeger R, Wasan H, Yoshino T, et al. Encorafenib plus cetuximab as a new standard of care for previously treated BRAF V600E-mutant metastatic colorectal cancer: updated survival results and subgroup analyses from the BEACON study. J Clin Oncol. 2021;39:273-84.
    Pubmed KoreaMed CrossRef
  17. Kopetz S, Guthrie KA, Morris VK, Lenz HJ, Magliocco AM, Maru D, et al. Randomized trial of irinotecan and cetuximab with or without vemurafenib in BRAF-mutant metastatic colorectal cancer (SWOG S1406). J Clin Oncol. 2021;39:285-94.
    Pubmed KoreaMed CrossRef
  18. Takashima A, Kotani D, Kato T, Satoh T, Masuishi R, Komatsu Y, et al. Safety and efficacy of encorafenib, binimetinib, plus cetuximab for BRAF V600E-mutant metastatic colorectal cancer: results of a prospective study as an expanded access program. J Clin Oncol. 2022;40(4 Suppl):199-9.
    CrossRef
  19. Van Cutsem E, Taieb J, Yaeger R, Yoshino T, Maiello E, Elez E. O-10 ANCHOR CRC: results from a single-arm, phase 2 study of encorafenib, binimetinib plus cetuximab in previously untreated BRAF V600E-mutant metastatic colorectal cancer. Ann Oncol. 2021;32(Suppl 3):S222.
    CrossRef
  20. Kaiser J, Couzin-Frankel J. Cancer immunotherapy sweeps nobel for medicine. Science. 2018;362:13.
    Pubmed CrossRef
  21. Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021;16:223-49.
    Pubmed CrossRef
  22. Moslehi J, Lichtman AH, Sharpe AH, Galluzzi L, Kitsis RN. Immune checkpoint inhibitor-associated myocarditis: manifestations and mechanisms. J Clin Invest. 2021;131:e145186.
    Pubmed KoreaMed CrossRef
  23. Vilar E, Gruber SB. Microsatellite instability in colorectal cancer-the stable evidence. Nat Rev Clin Oncol. 2010;7:153-62.
    Pubmed KoreaMed CrossRef
  24. Aasebø KØ, Dragomir A, Sundström M, Mezheyeuski A, Edqvist PH, Eide GE, et al. Consequences of a high incidence of microsatellite instability and BRAF-mutated tumors: a population-based cohort of metastatic colorectal cancer patients. Cancer Med. 2019;8:3623-35.
    Pubmed KoreaMed CrossRef
  25. Diaz LA Jr, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt C, et al; KEYNOTE-177 Investigators. Pembrolizumab versus chemotherapy for microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer (KEYNOTE-177): final analysis of a randomised, open-label, phase 3 study. Lancet Oncol. 2022;23:659-70.
    Pubmed CrossRef
  26. Johnson B, Haymaker CL, Parra ER, Soto LMS, Wang X, Thomas JV, et al. Phase II study of durvalumab (anti-PD-L1) and trametinib (MEKi) in microsatellite stable (MSS) metastatic colorectal cancer (mCRC). J Immunother Cancer. 2022;10:e005332.
    Pubmed KoreaMed CrossRef
  27. Tabernero J, Ros J, Élez E. The evolving treatment landscape in BRAF-V600E-mutated metastatic colorectal cancer. Am Soc Clin Oncol Educ Book. 2022;42:1-10.
    Pubmed CrossRef
  28. Nieuwenhuis L, Simons CCJM, Weijenberg MP, van den Brandt PA. Nut and peanut butter intake and the risk of colorectal cancer and its anatomical and molecular subtypes: the Netherlands cohort study. Carcinogenesis. 2020;41:1368-84.
    Pubmed KoreaMed CrossRef
  29. Kambara T, Simms LA, Whitehall VL, Spring KJ, Wynter CV, Walsh MD, et al. BRAF mutation is associated with DNA methylation in serrated polyps and cancers of the colorectum. Gut. 2004;53:1137-44.
    Pubmed KoreaMed CrossRef
  30. Leach JDG, Vlahov N, Tsantoulis P, Ridgway RA, Flanagan DJ, Gilroy K, et al; S:CORT consortium. Oncogenic BRAF, unrestrained by TGFβ-receptor signalling, drives right-sided colonic tumorigenesis. Nat Commun. 2021;12:3464.
    Pubmed KoreaMed CrossRef
  31. Dankner M, Rose AAN, Rajkumar S, Siegel PM, Watson IR. Classifying BRAF alterations in cancer: new rational therapeutic strategies for actionable mutations. Oncogene. 2018;37:3183-99.
    Pubmed CrossRef
  32. Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, et al; Cancer Genome Project. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell. 2004;116:855-67.
    Pubmed CrossRef
  33. Schirripa M, Biason P, Lonardi S, Pella N, Pino MS, Urbano F, et al. Class 1, 2, and 3 BRAF-mutated metastatic colorectal cancer: a detailed clinical, pathologic, and molecular characterization. Clin Cancer Res. 2019;25:3954-61.
    Pubmed CrossRef
  34. Müller MF, Ibrahim AE, Arends MJ. Molecular pathological classification of colorectal cancer. Virchows Arch. 2016;469:125-34.
    Pubmed KoreaMed CrossRef
  35. Pino MS, Chung DC. The chromosomal instability pathway in colon cancer. Gastroenterology. 2010;138:2059-72.
    Pubmed KoreaMed CrossRef
  36. Dienstmann R, Vermeulen L, Guinney J, Kopetz S, Tejpar S, Tabernero J. Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer. 2017;17:79-92.
    Pubmed CrossRef
  37. Castro MP, Goldstein N. Mismatch repair deficiency associated with complete remission to combination programmed cell death ligand immune therapy in a patient with sporadic urothelial carcinoma: immunotheranostic considerations. J Immunother Cancer. 2015;3:58.
    Pubmed KoreaMed CrossRef
  38. Kochi M, Hinoi T, Niitsu H, Miguchi M, Saito Y, Sada H, et al. Oncogenic mutation in RAS-RAF axis leads to increased expression of GREB1, resulting in tumor proliferation in colorectal cancer. Cancer Sci. 2020;111:3540-9.
    Pubmed KoreaMed CrossRef
  39. Avvaru AK, Saxena S, Sowpati DT, Mishra RK. MSDB: a comprehensive database of simple sequence repeats. Genome Biol Evol. 2017;9:1797-802.
    Pubmed KoreaMed CrossRef
  40. Nakamura Y. Biomarkers for immune checkpoint inhibitor-mediated tumor response and adverse events. Front Med (Lausanne). 2019;6:119.
    Pubmed KoreaMed CrossRef
  41. Kreidieh M, Mukherji D, Temraz S, Shamseddine A. Expanding the scope of immunotherapy in colorectal cancer: current clinical approaches and future directions. Biomed Res Int. 2020;2020:9037217.
    Pubmed KoreaMed CrossRef
  42. Venderbosch S, Nagtegaal ID, Maughan TS, Smith CG, Cheadle JP, Fisher D, 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.
    Pubmed KoreaMed CrossRef
  43. Sahin IH, Klostergaard J. BRAF mutations as actionable targets: a paradigm shift in the management of colorectal cancer and novel avenues. JCO Oncol Pract. 2021;17:723-30.
    Pubmed CrossRef
  44. Guinney J, Dienstmann R, Wang X, de Reyniès A, Schlicker A, Soneson C, et al. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21:1350-6.
  45. Loupakis F, Biason P, Prete AA, Cremolini C, Pietrantonio F, Pella N, et al. CK7 and consensus molecular subtypes as major prognosticators in V600EBRAF mutated metastatic colorectal cancer. Br J Cancer. 2019;121:593-9.
    Pubmed KoreaMed CrossRef
  46. Barras D, Missiaglia E, Wirapati P, Sieber OM, Jorissen RN, Love C, et al. BRAF V600E mutant colorectal cancer subtypes based on gene expression. Clin Cancer Res. 2017;23:104-15.
    Pubmed CrossRef
  47. Middleton G, Yang Y, Campbell CD, André T, Atreya CE, Schellens JHM, et al. BRAF-mutant transcriptional subtypes predict outcome of combined BRAF, MEK, and EGFR blockade with dabrafenib, trametinib, and panitumumab in patients with colorectal cancer. Clin Cancer Res. 2020;26:2466-76.
    Pubmed KoreaMed CrossRef
  48. Le DT, Kim TW, Van Cutsem E, Geva R, Jäger D, Hara H, 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;38:11-9.
    Pubmed KoreaMed CrossRef
  49. Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, 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.
    Pubmed CrossRef
  50. André T, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, et al. Nivolumab plus low-dose ipilimumab in previously treated patients with microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: 4-year follow-up from CheckMate 142. Ann Oncol. 2022;33:1052-60.
    Pubmed CrossRef
  51. Lenz HJ, Van Cutsem E, Luisa Limon M, Wong KYM, Hendlisz A, Aglietta M, et al. First-line nivolumab plus low-dose ipilimumab for microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: the phase II CheckMate 142 study. J Clin Oncol. 2022;40:161-70.
    Pubmed CrossRef
  52. André T, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt C, et al. ; KEYNOTE-177 Investigators. Pembrolizumab in microsatellite-instability-high advanced colorectal cancer. N Engl J Med. 2020;383:2207-18.
    Pubmed CrossRef
  53. Andre T, Amonkar M, Norquist JM, Shiu KK, Kim TW, Jensen BV, et al. Health-related quality of life in patients with microsatellite instability-high or mismatch repair deficient metastatic colorectal cancer treated with first-line pembrolizumab versus chemotherapy (KEYNOTE-177): an open-label, randomised, phase 3 trial. Lancet Oncol. 2021;22:665-77.
    Pubmed CrossRef
  54. Sahin IH, Goyal S, Pumpalova Y, Sonbol MB, Das S, Haraldsdottir S, et al. Mismatch repair (MMR) gene alteration and BRAF V600E mutation are potential predictive biomarkers of immune checkpoint inhibitors in MMR-deficient colorectal cancer. Oncologist. 2021;26:668-75.
    Pubmed KoreaMed CrossRef
  55. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509-20.
  56. Overman MJ, Kopetz S, McDermott RS, Leach J, Lonardi S, Lenz HJ, et al. Nivolumab ± ipilimumab in treatment (tx) of patients (pts) with metastatic colorectal cancer (mCRC) with and without high microsatellite instability (MSI-H): CheckMate-142 interim results. J Clin Oncol. 2016;34(15 Suppl):3501.
    CrossRef
  57. Kim CW, Chon HJ, Kim C. Combination immunotherapies to overcome intrinsic resistance to checkpoint blockade in microsatellite stable colorectal cancer. Cancers (Basel). 2021;13:4906.
    Pubmed KoreaMed CrossRef
  58. Bottos A, Martini M, Di Nicolantonio F, Comunanza V, Maione F, Minassi A, et al. Targeting oncogenic serine/threonine-protein kinase BRAF in cancer cells inhibits angiogenesis and abrogates hypoxia. Proc Natl Acad Sci U S A. 2012;109:E353-9.
    Pubmed KoreaMed CrossRef
  59. Apte RS, Chen DS, Ferrara N. VEGF in signaling and disease: beyond discovery and development. Cell. 2019;176:1248-64.
    Pubmed KoreaMed CrossRef
  60. Bourhis M, Palle J, Galy-Fauroux I, Terme M. Direct and indirect modulation of T cells by VEGF-A counteracted by anti-angiogenic treatment. Front Immunol. 2021;12:616837.
    Pubmed KoreaMed CrossRef
  61. Khan KA, Kerbel RS. Improving immunotherapy outcomes with anti-angiogenic treatments and vice versa. Nat Rev Clin Oncol. 2018;15:310-24.
    Pubmed CrossRef
  62. Fukuoka S, Hara H, Takahashi N, Kojima T, Kawazoe A, Asayama M, et al. Regorafenib plus nivolumab in patients with advanced gastric or colorectal cancer: an open-label, dose-escalation, and dose-expansion phase Ib trial (REGONIVO, EPOC1603). J Clin Oncol. 2020;38:2053-61.
    Pubmed CrossRef
  63. Damato A, Bergamo F, Antonuzzo L, Nasti G, Pietrantonio F, Tonini G, et al. Phase II study of nivolumab in combination with FOLFOXIRI/bevacizumab as first-line treatment in patients with advanced colorectal cancer RAS/BRAF mutated (mut): NIVACOR trial (GOIRC-03-2018). J Clin Oncol. 2022;40(16 Suppl):3509.
    CrossRef
  64. Fang C, Lin J, Zhang T, Luo J, Nie D, Li M, et al. Metastatic colorectal cancer patient with microsatellite stability and BRAFV600E mutation showed a complete metabolic response to PD-1 blockade and bevacizumab: a case report. Front Oncol. 2021;11:652394.
    Pubmed KoreaMed CrossRef
  65. Avery TY, Köhler N, Zeiser R, Brummer T, Ruess DA. Onco-immunomodulatory properties of pharmacological interference with RAS-RAF-MEK-ERK pathway hyperactivation. Front Oncol. 2022;12:931774.
    Pubmed KoreaMed CrossRef
  66. Ascierto PA, Ferrucci PF, Fisher R, Del Vecchio M, Atkinson V, Schmidt H, et al. Dabrafenib, trametinib and pembrolizumab or placebo in BRAF-mutant melanoma. Nat Med. 2019;25:941-6.
    Pubmed CrossRef
  67. Ribas A, Lawrence D, Atkinson V, Agarwal S, Miller WH Jr, Carlino MS, et al. Combined BRAF and MEK inhibition with PD-1 blockade immunotherapy in BRAF-mutant melanoma. Nat Med. 2019;25:936-40.
    Pubmed KoreaMed CrossRef
  68. Gutzmer R, Stroyakovskiy D, Gogas H, Robert C, Lewis K, Protsenko S, et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAFV600 mutation-positive melanoma (IMspire150): primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2020;395:1835-44.
    Pubmed CrossRef
  69. Zimmer L, Livingstone E, Krackhardt A, Schultz ES, Göppner D, Assaf C, et al. Encorafenib, binimetinib plus pembrolizumab triplet therapy in patients with advanced BRAFV600 mutant melanoma: safety and tolerability results from the phase I IMMU-TARGET trial. Eur J Cancer. 2021;158:72-84.
    Pubmed CrossRef
  70. Corcoran R, Giannakis M, Allen J, Chen J, Pelka K, Chao S, et al. SO-26 clinical efficacy of combined BRAF, MEK, and PD-1 inhibition in BRAFV600E colorectal cancer patients. Ann Oncol. 2020;31(Suppl 3):S226-7.
    CrossRef
  71. Elez E, Ros J, Tabernero J. Seeking therapeutic synergy in BRAF mutant colorectal cancer. Nat Med. 2023;29:307-8.
    Pubmed CrossRef
  72. Corcoran RB, André T, Atreya CE, Schellens JHM, Yoshino T, Bendell JC, et al. Combined BRAF, EGFR, and MEK inhibition in patients with BRAFV600E-mutant colorectal cancer. Cancer Discov. 2018;8:428-43.
    Pubmed KoreaMed CrossRef
  73. Morris VK, Parseghian CM, Escano M, Johnson B, Raghav KPS, Dasari A, et al. Phase I/II trial of encorafenib, cetuximab, and nivolumab in patients with microsatellite stable (MSS), BRAFV600E metastatic colorectal cancer. J Clin Oncol. 2022;40(16 Suppl):3598.
    CrossRef
  74. Baghban R, Roshangar L, Jahanban-Esfahlan R, Seidi K, Ebrahimi-Kalan A, Jaymand M, et al. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal. 2020;18:59.
    Pubmed KoreaMed CrossRef
  75. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24:541-50.
    Pubmed KoreaMed CrossRef
  76. Leone RD, Powell JD. Metabolism of immune cells in cancer. Nat Rev Cancer. 2020;20:516-31.
    Pubmed KoreaMed CrossRef
  77. Bai Z, Zhou Y, Ye Z, Xiong J, Lan H, Wang F. Tumor-infiltrating lymphocytes in colorectal cancer: the fundamental indication and application on immunotherapy. Front Immunol. 2022;12:808964.
    Pubmed KoreaMed CrossRef
  78. Paijens ST, Vledder A, de Bruyn M, Nijman HW. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 2021;18:842-59.
    Pubmed KoreaMed CrossRef
  79. Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer. 2019;19:133-50.
    Pubmed KoreaMed CrossRef
  80. Fuchs TL, Sioson L, Sheen A, Jafari-Nejad K, Renaud CJ, Andrici J, et al. Assessment of tumor-infiltrating lymphocytes using international TILs working group (ITWG) system is a strong predictor of overall survival in colorectal carcinoma: a study of 1034 patients. Am J Surg Pathol. 2020;44:536-44.
    Pubmed CrossRef
  81. Williams DS, Mouradov D, Newman MR, Amini E, Nickless DK, Fang CG, et al. Tumour infiltrating lymphocyte status is superior to histological grade, DNA mismatch repair and BRAF mutation for prognosis of colorectal adenocarcinomas with mucinous differentiation. Mod Pathol. 2020;33:1420-32.
    Pubmed CrossRef
  82. Nazemalhosseini-Mojarad E, Mohammadpour S, Torshizi Esafahani A, Gharib E, Larki P, Moradi A, et al. Intratumoral infiltrating lymphocytes correlate with improved survival in colorectal cancer patients: independent of oncogenetic features. J Cell Physiol. 2019;234:4768-77.
    Pubmed CrossRef
  83. Nosho K, Baba Y, Tanaka N, Shima K, Hayashi M, Meyerhardt JA, et al. Tumour-infiltrating T-cell subsets, molecular changes in colorectal cancer, and prognosis: cohort study and literature review. J Pathol. 2010;222:350-66.
    Pubmed KoreaMed CrossRef
  84. Kwak Y, Koh J, Kim DW, Kang SB, Kim WH, Lee HS. Immunoscore encompassing CD3+ and CD8+ T cell densities in distant metastasis is a robust prognostic marker for advanced colorectal cancer. Oncotarget. 2016;7:81778-90.
    Pubmed KoreaMed CrossRef
  85. Cen S, Liu K, Zheng Y, Shan J, Jing C, Gao J, et al. BRAF mutation as a potential therapeutic target for checkpoint inhibitors: a comprehensive analysis of immune microenvironment in BRAF mutated colon cancer. Front Cell Dev Biol. 2021;9:705060.
    Pubmed KoreaMed CrossRef
  86. Digiacomo N, Bolzacchini E, Veronesi G, Cerutti R, Sahnane N, Pinotti G, et al. Neuroendocrine differentiation, microsatellite instability, and tumor-infiltrating lymphocytes in advanced colorectal cancer with BRAF mutation. Clin Colorectal Cancer. 2019;18:e251-60.
    Pubmed CrossRef
  87. Wang H, Tian T, Zhang J. Tumor-associated macrophages (TAMs) in colorectal cancer (CRC): from mechanism to therapy and prognosis. Int J Mol Sci. 2021;22:8470.
    Pubmed KoreaMed CrossRef
  88. Aasebø K, Bruun J, Bergsland CH, Nunes L, Eide GE, Pfeiffer P, et al. Prognostic role of tumour-infiltrating lymphocytes and macrophages in relation to MSI, CDX2 and BRAF status: a population-based study of metastatic colorectal cancer patients. Br J Cancer. 2022;126:48-56.
    Pubmed KoreaMed CrossRef
  89. Zhi J, Jia XJ, Yan J, Wang HC, Feng B, Xing HY, et al. BRAFV600E mutant colorectal cancer cells mediate local immunosuppressive microenvironment through exosomal long noncoding RNAs. World J Gastrointest Oncol. 2021;13:2129-48.
    Pubmed KoreaMed CrossRef
  90. Kornepati AVR, Vadlamudi RK, Curiel TJ. Programmed death ligand 1 signals in cancer cells. Nat Rev Cancer. 2022;22:174-89.
    Pubmed KoreaMed CrossRef
  91. Yi M, Zheng X, Niu M, Zhu S, Ge H, Wu K. Combination strategies with PD-1/PD-L1 blockade: current advances and future directions. Mol Cancer. 2022;21:28.
    Pubmed KoreaMed CrossRef
  92. Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14:847-56.
    Pubmed CrossRef
  93. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455-65.
    Pubmed KoreaMed CrossRef
  94. Salem ME, Puccini A, Grothey A, Raghavan D, Goldberg RM, Xiu J, et al. Landscape of tumor mutation load, mismatch repair deficiency, and PD-L1 expression in a large patient cohort of gastrointestinal cancers. Mol Cancer Res. 2018;16:805-12.
    Pubmed KoreaMed CrossRef
  95. Rosenbaum MW, Bledsoe JR, Morales-Oyarvide V, Huynh TG, Mino-Kenudson M. PD-L1 expression in colorectal cancer is associated with microsatellite instability, BRAF mutation, medullary morphology and cytotoxic tumor-infiltrating lymphocytes. Mod Pathol. 2016;29:1104-12.
    Pubmed CrossRef
  96. Sudoyo AW, Kurniawan AN, Kusumo GD, Putra TP, Rexana FA, Yunus M, et al. Increased CD8 tumor infiltrating lymphocytes in colorectal cancer microenvironment supports an adaptive immune resistance mechanism of PD-L1 expression. Asian Pac J Cancer Prev. 2019;20:3421-7.
    Pubmed KoreaMed CrossRef
  97. Inaguma S, Lasota J, Wang Z, Felisiak-Golabek A, Ikeda H, Miettinen M. Clinicopathologic profile, immunophenotype, and genotype of CD274 (PD-L1)-positive colorectal carcinomas. Mod Pathol. 2017;30:278-85.
    Pubmed KoreaMed CrossRef
  98. Siraj AK, Parvathareddy SK, Annaiyappanaidu P, Haqawi W, Al-Rasheed M, AlManea HM, et al. PD-L1 expression is associated with deficient mismatch repair and poor prognosis in middle eastern colorectal cancers. J Pers Med. 2021;11:73.
    Pubmed KoreaMed CrossRef
  99. Carballal S, Balaguer F, IJspeert JEG. Serrated polyposis syndrome; epidemiology and management. Best Pract Res Clin Gastroenterol. 2022;58-59:101791.
    Pubmed CrossRef
  100. Ma R, Liu Y, Che X, Li C, Wen T, Hou K, et al. Nuclear PD-L1 promotes cell cycle progression of BRAF-mutated colorectal cancer by inhibiting THRAP3. Cancer Lett. 2022;527:127-39.
    Pubmed CrossRef
  101. Feng D, Chen Z, He X, Huang S, Zhang Z. Loss of tumor intrinsic PD-L1 confers resistance to drug-induced apoptosis in human colon cancer. Neoplasma. 2021;68:144-53.
    Pubmed CrossRef
  102. Feng D, Qin B, Pal K, Sun L, Dutta S, Dong H, et al. BRAFV600E-induced, tumor intrinsic PD-L1 can regulate chemotherapy-induced apoptosis in human colon cancer cells and in tumor xenografts. Oncogene. 2019;38:6752-66.
    Pubmed KoreaMed CrossRef
  103. Sautès-Fridman C, Petitprez F, Calderaro J, Fridman WH. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019;19:307-25.
    Pubmed CrossRef
  104. Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer. Science. 2022;375:eabf9419.
    Pubmed CrossRef
  105. Posch F, Silina K, Leibl S, Mündlein A, Moch H, Siebenhüner A, et al. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. Oncoimmunology. 2017;7:e1378844.
    Pubmed KoreaMed CrossRef
  106. Elez E, Ros J, Fernández J, Villacampa G, Moreno-Cárdenas AB, Arenillas C, et al. RNF43 mutations predict response to anti-BRAF/EGFR combinatory therapies in BRAFV600E metastatic colorectal cancer. Nat Med. 2022;28:2162-70.
    Pubmed KoreaMed CrossRef