Back to Journals » OncoTargets and Therapy » Volume 17
Progress of Immune Checkpoint Inhibitors Therapy for pMMR/MSS Metastatic Colorectal Cancer
Received 11 October 2024
Accepted for publication 12 December 2024
Published 24 December 2024 Volume 2024:17 Pages 1223—1253
DOI https://doi.org/10.2147/OTT.S500281
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Dr John Maher
Fanjie Qu, Shuang Wu, WeiWei Yu
Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
Correspondence: Fanjie Qu, Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, No. 40 Qianshan Road, Dalian, Liaoning, 116033, People’s Republic of China, Email [email protected]
Abstract: Immunotherapy is one of the research hotspots in colorectal cancer field in recent years. The colorectal cancer patients with mismatch repair-deficient (dMMR) or high microsatellite instability (MSI-H) are the primary beneficiaries of immunotherapy. However, the vast majority of colorectal cancers are mismatch repair proficient (pMMR) or microsatellite stability (MSS), and their immune microenvironment is characterized by “cold tumors” that are generally insensitive to single immunotherapy based on immune checkpoint inhibitors (ICIs). Studies have shown that some pMMR/MSS colorectal cancer patients regulate the immune microenvironment by combining other treatments, such as multi-target tyrosine kinase inhibitors, anti-vascular endothelial growth factor (VEGF) monoclonal antibodies, chemotherapy, radiotherapy, anti-epithelial growth factor receptor (EGFR) monoclonal antibodies, and mitogen-activated protein kinase (MAPK) signaling pathway inhibitors and oncolytic viruses, etc. to transform “cold tumor” into “hot tumor”, thereby improving the response to immunotherapy. In addition, screening for potential prognostic biomarkers can also enrich the population benefiting from immunotherapy for microsatellite stable colorectal cancer. Therefore, in pMMR or MSS metastatic colorectal cancer (mCRC), the optimization of immunotherapy regimens and the search for effective efficacy prediction biomarkers are currently important research directions. In this paper, we review the progress of efficacy of immunotherapy (mainly ICIs) in pMMR /MSS mCRC, challenges and potential markers, in order to provide research ideas for the development of immunotherapy for mCRC.
Keywords: immune checkpoint inhibitors, metastatic colorectal cancer, pMMR/MSS, combination immunotherapy, biomarkers
Background
Immune checkpoint inhibitors (ICIs) enhance anti-tumor immunity by regulating the interaction between T lymphocytes, antigen presenting cells (APC) and tumor cells, and its mechanism is to reactivate the immune response of T cells to tumor cells and inhibit tumor growth, which has brought long-term and lasting benefits to patients with multiple types of solid tumors.1,2 The dMMR/MSI-H tumors are the main beneficiaries of immunotherapy due to their high immunogenicity, strong lymphocyte infiltration in tumor microenvironment, good prognosis, and insensitivity to conventional radiotherapy and chemotherapy.3–5
The results of KEYNOTE 016, reported by Professor Le’s team at Hopkins Hospital in 2015, showed that dMMR metastatic colorectal cancer (mCRC) could benefit from pembrolizumab monotherapy, This study opened the door of immunotherapy for colorectal cancer and opened the era of immunotherapy for colorectal cancer.6 Subsequently, data from the first phase of the CheckMate142 study, reported in 2016, suggested that recurrent dMMR/MSI-H mCRC could benefit from a combination of nivolumab and ipilimumab.7 Long-term follow-up showed that the combination helped dMMR/MSI-H mCRC patients achieve 77% PFS and 83% OS at 1 year. Based on these data, the US Food and Drug Administration (FDA) has approved the combination of nivolumab and ipilimumab for previously treated dMMR/MSI-H mCRC patients.8 The KEYNOTE-177 study, published in 2020, is the most important clinical trial in first-line therapy to date, which confirmed that pembrolizumab monotherapy can be used as the first-line standard treatment for dMMR/MSI-H colorectal cancer.9,10 KEYNOTE-177 successfully rewrote the NCCN colorectal cancer treatment guidelines.11
Studies on the mechanism have found that somatic mutations in MSI-H colorectal cancer produce a large number of nascent peptides, which can act as specific antigens of tumor cells.11 The combination of these neoantigens with the major histocompatibility complex (MHC) can induce the immune response of CD4+ or CD8+ T cells, while attracting the infiltration of tumor infiltrating lymphocyte (TIL) in the tumor microenvironment, thus making the tumor “hot” and increasing the response of CRC to immunotherapy.12,13
Although dMMR/MSI-H patients belong to the “hot tumor” population that is sensitive to immunotherapy, the proportion of dMMR/MSI-H patients in all mCRC are only 4% to 5%,10 and a larger proportion of pMMR /MSS patients belong to the “cold tumor” population that is insensitive to immunotherapy.14 Studies have found that for pMMR /MSS colorectal cancer, the tumor lacks antigens that induce T cell initiation, and the tumor microenvironment lacks T cell infiltration, so the antitumor effect of ICIs in these “cold tumor” populations that are not sensitive to immunotherapy has certain limitations.15–17 Existing studies have shown that the use of a single ICIs alone is essentially ineffective for pMMR/MSS mCRC.6,18 How to break through this dilemma has always been the challenge. Most of the current treatment strategies use combination therapy to reverse the “cold tumor” into the “hot tumor” sensitive to immunotherapy, so as to improve the efficacy of immunotherapy in pMMR/MSS colorectal cancer, so that immunotherapy can benefit more colorectal cancer patients.16,17,19,20 Although some combination treatment strategies have achieved good short-term efficacy and are promising in the treatment of patients with mCRC, most of them are single-arm small sample exploratory studies, and the research data are not completely stable and consistent (Table 1), and the safety and efficacy need to be verified by more prospective clinical trials with large samples.14,21
![]() |
Table 1 The Results of Clinical Trials Based on ICIs in Combination with Other Modalities in pMMR/MSS Metastatic Colorectal Cancer |
In addition, several studies are looking for other markers with potential to predict immunotherapy efficacy, not only for mismatch repair genes, but also for other molecular markers to enrich the population who can benefit from immunotherapy for pMMR/MSS colorectal cancer. Current studies have found that multiple molecular markers such as tumor mutation burden (TMB), polymerase ε and polymerase δ (POLE/POLD1) mutations, PD-L1 expression, tumor infiltrating lymphocytes, and gut microbiota have the potential and value to predict the efficacy of ICIs for pMMR/MSS colorectal cancer.29,68–76 This article aims to review the research progress of ICIs combination therapy for pMMR/MSS mCRC and related studies on the predictive markers of ICIs efficacy, in order to provide references for finding the best immunotherapy strategy for pMMR/MSS mCRC patients.
ICIs Combined with Multi-Target Tyrosine Kinase Inhibitors
The mechanism of primary resistance to immunotherapy in pMMR/MSS colorectal cancer patients is complex and involves many factors in tumor cells and tumor microenvironment. The tumor microenvironment is composed of blood and lymphovascular networks, extracellular matrix (ECM), various stromal cells (including fibroblasts, mesenchymal stromal cells, and pericytes), and resident and infiltrating immune cells (including T lymphocytes, B lymphocytes, natural killer cells, and tumor-associated macrophages.77,78 Tumor neovascularization plays an important role in tumor microenvironment. Previous studies have shown that overexpression of VEGF and the resulting over-activation of VEGF receptors stimulate endothelial cell proliferation, promote angiogenesis, and improve the availability of oxygen and nutrients in tumors.79 Meanwhile, abnormal activation of receptor tyrosine kinase (RTK), including endothelial growth factor (EGF), platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), and hepatocyte growth factor (HGF), plays a key role in the development and progression of cancer. This makes abnormal RTK a potential therapeutic target for cancer therapy.79,80
Preclinical studies have shown that inhibitors targeting VEGF/VEGF receptor have both effective anti-angiogenesis and immunomodulatory effects, can induce tumor vascular degeneration, improve hypoxia and low pH in the tumor microenvironment, and increase the infiltration of immune T lymphocytes in the tumor microenvironment through tumor revascularization, then an inflammatory tumor microenvironment is generated;In addition, anti-VEGF therapy has a synergistic effect with immunotherapy by up-regulating the expression of PD-L1, reducing immunosuppressive cells [such as tumour-associated macrophages (TAMs) and regulatory T cells (Tregs)], and enhancing the anti-tumor activity of CD8+T cells.81,82 Studies in mouse models have confirmed that the combination of anti-VEGFR2 and anti-PD-1 monoclonal antibodies can increase the production of tumor interferon-gamma, tumor necrosis factor-α and granzyme B, thereby enhancing the immune response of mice.83
Regorafenib is the first FDA-approved multi-target TKI in metastatic colorectal cancer that has progressed after treatment with standard treatment, targeting angiogenesis (VEGFR1-3, TIE2), stroma (PDGFR-β) and cancer-causing receptor tyrosine kinases (KIT, RET and RAF).84,85 Regorafenib has been shown to regulate anti-tumor immunity through different mechanisms. In a mouse hepatocellular carcinoma model, regorafenib normalizes tumor blood vessels by targeting VEGFR2/3, promotes the proliferation and activation of CD8+T cells, and improves the anti-tumor immunity of PD-1 blockers.86
In addition, Indoleamine 2,3-dioxygenase 1 (IDO1) overexpression has been shown to induce dendritic cells and regulatory T cells in a variety of cancers, leading to immunosuppressive effects, and therefore IDO1 is likely to be a potential target for anti-tumor immunotherapeutic intervention.87,88 Studies have shown that regorafenib can simultaneously reduce the expression of PD-L1 and IDO1, reduce the TAM infiltration, and produce synergistic effects with anti-PD-1 monoclonal antibodies to significantly improve the anti-tumor effect.89 Doleschel also found that regorafenib significantly reduced the infiltration of immunosuppressive TAM and Treg cells into the tumor microenvironment in his study of CRC mouse models developed after chemotherapy, while anti-PD-1 treatment significantly increased intratumoral IFN-γ levels, thus the two have a synergistic effect in regulating anti-tumor immune response, thus achieving sustained tumor suppression.90 Based on the above theoretical basis, in recent years, the combination of multi-target receptor tyrosine kinase inhibitors, including regorafenib, and immune checkpoint inhibitors has been attempted in pMMR/MSS mCRC.
The REGONIVO study, reported in Japan in 2020, is a Phase I b trial that enrolled 24 patients with pMMR/MSS colorectal cancer after failure of standard treatment to evaluate the safety and efficacy of the combination of regorafenib and nivolumab. The results showed that the objective response rate (ORR) was 33.3% and the median progression free survival (PFS) was 7.9 months. The 1-year PFS rate and overall survival (OS) rate were 41.8% and 68.0%, respectively.22 This study shows that some MSS mCRC patients may benefit from the combination of PD-1 inhibitors with multi-target TKI. However, this result was not replicated in the North American Phase II study, with an ORR of only 7.1%, median PFS of 1.8 months, and median OS (data immature) of 11.9 months. All responders had no liver metastases at baseline.23 It is suggested that the benefit group should be screened in this treatment mode. The researchers subsequently conducted a retrospective exploratory analysis, and the results suggested that there was a certain correlation between some specific biomarkers and treatment response.91
The Phase II REGOMUNE study is evaluating the efficacy and safety of regorafenib in combination with Avelumab in treated MSS colorectal cancer. Of the 40 evaluable patients, the best response was in 23 patients (57.5%) who were stable, with a median PFS and OS of 3.6 and 10.8 months, respectively.24,25 Studies have shown that PFS and OS obtained by this combination therapy are superior to historical data obtained by regorafenib alone.92 The REGOTORI study, led by Professor Xu Ruihua, is a Phase Ib/ II clinical trial evaluating the efficacy of regorafenib combined with the PD-1 inhibitor toripalimab in treated MSS mCRC. The ORR of 33 enrolled patients was 15.2%, and the median PFS and OS were 2.1 and 15.5 months, respectively. Studies have also shown that patients with lung metastasis have a relatively higher effective rate, while patients with only liver metastasis have a relatively poor effect.26
Lenvatinib is another multi-target receptor TKI. In pre-clinical studies, it was found that Lenvatinib combined with pembrolizumab can activate CD8+ T cells by reducing TAM and activating interferon pathway, thus producing synergistic effects with anti-PD-1 antibodies and enhancing anti-tumor effects.93 LEAP-005 (NCT03797326) is a Phase II study of lenvatinib and pembrolizumab in previously treated patients with advanced solid tumors of pMMR/MSS.27 The 32 MSS colorectal cancer patients had an ORR of 22% and a disease control rate (DCR) of 47%; and the median PFS and OS were 2.3 months and 7.5 months, respectively. It showed good antitumor activity and a manageable safety profile. The Phase III LEAP 017 study (NCT04776148) is being recruited worldwide based on the results of pembrolizumab in combination with lenvatinib in treated pMMR/MSS mCRC patients.
Fruquintinib is a potent and highly selective small-molecule TKI against VEGFR1, 2, and 3,94 and its survival benefits have been demonstrated in patients with mCRC in the Phase 3 FRESCO clinical trial.95 Clinical trials of fruquintinib in combination with immunotherapy are also underway. Preliminary results from a Phase 1b/2 study of sintilimab combined with fruquintinib in the treatment of advanced colorectal cancer (NCT03903705) suggest that the combination of sintilimab and fruquintinib is effective and safe to tolerate in patients with mCRC who have failed standard therapy. In this study,44 mCRC patients with disease progression after receiving at least two-line chemotherapy containing fluorouracil, oxaliplatin or irinotecan in the past were included. Patients who received intermittent administration of Fruquintinib combined with sintilimab had ORR of 23.8% and mPFS of 6.9 months, and mOS of 14.8 months respectively.28
It can be seen that despite the exciting results of the REGONIVO Phase Ib study, and ORR in MSS colorectal cancer patients reached 33%,22 thus opening the paradigm of refractory MSS colorectal cancer with regorafenib combined with anti-PD-(L)1 monoclonal antibody. However, subsequent single-arm studies using immune checkpoint inhibitors in combination with different anti-angiogenesis targeting agents (tyrosine kinase inhibitors) as previously treated mCRC therapy have been reported, including the North American REGONIVO study,23 REGOMUNE study (regorafenib combined with Avelumab),25 REGOTORI study (regorafenib combined with toripalimab),26 LEAP-005 colorectal cancer cohort (Lenvatinib combined with pembrolizumab).27 Most studies did not repeat the results consistent with the Japanese REGONIVO study, but in general, the reported ORR of these studies ranged from 7% to 27%, DCR from 39% to 80%, and median OS from 7.5 to 15.5 months. Compared with previous studies on standard third-line monotherapy (regorafenib, fruquintinib or TAS-102), both ORR and OS were numerically improved.92,95,96 However, the sample size of these studies is limited, so more high-level phase III randomized controlled trials are needed for validation, and the preliminary study supports the need to further explore the screening of advantageous populations in the future.
Combination Therapy with Two ICIs
Anti-PD-(L)1 Combined with Anti-CTLA-4 Therapy
Cytotoxic T lymphocyte antigen-4(CTLA-4) mainly inhibits T cell activation in the initial phase, thereby inhibiting T cell activity.97 Anti-CTLA-4 monoclonal antibody can block the inhibitory signal transduction of CTLA-4, thereby promoting T cell activation and immune response to tumor cells; However, anti-PD-(L)1 monoclonal antibody removes the inhibition of the combination of PD-1 and PD-L1 on activated T cells and promotes the killing effect of T cells on tumor cells.1,98 Therefore, the combination of the two can synergistically promote the anti-tumor immune response through the blocking complementary mechanism, thereby the acquired resistance to ICIs monotherapy is overcome1,99 and has a good safety profile.100 It has been observed that anti-PD (L)-1 antibody combined with anti-CTLA-4 antibody treatment has shown relatively excellent anti-tumor effects in a variety of malignant tumors (including melanoma, lung cancer, etc),101,102 and has begun to be explored in colorectal cancer, although the exact mechanism of action is still unclear.103
The CCTG CO.26 randomized Phase II trial evaluated whether the combination of durvalumab and tremelimumab improved survival in patients with advanced refractory colorectal cancer compared to best supportive care (BSC). A total of 180 patients were included in the study, excluding patients with dMMR, and the results showed that the study met the primary endpoint, with the median OS increased from 4.1 months in the BSC group to 6.6 months in the dual immunotherapy group, while the incidence of grade 3 or 4 adverse events also increased significantly.29 Although this study was the first to report positive results for PD-L1 monoclonal antibody combined with CTLA-4 monoclonal antibody in MSS mCRC, it was not widely accepted in the clinic, mainly because no patients in the study achieved complete response (CR). Only one patient with MSS type achieved partial response (PR). This study suggests the potential role of dual immunotherapy in MSS mCRC.29
In a clinical trial (NCT02754856), a combination of durvalumab and tremelimumab was administered to 21 pMMR patients with only liver metastasis before surgery, and evidence of extended relapse-free survival (RFS) and T cell activation was initially observed.104 In addition, the Fakih team conducted a phase I study on nivolumab combined with ipilimumab and regorafenib in MSS mCRC patients who had not been treated with regorafenib and anti-PD (L) −1 monoclonal antibody,30 and also achieved surprising results. ORR reached 31%, and the median PFS and OS were 4 months and 19.6 months, respectively, which were significantly improved compared with traditional third-line treatment for mCRC.92,95,96
Botensilimab (AGEN1181) is a new generation FC-enhanced CTLA-4 inhibitor that blocks the inhibitory function of CTLA-4 interactions with its ligands CD80 and CD86. A Phase 1A/B study by Professor Bullock et al explored the efficacy and safety of botensilimab in combination with balstilimab (AGEN2034, a PD-1 inhibitor) in treated MSS patients with mCRC. 34% of enrolled patients had previously received immunotherapy, and the median number of prior treatment lines was 4. Results showed that dual immunotherapy had an ORR of 24% and a disease control rate (DCR) of 73% in 41 evaluable patients. At the same time, the safety status was good, and most treatment-related adverse events were grade 1–3.31 A randomized Phase II trial (NCT05608044) is about to start.
Anti- PD-(L)1 Combined with New Generation of Immune Checkpoint Inhibitors
To enable more patients to benefit from immunotherapy, there is growing interest in other immune checkpoints and new potential targets have been identified, such as LAG-3, TIGIT, TIM-3 or VISTA. These receptors, like PD-1 or CTLA-4, are physiologically classified as inhibitory immune checkpoint receptors that are induced after T cell activation to prevent excessive stimulation.105–108 LAG-3,109 TIGIT,110 TIM-3,105,111 and VISTA112 are frequently co-expressed with PD-1 on tumor infiltrating lymphocytes (TILs) in a variety of cancers, including renal cell carcinoma (RCC), non-small cell lung cancer (NSCLC), ovarian cancer, and melanoma.
Lymphocyte activation Gene-3(LAG-3) is an inhibitory receptor on the surface of T cells and is often overexpressed in colorectal cancer.113 LAG-3 molecule can negatively regulate T lymphocytes, and the combination of LAG-3 inhibitor and PD-(L)1 inhibitor can enhance anti-tumor activity.114 As one of the new targets, LAG-3 has great potential in tumor immunotherapy. Targeting LAG-3 and its corresponding inhibitors may have a synergistic effect with anti-PD-(L)1 drugs to improve the immune response to cancer cells.113
The LAG-3 antibody favezelimab combined with pembrolizumab is currently in Phase I studies,32 with an ORR of 6.3% in 89 patients with pMMR/MSS mCRC who progressed after prior ≥2line therapy. The median PFS and OS were 2.1 and 8.3 months, respectively. Meanwhile, the median OS (12.7 months) in Combined Positive Score (CPS) ≥1 group was better than that in CPS<1 group (only 6.7 months), suggesting that further screening according to the level of PD-L1 expression in pMMR/MSS patients can identify the advantaged patients receiving LAG-3 antibody combined with anti-PD -(L)1 therapy.
T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory domains (TIGIT) is an inhibitory receptor shared by T cells and natural killer (NK) cells. On the one hand, TIGIT can inhibit the function of NK cells, prevent their killing of tumor cells and release of tumor cell antigens. On the other hand, it can directly inhibit the function of CD8+T cells, thereby inhibiting the killing effect of CD8+T cells on tumors; Meanwhile, Tregs with high expression of TIGIT can inhibit a variety of immune cells in the tumor immune cycle.115–118 Therefore, antibodies targeting TIGIT can restore the tumor killing ability of NK cells and CD8+T cells, and restore the anti-tumor immune response to normal.118,119 Based on the inhibitory effect of TIGIT in anti-tumor immune response, it has become a new target of tumor immunotherapy with potential.
Some studies have shown that although blocking TIGIT or PD-1 alone can enhance the function of CD8+ T cells, blocking both receptors at the same time is more effective in enhancing anti-tumor immunity of CD8+ T cells.120–122 Thibaudin et al found in vitro that Atezolizumab alone could only reactivate T cells from MSI-H tumors. The combination of Atezolizumab and anti-TIGIT monoclonal antibody tiragolumab reactivated T cells in 46% of MSS CRC samples, suggesting that the combination could initiate clinical trials in colorectal cancer patients with MSS status.123
GO30103 is the first human study to study the treatment of advanced solid tumors with anti-TIGIT monoclonal antibody tiragolumab.124 In this non-randomized controlled trial, tiragolumab was well tolerated with or without atezolizumab and showed initial antitumor activity of the combination therapy. Later, some other anti- TIGIT monoclonal antibodies such as vibostolimab and etigilimab combined with anti-PD-1 monoclonal antibodies in the treatment of advanced solid tumors were also reported, and the results were basically consistent.125,126 CITYSCAPE study is a phase II clinical study of TIGIT inhibitor tiragolumab combined with PD-L1 inhibitor Atezolizumab in the treatment of patients with PD-L1 positive metastatic NSCLC.127 The combination of tiragolumab and Atezolizumab resulted in a significant improvement in PFS compared to Atezolizumab monotherapy, with no increased safety risk. However, unfortunately, the PFS of the subsequent Phase III SKYSCRAPER 01 clinical study on NSCLC patients with high PD-L1 expression did not reach positive results, and the OS data was not mature.
T cell immunoglobulin and mucin domain 3 (TIM-3) are activation-induced inhibitory molecules involved in tumor immune tolerance and T cell depletion, while blocking TIM-3 pathway can promote the production of interferon-γ by T cells and enhance the immune function of tumor cells.128 Studies have found that in colorectal cancer, tumor-infiltrating CD8+ T cells that express both PD-1 and TIM3 have significantly lower levels of IFN-γ production, indicating that PD-1+ TIM-3+ CD8+ T cells are more dysfunctional than PD-1+ TIM-3-CD8 +T cells. Blocking TIM-3 may restore T cell response.129 Therefore, targeting TIM-3 is a promising approach to further improve the effectiveness of current immunotherapy. Data from several I/II clinical trials indicate that the anti-TIM-3 antibodies LY3321367 and MBG453 are well tolerated as monotherapy and in combination with anti-PD-L1 LY3300054 or anti-PD-1 PDR001 monoclonal antibodies, respectively; and showed preliminary antitumor activity.130,131 Encouragingly, partial responses were observed in 2 of the 6 patients with colorectal cancer who received the combination therapy.130
The immune regulation of colorectal cancer is a multi-link, multi-step and multi-mechanism process. These findings suggest that the use of single-agent anti-PD-1 monoclonal antibodies is not always sufficient to effectively restore T cell function, especially when these T cells co-express a variety of other immunosuppressive receptors. The results of the above clinical studies initially confirmed the effectiveness and safety of the combined treatment strategy of PD-L1 inhibitor and CTLA-4 inhibitor in MSS colorectal cancer, but most of the current studies are in the early stage, and more randomized controlled studies are still needed to continue to expand the sample for further verification. In addition, although the efficacy and safety of the combined treatment strategy of PD-L1 inhibitor and novel immune checkpoint inhibitors (LAG-3, TIGIT, TIM-3, VISTA, etc.) in MSS colorectal cancer has not yet been studied in clinical studies, according to the results of current clinical studies, They are expected to block multiple inhibitory signal pathways through combination therapy, thereby enhancing the anti-tumor immune response of the body to tumor cells, which is a promising anti-tumor immunotherapy strategy. In conclusion, combined immunotherapy strategies based on different mechanisms of action should be one of the models to continue to be explored in MSS colorectal cancer in the future.
ICIs Combined with Chemotherapy
ICIs Combined with Chemotherapy Alone
Chemotherapy-induced tumor cell death can promote the release and presentation of tumor antigens, increase tumor immunogenicity, and activate and improve the activity of CD8+ T lymphocytes, so immune checkpoint inhibitors combined with cytotoxic chemotherapy can theoretically improve the efficacy.19,132–134 At present, basic chemotherapy drugs for colorectal cancer include platinum (such as oxaliplatin), topoisomerase inhibitors (such as irinotecan) and antimetabolic drugs (such as 5-fluorouracil).135 It was found that both 5-fluorouracil and oxaliplatin were immunogenic. 5-fluorouracil (5FU) has selective cytotoxicity to myeloid-derived suppressor cells (MDSCs). In vivo, 5FU treatment of tumor-bearing mice resulted in a significant decrease in the number of MDSCs in the mice’s spleen and tumor bed, but had no significant effect on T lymphocytes, natural killer cells, dendritic cells, or B lymphocytes.136 Oxaliplatin can also induce immunogenic cell death and increase the immunogenicity of MSS tumors, thereby improving the efficacy of immunotherapy when combined with PD-(L)1 inhibitors.137,138
Studies of Dosset et al showed that 5-FU combined with oxaliplatin can improve anti-tumor immune response in MSS CRC mouse model, and then FOLFOX regimen can induce PD-L1 expression and high CD8+T lymphocyte infiltration in tumor microenvironment in tumor samples of colorectal cancer patients. Thus, the FOLFOX regimen has a synergistic effect with anti-PD-(L)1 monoclonal antibody.139 These preclinical data suggest that the combination of a PD-(L)1 inhibitor with an inducer of immunogenic cell death can improve immunotherapy efficacy and support a therapeutic strategy of combining ICIs with chemotherapy such as FOLFOX regimens. Based on these data, a series of clinical studies were gradually carried out.
The METIMMOX study evaluated the efficacy of short-course repeat sequential oxaliplatin-based chemotherapy (FLOX) (oxaliplatin + fluorouracil) combined with nivolumab in first-line treatment of MSS mCRC. A total of 54 patients were enrolled, all of whom were RAS/BRAF mutation patients. ORR rates in the combined immunotherapy group and chemotherapy group were 48% and 23%, CR rates were 16% and 0%, respectively, indicating that immunotherapy combined chemotherapy could improve the chances of immunotherapy response for patients. However, the difference in median PFS between the two groups was not significant, 6.6 months and 5.6 months, respectively.33 KEYNOTE-651 evaluated the efficacy and safety of Pembrolizumab in combination with mFOLFOX7 or FOLFIRI in mCRC.34 The latest results showed that Pembrolizumab combined with mFOLFOX7 had an ORR of 61% for first-line therapy and Pembrolizumab combined with FOLFIRI had an ORR of 25% for second-line therapy. Both combination regimens showed antitumor activity in pMMR/MSS mCRC patients.
In addition, some studies have found that chemotherapy drugs related to DNA repair enzymes can produce better synergies with ICIs. O 6-methylguanine-DNA methyltransferase (MGMT) is designed to repair DNA damage caused by alkylating agents.140 If the tumor has MGMT gene silence (promoter methylation and protein expression loss), it will enhance the sensitivity of tumor cells to alkylate agents such as temozolomide (TMZ),141 and more secondary gene mutations may occur in the tumor after receiving TMZ, resulting in secondary high tumor mutation burden (TMB). The special synergistic mechanism makes TMZ a sensitizer for immunotherapy. Studies have reported that nearly 40% of mCRC patients have MGMT silencing.142 The MAYA study is based on this theory to explore a TMZ induction chemotherapy followed by combined immunotherapy strategy in MSS mCRC. This Phase II study included MSS mCRC patients who failed to respond to multi-line therapy and had silent MGMT expression, and received 2 cycles of TMZ induction chemotherapy followed by nivolumab combined with low-dose ipilimumab. The results showed that the median PFS and OS were 7.1 months and 18.5 months, respectively, and the ORR was as high as 39%; the incidence of grade 3 and above adverse reactions is low.35,36 This appears to be better than the results of a previous Phase II study of TMZ monotherapy in MGMT- silenced mCRC, which enrolled 41 patients with an ORR rate of only 10% and a median PFS and OS of only 1.9 and 5.1 months.143
ICIs Combined with Chemotherapy and Bevacizumab
The anti-angiogenic drug bevacizumab has been an important component of targeted therapy for mCRC and is approved for mCRC when used in combination with chemotherapy. By inhibiting the VEGF/VEGFR pathway, bevacizumab normalizes blood vessels, increases the infiltration of T lymphocytes, and activates immune effector cells by stimulating the maturation of dendritic cells and reducing the expansion of Tregs and MDSCs.144–146 Therefore, ICIs combined with chemotherapy and anti-angiogenesis targeted therapy have been the focus of research.
A Phase II placebo-controlled BACCI study evaluated the clinical activity of capecitabine, bevacizumab combined with atezolizumab in advanced refractory MSS mCRC, showing that the addition of atezolizumab failed to significantly improve PFS and OS.37 Another Phase II study evaluated the safety and tolerability of capecitabine, bevacizumab and pembrolizumab in the treatment of MSS mCRC. The ORR in the three-drug combination treatment group was 5%, which did not meet the predetermined target, but the median PFS and OS reached 4.3 and 9.6 months, respectively, and were tolerable.38 Meanwhile, after FOLFOX combined with bevacizumab induction therapy in MSS mCRC patients in MODUL Cohort 2 studies, atezolizumab is added to Fluoropyrimidine (FP) + bevacizumab as first-line maintenance therapy. The results showed that although no new safety signals were found, there was no improvement in efficacy.39,40
ICIs in combination with standard chemotherapy and bevacizumab is also being explored as a first-line treatment. The researchers first evaluated the efficacy of atezolizumab combined with bevacizumab and FOLFOX in the first-line treatment of MSS mCRC patients, with an ORR of 52% and a median PFS of 14.1 months, showing no significant activity compared with standard treatment.147 CHECKMATE-9X8 evaluated nivolumab combined with mFOLFOX6/ bevacizumab (NIVO+SOC) versus mFOLFOX6/ bevacizumab (SOC) for first-line treatment of mCRC. Most of the patients were MSS type. The ORR was 60% in the NIVO+SOC group and 46% in the SOC group, but the median PFS in both groups was 11.9 months, which did not meet the primary endpoint of PFS. However, after 12 months, the PFS rate in the NIVO + SOC group was higher than that in the SOC group.41
Two other clinical studies conducted in China have given some positive signals in terms of survival benefits. The Phase II BBCAPX study explored the efficacy of sintilimab combined with CAPEOX/bevacizumab in the first-line treatment of RAS mutation and MSS mCRC.42 The results showed that ORR and DCR were 84.0% and 100.0%, respectively. This efficacy data shows that the addition of immunotherapy improves efficacy by about 20% compared to the existing standard regimen. The results of follow-up were published in 2023, suggesting that the ORR and DCR of the combination treatment regimen remained stable, with a median PFS of 18.2 months, and were generally well tolerated.42 The study offers new hope for the up to 95% of MSS mCRC patients. However, the sample size of this study was small, only 25 people were included, and further large-scale studies are needed for verification. At present, the Phase III study of BBCAPX is in progress (NCT04194359). Another randomized, double-blind, phase 2 part of a phase 2/3 trial in China assessed the preliminary anti-tumor activity and safety of serplulimab plus HLX04 and XELOX as a treatment option for patients with MSS mCRC.148 The results showed that the ORR reached 65.5% and the median PFS reached 17.2 months. Although the median overall survival (OS) was not reached for either group, a trend of an OS benefit was observed for the serplulimab group (HR, 0.77; 95% CI, 0.41–1.45).
In addition, ICIs combined with three-drug chemotherapy and bevacizumab have also been explored for first-line treatment, equivalent to the use of the strongest induction chemotherapy, in order to kill more tumor cells and produce more neoantigens, so as to enhance the effect of immunotherapy and improve the survival outcome of patients. Recently, two related clinical studies (NIVACOR study and AtezoTRIBE study43,44 were published. Unfortunately, there was only some improvement in efficiency, but the survival benefit was not obviously. The single-arm Phase II NIVACOR study evaluated the efficacy of nivolumab combined with FOLFOXIRI/bevacizumab in first-line treatment of RAS/BRAF mutated MSS mCRC patients. The results showed that ORR and DCR were 78.9% and 96.2%, but mPFS were only 9.8 months.43 The AtezoTRIBE study is a multicenter, open-label, randomized, controlled phase 2 clinical trial that included unresectable mCRC patients regardless of MMR status. The results show that adding atezolizumab to first-line FOLFOXIRI/bevacizumab is safe. The median PFS of 13.1 months in the altazomab group was better than that of 11.5 months in the control group.44 At present, the OS data of AtezoTRIBE is still immature, and people are interested in further results of this study.
So far, immunotherapy combined with chemotherapy (including three drugs) and bevacizumab in the first-line treatment of MSS mCRC has shown very limited efficacy, and whether it can bring survival benefits to patients is still controversial. Further consideration should be given to the combination mode and related value of this treatment mode in the future. We look forward to the Phase III clinical study to obtain more data.
Dual ICIs Regimen Combined with Chemotherapy
The single-arm Phase 1b/2 MEDITREME trial evaluated whether the addition of durvalumab and tremelimumab to standard chemotherapy mFOLFOX6 improved therapeutic efficacy.45,46 Preliminary results showed that ORR and CR were 62.5% and 25%, respectively, and 1-year PFS were 50%, indicating that the above regimen was initially effective in treating MSS mCRC patients.47 In 2023, the results of the MEDETREME study were republished after long-term follow-up. Among them, 48 patients were MSS type, with a median PFS of 8.2 months, showing good safety. The study met its primary endpoint and achieved survival outcomes similar to those previously observed in bevacizumab combined chemotherapy regimens, which resulted in approximately 8 months of PFS.149 This study showed that the dual immunotherapy combined with standard chemotherapy was significantly better than chemotherapy alone or immunotherapy alone for MSS mCRC on two key efficacy indicators of ORR and PFS. Researchers have also explored predictive biomarkers, and the relevant information is expected to guide more accurate CRC combined immunotherapy strategies in the future.46
To sum up, the overall efficacy of ICIs combined with chemotherapy in MSS mCRC patients has been limited. Compared with previous phase III clinical studies of first-line or second-line chemotherapy combined with targeted therapy, there seems to be no obvious advantage in ORR and survival. In the context of targeted therapy being the front-line standard treatment, In the future, more exploration directions will focus on the triple regimen such as immunotherapy combined with chemotherapy and anti-vascular targeted therapy, as well as the “AtezoTRIBE” model with three-drug chemotherapy, and double immunotherapy combined with chemotherapy. The optimal combined model is worthy of further exploration.
ICIs Combined with Radiotherapy
The combination of radiotherapy and immunotherapy may have synergistic effects based on several mechanisms. Radiotherapy can induce tumor cell death, release tumor antigens and activate immune signals, thus promoting systemic immune response.150,151 Radiotherapy changed the tumor immune microenvironment and inhibited the immune escape of tumor cells, thus enhancing the killing ability of immune cells to tumor cells.152–154 Radiotherapy can also promote the diversification of tumor antigen pool, promote the antigen presentation of dendritic cells and stimulate the body’s specific immune response.152 Radiotherapy can induce the aggregation of TILs and the up-regulation of PD-L1 expression, thereby increasing the sensitivity of immunotherapy.155
Preclinical data suggest that the combination of ICIs with an inducer of immunogenic cell death, such as radiotherapy, improves the efficacy of immunotherapy.152,156 The synergistic effect of radiation therapy and ICIs has also been demonstrated in several studies of preclinical CRC tumor models.157,158 These mechanisms provide a basis for the clinical study of radiotherapy combined with immunotherapy for mCRC patients.
The ANAVA study enrolled 101 patients with locally advanced rectal cancer (LARC), and 96 patients who were pathologically evaluable underwent preoperative chemoradiotherapy (capecitabine +conventional fractionated long-term radiotherapy + avelumab). The results showed that 22 cases (23%) achieved pathological complete response (pCR),59 cases (61.5%) achieved obvious pathological remission, the rates of non-immune and immune-related adverse reactions above grade 3 were only 8% and 4%. There were 39 cases of MSS rectal cancer in the study, suggesting that radiotherapy combined with immunotherapy may be a potentially effective program for patients with MSS rectal cancer.48 The VOLTAGE-A study showed that 39 MSS LARC patients treated with nivolumab after the end of long-term concurrent chemoradiotherapy achieved 38% major pathological response (MPR) and 30% pathological complete response (pCR).49
A single-arm, non-randomized Phase II clinical trial (NCT03104439) enrolled 40 patients with MSS mCRC and administered nivolumab + ipilimumab + hypofractionated short-course radiotherapy. The results showed that the ORR and DCR were 10% and 25% respectively, and the mOS was 7.1 months, which indicated that radiotherapy combined with immunotherapy could improve the ICIs response rate and prolong the overall survival of MSS mCRC patients, providing a new treatment idea for MSS mCRC patients.50 However, in a Phase II study, 24 patients with chemotherapy-refractory MSS mCRC who received durvalumab combined with tremelimumab and radiotherapy achieved an 8.3% response rate, which did not meet the preset ORR of at least 25%, despite a median OS of 11.4 months.51 Another study explored the feasibility of Y90 liver radioembolization combined with durvalumab and tremelimumab for MSS mCRC and found that although the combination regimen was well tolerated by patients, there was no significant benefit.159 Similarly, Monjazeb et al found that the addition of hypofractionated radiotherapy to PD-1 and CTLA-4 inhibitors had no significant clinical benefit for MSS mCRC patients resistant to first-line chemotherapy.52 Therefore, the role of radiation therapy in overcoming therapeutic resistance to MSS mCRC remains to be further validated.
In summary, the preclinical model showed that combined radiotherapy on the basis of ICIs induced and enhanced systemic anti-tumor immune response. Relevant clinical studies have also confirmed that it can improve the pathological remission of MSS LARC patients, showing a relatively optimistic short-term efficacy. However, the safety and efficacy of this combination therapy model in MSS mCRC are not very consistent, and more large-scale prospective clinical trials are needed to further confirm it. At the same time, the choice of radiotherapy mode (including dose, segmentation, treatment order, etc.) is still inconclusive, and more studies are expected to further explore.
ICIs Combined with Targeted Drug
ICIs Combined with Anti-EGFR Monoclonal Antibody
Anti-EGFR therapy is the standard treatment for RAS wild-type mCRC. The Fc segment of cetuximab can bind to CD16 receptors on NK cells and dendritic cells, and direct killing of target cells by effector cells is mediated through antibody-dependent cell-mediated cytotoxicity (ADCC). Meanwhile, cetuximab can effectively improve the immune microenvironment and promote the infiltration of immune cells within the tumor.160,161 In addition, cetuximab alone or in combination with chemotherapy can induce immunogenic cell death (ICD) of colorectal cancer cells, thereby activating an effective anti-tumor response.162,163 Therefore, anti-EGFR monotherapy or combination chemotherapy and ICIs have a good synergistic mechanism basis.
Several studies have confirmed the initial efficacy of ICIs combined with anti-EGFR mab. The Phase II CAVE study included RAS wild-type mCRC patients treated with cetuximab, and 92% of enrolled patients had MSS status. When cetuximab and Avelumab were combined for third-line treatment, the median PFS and OS of the population were 3.6 months and 11.6 months, respectively, but the overall objective response rate was not high, only about 7%.53 An updated long-term follow-up of the study found that the median OS was 18.6 months. At the same time, the significant effectiveness of cetuximab rechallenge combined with Avelumab was further verified in patients with RAS wild-type mCRC based on ctDNA cues.54
Another AVETUXIRI study was A Phase II study of Avelumab combined with cetuximab combined with irinotecan in the treatment of refractory MSS mCRC, in which cohort A was RAS wild type and cohort B was RAS mutant. The interim analysis results showed that the ORR of cohort A and cohort B were 30% and 0%, respectively, which seemed to be quite different. However, 6-month PFS rates were 40% and 38.5%, and 1-year OS rates were 50% and 46.2%, respectively. Finally, the RAS wild-type cohort reached the primary efficacy endpoint, and the RAS mutant cohort also saw some survival benefits. These results also suggest that regardless of RAS status, ICIs combined with anti-EGFR mab can improve the outcome of immunotherapy to some extent.55 The study is ongoing. A similar study evaluated the efficacy and safety of tislelizumab in combination with cetuximab and irinotecan in previously treated RAS wild-type mCRC patients in a single-arm Phase II TEC trial.56 The results showed an ORR of 33%, exceeding the study’s preset value of 30%, and a DCR of 79%, and the median PFS and OS were 7.3 and 17.4 months respectively.
Immunotherapy combined with anti-EGFR mab in first-line therapy is also being explored. A single-arm, multicenter Phase II AVETUX trial evaluated avelumab in combination with cetuximab and mFOLFOX6 in previously untreated RAS/BRAF wild-type, MSI-independent mCRC patients.57 Among the 41 MSS mCRC patients, the 12-month PFS rate was 40% and the median PFS was 11.1 months. The overall ORR and DCR were 81% and 89%, respectively. This study preliminarily suggests that immunotherapy combined with anti-EGFR monoclonal antibody and chemotherapy as first-line treatment may further improve efficacy.
In addition, the Phase II AVETRIC study of cetuximab combined with Avelumab and mFOLFOXIRI three-drug chemotherapy regimen for first-line treatment of MSS RAS wild-type mCRC patients is ongoing and is expected to bring more tips on immunotherapy for advanced bowel tumors. There is also a phase II study that is changing ICIs from single to double immunotherapy, with a stronger combination therapy. In RAS wild-type and MSS type mCRC patients who had not been treated with anti-EGFR mab, the combination of panitumumab, ipilimumab, and nivolumab resulted in an ORR of 35%. This study showed that wild-type patients with RAS could have more choices and could try to adopt a combination regimen of anti-EGFR monoclonal antibody plus dual immunotherapy.58
In conclusion, the combination mode of anti-EGFR monoclonal antibody plus immunotherapy has a theoretical basis, and it also shows us better disease control. In particular, the effectiveness is more evident in the posterior treatment and in the RAS wild-type population, which is worthy of further exploration in the future.
ICIs Combined with Anti-EGFR Monoclonal Antibody and BRAF Inhibitor
Preclinical studies suggest possible synergies between BRAF inhibitors and PD-1/PD-L1 inhibitors. It has been reported that BRAF inhibitors combined with anti-EGFR mab can induce transient MSI-H phenotype in MSS colorectal cancer, suggesting that this combination model may bring better survival benefits for BRAF mutated MSS patients.164
A Phase I/II study (NCT04017650) evaluated the efficacy and safety of encorafenib (BRAF inhibitor) in combination with cetuximab and nivolumab in the treatment of BRAF V600E mutant MSS mCRC. A total of 26 patients were enrolled, and the ORR and DCR of 24 patients that could be evaluated reached 50% and 96%, respectively, and the median PFS and OS were 7.4 months and 15.1 months, respectively.59 This is the best outcome for second-line and above treatment in patients with BRAF mutations, and it is well tolerated, and the randomized Phase II SWOG 2107 study is also ongoing.
ICIs Combined with MAPK Signal Transduction Pathway Inhibitors
Overexpression and activation of mitogen-activated protein kinase (MAPK) pathways are often detected in mCRC, and dysregulation of these signal transduction pathways can be used as potential targets for cancer therapy.165 RAS, RAF, MEK and ERK proteins are key factors in this pathway. Some studies have shown that RAS-MAPK pathway can directly promote tumor cell proliferation,166 while BRAF mutations often lead to continuous activation of RAS/RAF/MEK/ERK pathway. Combined inhibition of upstream and downstream nodes of RAS/MAPK pathway is an important strategy to overcome drug resistance. Studies have found that the ORR of BRAF inhibitor monotherapy is only 0–5%,167,168 and the co-inhibition of BRAF and MEK contributes to the inhibition of MAPK pathway. Therefore, the combination of anti-EGFR drugs with BRAF inhibitor and MEK inhibitor is one of the treatment options for BRAF V600E mutant mCRC. However, the therapeutic effect is still limited.169–171
Based on the available clinical evidence, the US Food and Drug Administration (FDA) approved BRAF inhibitor encorafenib in combination with cetuximab for BRAFV600E mCRC. However, with an ORR of only 20% and a median PFS of only 4.3 months, the clinical benefit was not long-lasting.170,171
In the era of immunotherapy, more and more studies have found that the activation of RAS-MAPK pathway is also related to the decrease of T lymphocyte infiltration in tumors, while inhibition of MEK can enhance T lymphocyte infiltration in tumors and synergistic effect with PD-1 inhibitors to enhance anti-tumor activity.164,172,173 Therefore, on this basis, the exploration of clinical research has been carried out.
First, in a Phase IB study, Atezolizumab in combination with cobimetinib (a MEK inhibitor) achieved manageable safety and clinical activity in mCRC patients after standard treatment failure, with ORR and DCR of 17% and 39%, respectively. The 12-month PFS and OS rates were 11% and 43%, respectively.60,61 Unfortunately, the IMblaze370 Phase III study of atezolizumab in combination with cobimetinib, published in the Lancet, yielded negative results. Patients who had previously failed multiple lines of therapy were enrolled, and regorafenib was used as the standard control. Results showed that the efficacy of the combination regimen and regorafenib standard third-line therapy was similar, and atezolizumab alone or combined with cobimetinib achieved OS of 7.1 and 8.9 months, respectively, which was not significantly better than the standard treatment regorafenib monotherapy (OS of 8.5 months).62
In addition, durvalumab plus the MEK inhibitor trametinib had a similar effect in MSS mCRC patients, showing acceptable tolerability, but no improvement in OS was seen.63 A trial of another MEK inhibitor binimetinib combined with pembrolizumab combined with beizumab in mCRC patients who had failed multiline therapy showed that a total of 21 patients with pMMR/MSS mCRC were enrolled, with ORR and DCR of 12% and 94%, respectively. The median PFS was 6.4 months and the adverse reactions were tolerable.64 Studies have shown initial clinical efficacy, but Phase III clinical trials are needed to confirm this. Therefore, whether MEK inhibitor combined with immunotherapy has a synergistic effect remains to be confirmed by further studies.
The co-inhibition of BRAF and MEK contributes to the inhibition of MAPK pathway, so the combination of BRAF inhibitor and MEK inhibitor is also one of the treatment options for BRAF V600E mutant mCRC, although the efficacy is limited.169 Studies have shown that combining drugs targeting the RAS/MAPK pathway with immunotherapy may lead to an enhanced immune response, which may improve patient outcomes.65 A single-arm phase 2 clinical trial of combined PD-1, BRAF and MEK inhibition with sparatlizumab (PDR001), dabrafenib and trametinib in 37 patients with BRAFV600E CRC, met its primary end point. In 32 patients with microsatellite stabilization, ORR and DCR reached 25% and 75%, respectively, and durability that were favorable relative to historical controls of BRAF-targeted combinations alone, and the combination regimen was well tolerated.65
Based on the regulatory effect of RAS/MAPK pathway targeting drugs on tumor immune microenvironment, continuing to explore new combined strategies of RAS/MAPK pathway targeting drugs and immunotherapy will likely bring lasting survival benefits for tumor patients with RAS/MAPK pathway mutations.
ICIs Combined with KRAS-G12C Inhibitors
Kirsten rat sarcoma virus oncogene homology (KRAS) mutation is an important genetic driver of colorectal cancer (CRC).174,175 KRAS mutation can lead to activation of downstream signaling pathways, promote tumorigenesis and invasion, and thus affect the prognosis of colorectal cancer.176,177
Drug targeting of KRAS mutations is challenging. Although KRAS G12C inhibitors show significant clinical response after their emergence, the resistance mechanisms of KRAS G12C inhibitors are more complex and diverse. The main driving factors include secondary mutations of KRAS itself, reactivation of multiple nodes upstream and downstream of KRAS on the MAPK pathway, and immune deficiency.178 Therefore, combination therapy may be an effective means to overcome drug resistance.
Preclinical trials of KRAS G12C inhibitors in combination with PD-1/PD-L1 inhibitors are underway. In 2017, British scholars found that KRAS signal could inhibit T lymphocytes’ immune surveillance of tumor cells by up-regulating the protein expression of PD-L1 in tumor cells.179 It has been demonstrated in preclinical models that the KRAS-G12C inhibitor sotorasib can promote anti-tumor immunity by inhibiting PD-L1 signaling in tumors, by increasing and activating T cell infiltration and making tumor microenvironment (TME) a highly sensitive to immune checkpoint inhibitors. Therefore, the combination of KRAS-G12C inhibitor and immune checkpoint inhibitor can produce durable activity.180
Another KRAS G12C inhibitor MRTX849, had similar results when used in combination with anti-PD-1 antibodies. MRTX849 reversed the immunosuppressed tumor microenvironment, thereby sensitizing the tumor to ICIs therapy. The results showed that in a genetically engineered mouse (GEM) model, the combination of MRTX849 and anti-PD-1 antibodies improved progression-free survival compared with either drug alone.181
In summary, preclinical studies have found that the combination of immune checkpoint inhibitors with anti-EGFR mab, BRAF inhibitors, MAPK signaling pathway inhibitors, etc., has a good mechanism basis. At the same time, some clinical studies have shown that immunotherapy combined with these targeted therapies has a certain effect in the treatment of MSS colorectal cancer, showing a certain therapeutic prospect. Unfortunately, most of these existing studies are single-arm small sample exploratory studies, and the data direction is not completely stable and consistent, and more research results and long-term follow-up are still needed to prove the efficacy of immunotherapy combined with targeted therapy in MSS colorectal cancer.
ICIs Combined with Novel Development Drugs
ICIs Combined with Cancer Vaccines
Cancer vaccines are considered an option for immunotherapy in most solid tumors, including colorectal cancer. Several cancer vaccines have been studied, including autologous vaccines, dendritic cell vaccines, viral vector vaccines, and peptide-based vaccines, which stimulate anti-tumor immunity by containing tumor antigens, nucleic acids, whole cells, and peptides.182,183 These vaccines work by stimulating tumor antigen-specific cytotoxic T lymphocytes, which in turn recognize and potentially eliminate cancer cells in an antigen-specific manner.183,184 These results provide a theoretical basis for the combination of cancer vaccine and ICIs. Therapies involving cancer vaccines in combination with conventional treatments or different immunotherapies have been shown to be beneficial in combating tumor resistance and improving clinical outcomes, but clinical trials are still in the early stages and more research is needed to demonstrate their therapeutic potential.184–189
Oncolytic viruses (OVs) are cancer vaccines that use viruses as carriers to selectively infect tumor cells with natural or genetically modified viruses with replication ability, then induce tumor cell lysis and death, release cytokines and cell antigens, and make the tumor change from “cold” to “hot”. Thus, the anti-tumor effect of ICIs can be improved.190–194 Therefore, combining ICIs with OVs can enhance the anti-tumor ability of ICIs. OVs combined with PD-1/PD-L1 inhibitors has achieved good therapeutic effect in mouse animal models of colon cancer and ovarian cancer.195
The clinical trial of OVs drug PexaVec (a vaccinia virus) in combination with durvalumab for MSS colorectal cancer is ongoing, but it is still in the phase I/II study stage. Preliminary analysis shows that it is well tolerated, but the long-term curative effect has not been reported,196 which needs continuous attention.
In addition, avelumab plus autologous dendritic cell vaccine has entered clinical studies and has been shown to be effective and well tolerated in chemotherapy treated MSS mCRC.66 Although the primary endpoint of the study has not been met, the median PFS observed was 12.2 months, which was better than 5 months when anti-PD-1 monoclonal antibody was used as monotherapy.6
In conclusion, cancer vaccines may improve the efficacy of immunotherapy by altering the tumor microenvironment. To boost anti-tumor immunity, cancer vaccines in combination with currently used immunotherapy approaches such as ICIs are an attractive approach. This combination treatment approach can compensate for the limitations of each therapy when used alone, which also opens up the possibility for MSS metastatic colorectal cancer to benefit from immunotherapy.
ICIs Combined with Bispecific Antibody
Bispecific antibody (BsAb) is an artificial antibody that specifically binds to two antigens or epitopes at the same time, and can be prepared by cell fusion or recombinant DNA techniques. Due to its ability to simultaneously target two epitopes in tumor cells or TME, BsAb is gradually becoming an important component of the next generation of therapeutic antibodies.197,198 BsAb stimulate immune response by binding tumor-rich antigens (such as CEA, HER2, etc.) and immune cells, and their efficacy is being explored in different types of solid tumors,198,199 while BsAb in combination with ICIs can enhance anti-tumor activity.198,200,201
Cibisatamab is a T cell BsAb that targets both CEA on tumor cells and CD3 on T cells. Cibisatamab has shown effective antitumor activity in preclinical models, leading to increased intratumoral T cell infiltration and activation and upregulation of PD-1/PD-L1.200 Therefore, cibisatamab combined with PD-1/PD-L1 inhibitors may enhance the antitumor effect. In vitro tests have shown that anti-CEA BsAb combined with PD-1/PD-L1 inhibitors can kill tumor cells to the maximum extent.201 Considering that CEA is overexpressed in 80% of colorectal cancer and underexpressed in normal tissues, it can be used as an ideal tumor antigen for colorectal cancer. Clinical studies of BsAb for colorectal cancer are in the early stages. In two ongoing Phase I clinical studies, cibisatamab is being treated as a monotherapy or in combination with atezolizumab in patients with CEA expressing solid tumors (including MSS mCRC patients). Evidence of antitumor activity was observed with cibisatamab monotherapy at sustained dose escalation, and its combination with atezolizumab further enhanced antitumor activity with a manageable safety profile.67
Despite the potential therapeutic activity of BsAb in tumor therapy, its application still faces significant challenges, including tumor heterogeneity, difficult to control tumor microenvironment, insufficient co-stimulatory signaling to activate T cells, the need for continuous injection, fatal systemic side effects, and off-target toxic effects.
ICIs Combined with Other Novel Target Drugs
At present, the combination of ICIs with some novel target drugs (such as IL-17 inhibitors and TGF-β1 receptor inhibitors, etc.) has shown certain efficacy in basic studies, and is being further promoted in subsequent clinical studies.202–205
Interleukin-17a (also known as IL-17) is mainly secreted by T-helper lymphocyte-17 (Th17)206 and can up-regulate the expression of PD-L1 in multiple tumors. It induces the aggregation of MDSCs in the tumor, thereby leading to tumor immune escape and promoting tumor growth and invasion.202,203,207,208 Therefore, targeting IL-17 has become a promising strategy to overcome immunosuppression and improve the sensitivity of anti-PD-1 therapy.202,203 Inhibition of IL-17A has been observed to enhance tumor infiltration of lymphocytes in both colon and lung cancer tumor models in mice.204,209,210 It is suggested that IL-17A may be used as a therapeutic target, making MSS CRC patients sensitive to ICI treatment.204
Basic studies have shown that transforming growth factor-β (TGF-β1) signaling pathway can promote tumor growth, drug resistance and metastasis by inhibiting host immune response.211,212 At the same time, TGF-β1 was found to be the central mediator of immune tolerance in the tumor microenvironment of different cancer species, causing the immune escape of tumors,205 suggesting that inhibition of TGF-β1 pathway can help enhance the effect of immunotherapy and provide a new treatment strategy for the immunotherapy of solid tumors. It is expected that the combination of these novel target drugs will bring new hope for the treatment of bowel cancer.
In addition, existing studies have found that environmental factors, especially gut microbiome, can influence response to cancer immunotherapy. The gut microbiome plays a crucial role in various basic physiological and pathological processes. It has been found that in malignant melanoma and lung cancer, gut microbiome affects the human response to ICIs treatment, and it is the diversity and composition of intestinal microbiota that led to the different anti-tumor responses of patients against pd-1 monoclonal antibodies.213–216 Therefore, in order to improve the effectiveness of immunotherapy, the gut microbiome needs to be considered, especially in colorectal cancer, as it is closely related to the gut microbiome. These findings may provide a new therapeutic strategy for immunotherapy in patients with pMMR/MSS colorectal cancer.
To sum up, colorectal cancer is characterized by tumor complexity and heterogeneity, as well as the ability of tumor cells to evade immune surveillance through various means. Therefore, it is necessary to conduct personalized treatment targeting multiple targets and approaches to overcome the tumor evasion mechanism, so as to ensure satisfactory clinical results for different individuals. However, the development of these new therapeutic strategies, including cancer vaccines and specific double antibodies, has proven challenging, and these promising preclinical findings have not yet been translated into the clinic. Many factors contribute to this failure, including poor understanding of tumor biology and immunosuppressive tumor microenvironment, weak T cell response, and appropriate patient target selection.
Biomarkers for Predicting the Efficacy of ICIs in pMMR /MSS Colorectal Cancer
To date, dMMR/MSI-H status is an evidence-based biomarker for predictive ICIs benefit in mCRC.3,7,9,217 Finding a biomarker to predict the efficacy of ICIs in MSS colorectal cancer could enable more patients to benefit from immunotherapy, but this remains a clinical challenge. Several biomarkers other than dMMR/MSI-H are currently being studied as positive predictors of ICIs benefit in mCRC. These include TMB, mutations of polymerase ε and polymerase δ (POLE/POLD1), PD-L1 expression, tumor-infiltrating lymphocytes and immune scores, gut microbiome, etc.29,68–76
Tumor Mutational Burden
TMB refers to the total number of somatic mutations in each coding region of the tumor genome, which can measure all non-synonymous coding mutations in the tumor exome.218 TMB has been shown to be a molecular marker closely related to tumor immunotherapy efficacy in solid tumors, independent of MSI status and PD-1/PD-L1 expression.218,219 It is currently believed that higher TMB is associated with greater immunogenicity, which may enhance the antitumor activity of immunotherapy.220
The KEYNOTE158 study evaluated the efficacy of pembrolizumab therapy on previously treated patients with advanced, incurable solid tumors, and found that patients with high TMB (≥10 mutations per megabase) had significantly higher ORR than patients with low TMB (29% vs 6%). High tissue TMB (tTMB-high) may be a new and useful biomarker for predicting the efficacy of ICIs.221 Therefore, the US FDA has approved pembrolizumab for the treatment of refractory, advanced solid tumors with high TMB (≥10 mut/Mb) in tumor tissue.68
In colorectal cancer, only 16% of patients with high TMB colorectal cancer belong to MSI-H type, and the remaining 84% belong to MSS type. These patients with high TMB MSS colorectal cancer are likely to benefit from ICIs treatment.222 Fabrizio et al found that 2.9% of colorectal cancer patients with MSS were identified as having high TMB, and PD-1 inhibitors were shown to be effective in cases with concurrent MSS/ high TMB. TMB is superior to MSI status in predicting the efficacy of ICIs in colorectal cancer treatment, and this subgroup may expand the group of CRC patients who may benefit from ICIs based treatment.69
In the REGONIVO study, it was found that pMMR/MSS colorectal cancer patients with high TMB were more likely to benefit than patients with low TMB after failure of standard treatment with Regorafenib combined with nivolumab.22 In the follow-up molecular analysis of the CO.26 randomized Phase II trial, it was also found that MSS patients with TMB higher than 28 mts/Mb, the dual immunotherapy combination therapy can bring OS benefits, while patients with low TMB do not benefit. It was also found in the study that the proportion of plasma TMB higher than 28 mts/Mb in MSS patients was as high as 21%.29
However, the role of TMB in MSS mCRC has not been established, and there is currently no optimal mCRC-specific TMB cutoff to predict the benefit of ICIs-based treatment strategies. In the CO.26 study, if patients were divided into high and low TMB groups at a pre-specified cut-off point of 20 mts/Mb, the benefit of OS, PFS, or DCR could not be predicted.29 Another study also showed that a TMB cutoff point of 10 mut/Mb does not predict the benefit of ICIs-based treatment of pMMR/MSS for mCRC.223
In summary, TMB is a promising biomarker, and high TMB status is associated with immunotherapy benefit in MSS colorectal cancer, a subpopulation that may expand the population benefiting from ICIs-based therapies. However, there are still problems such as limited sample size of existing studies, different TMB analysis methods, and different optimal cutoff values for high TMB (9–28 mut/Mb),22,29,69 so the application of TMB as a biomarker to predict the efficacy of pMMR/MSS mCRC immunotherapy is still controversial.
Polymerase ε and Polymerase δ (POLE/POLD1) Mutations
DNA polymerase ε and polymerase δ (POLE/POLD1) genes encode the exonuclease domain of DNA polymerase, which is involved in DNA replication and proofreading. When the POLE/POLD1 gene is mutated, it may cause the dysfunction of the DNA damage repair system and lead to the increase of related neoantigens, making it have the characteristics of TMB-H.70,71,224–226 Relevant studies have shown that POLE/POLD1 mutations can predict the efficacy of ICIs in the treatment of non-small cell lung cancer, endometrial cancer and other solid tumors.71,227 POLE/POLD1 gene mutation has gradually become an independent biomarker for predicting the efficacy of pan-cancer immunotherapy.71
Studies in colorectal cancer have shown that tumors with POLE/POLD1 gene mutations are basically pMMR/MSS types, POLE mutant tumors exhibit higher CD8+ T cell infiltration and up-regulation of PD-1/PD-L1 and CTLA4 expression compared with POLE wild-type tumors, which are similar to dMMR/MSI-H tumors and therefore may be sensitive to ICIs treatment.70 However, this type of mutation only accounts for 1% of colorectal cancer patients.16,70 In the treatment of MSS mCRC, the predictive value of POLE/POLD1 mutation on ICIs efficacy has also been confirmed in clinical studies, and pembrolizumab has a significant effect on MSS metastatic colorectal cancer with POLE/POLD1 mutation.228,229 The combination of durvalumab and Avelumab has also shown promising clinical activity in POLE mutation mCRC patients. However, the clinical response to ICIs may be limited to patients with POLE exonuclease domain mutation (EDM).230,231 Other studies have shown that not all POLE/POLD1 mutations cause TMB-H, and that patients with POLE/POLD1 mutations without TMB-H may not benefit from immunotherapy. Subsequent retrospective studies have shown that POLE pathogenic mutations may be a key factor in the benefit of immunotherapy.232,233
Expression of PD-L1
PD-1 is a transmembrane protein that is expressed on the surface of activated T cells, B cells and natural killer cells. PD-L1 is a ligand of PD-1, which is usually expressed on the surface of tumor cells.234 In addition, PD-L1 is also expressed in lymphocytes, macrophages and stromal cells in the tumor immune microenvironment. The binding of PD-1 and PD-L1 can inhibit the proliferation and function of effector T cells, promote the transformation of effector T cells into regulatory T cells, and inhibit the apoptosis of tumor cells, thus escaping the surveillance and attack of the immune system.235
The high expression of PD-L1 in tumor tissues is correlated with the efficacy of immunotherapy in many solid tumors. In gastric cancer, non-small cell lung cancer and esophageal cancer, the PD-L-1 expression scoring system was used to predict the efficacy of ICIs.236–239 However, prospective studies in advanced colorectal cancer have shown that the expression of PD-L1 is not predictive of immune efficacy.240 At the same time, there is no correlation with dMMR/MSI-H.241
In a retrospective study, the association between PD-L1 expression and prognosis in patients with pMMR/MSS and dMMR/MSI-H CRC was investigated. Among 389 CRC patients, 68 were dMMR/MSI-H type, but PD-L1 expression was only confirmed to be associated with survival in the dMMR/MSI-H group, while no correlation was observed in the pMMR/MSS group.72 Another single-arm, multicenter Phase II AVETUX trial evaluating avelumab in combination with cetuximab and mFOLFOX6 in previously untreated RAS/BRAF wild-type, MSS mCRC patients also did not observe any correlation between PD-L1 expression and PFS.57
In conclusion, although PD-L1 expression has an important predictive role in other gastrointestinal tumors (such as gastric and esophageal cancers) when using ICIs, there is no evidence to date that PD-L1 expression can be used as a predictive biomarker in pMMR/MSS mCRC patients. In addition, the expression of PD-L1 is a dynamic biomarker that can be changed under drug treatment. Dynamic monitoring techniques to detect the expression of PD-L1 by circulating tumor cells can be explored in the future, and more evidence is needed to support the role of PD-L1 expression in predicting the efficacy of ICIs in colorectal cancer.
Tumor Infiltrating Lymphocytes and Immunoscore
There is usually infiltration of various lymphocytes in tumor parenchyma and stroma. These lymphocytes are collectively called TILs. TILs include T cells, B cells, NK cells, dendritic cells, macrophages and other subsets, and are an important part of tumor immune microenvironment. Existing studies have shown that anti- PD-1 /PD-L1 or anti-CTLA-4 treatment can induce increased CD8+ cytotoxic T lymphocyte infiltration in tumor, and these activated T cells may be related to tumor regression.242–244
Intratumor lymphocyte infiltration, especially the density of CD8+ T lymphocytes, is correlated with the prognosis of colorectal cancer.245 Masucci’s study showed that the infiltration of cytotoxic T cells and Th1 cells and the up-regulation of IFN-γ expression in colorectal cancer tissues could indicate the efficacy of ICIs, which also indicated that TILs could predict the efficacy of immunotherapy.246 This is consistent with the findings of Loupakis et al. TILs and TMB were detected in 85 dMMR/MSI-H colorectal cancer samples treated with ICIs, and the results showed that PFS and OS were significantly improved in the high TILs group, suggesting that TILs is a promising biomarker for predicting the efficacy of ICIs in dMMR/MSI-H mCRC.247
In recent years, the role of TILs in MSS colorectal cancer has been studied. Giannakis et al conducted whole exome sequencing on 619 colorectal cancers and found a significant association between high TILs and higher neoantigen load in MSS CRC, suggesting a possible sensitivity to ICIs treatment.73 However, previous studies have suggested that MSS CRC has low TMB and little TILs, and is not sensitive to immunotherapy.16 Interestingly, CD3+ CD8+ T lymphocytes were highly infiltrated in the microenvironment of approximately 45% of MSS CRC tumors,74 suggesting that some specific populations of MSS patients may respond to immunotherapy.16,248 NICOLE’s study showed that pMMR/MSS CRC patients could benefit from nivolumab. Further immune microenvironment analysis showed that tumor-infiltrating CD3+ and CD8+T lymphocytes may be biomarkers of immune response in pMMR CRC.249 Of course, unlike advanced CRC, the immune microenvironment in early CRC is relatively complete, and neoadjuvant immunotherapy may be able to better induce T lymphocyte proliferation and play a more significant curative effect.250–252 In addition, increased CD8+ T cell infiltration was associated with better survival outcomes in MSS mCRC patients treated with Regorafenib and avelumab.25
In addition, researchers have found that the combination of CD8+T lymphocyte infiltration and PD-L1 expression can be used to predict the efficacy of ICIs in the treatment of various tumors.253,254 Llosa et al found that MSS mCRC patients had a tumor immune microenvironment similar to dMMR/MSI-H patients when tumor tissue was infiltrated by highly expressed PD-L1 and highly expressed CD8+T cells, and patients could benefit from Pembrolizumab treatment. It is suggested that combined detection can predict the efficacy of ICIs in the treatment of MSS CRC patients.255 According to the analysis of tumor samples before and after neoadjuvant therapy, the VOLTAGE-A study showed that the pCR rate of patients with PD-L1 tumor ratio ≥1% was 75%, and the pCR rate of patients with CD8+T cells/effector regulatory T cells (CD8+/eTreg) ratio ≥2.5 in tumor infiltrating lymphocytes was 78%. The pCR rate was 100% when the above two conditions were met simultaneously. The results show that PD-L1 expression and increased CD8+/eTreg ratio are effective predictors of the benefit of immunocombined chemoradiotherapy in MSS locally advanced rectal cancer (LARC)patients.49
These studies suggest the potential of TILs as a predictor of the efficacy of ICIs in MSS CRC. In order to more accurately predict the efficacy of ICIs, the researchers proposed an immune score based on the distribution of TILs, which graded the number of CD8+T cells, CD3+T cells, and CD45RO+ memory T cells located in the core and border areas of the tumor. The density of lymphocyte populations in the two regions was rated from low to high as 1 to 4 points.256 The prognostic and predictive value of immune score based on the proportion of tumor immune cells in early and middle CRC is even higher than TNM stage and MSI status.74 Meanwhile, the prognostic value of immune score on clinical outcome of mCRC patients has also been confirmed.257 However, the predictive value of immunotherapy efficacy in mCRC has not yet been shown. At present, several studies have used immune scoring system to predict the efficacy of ICIs.75,183,258 AtezoTRIBE Study This study uses an immune score. pMMR/MSS patients who may benefit from the combination regimen of immunization were screened by the immune score. Studies have shown that patients with higher immune scores have more OS benefits than patients with lower immune scores,44 indicating that the immune score has the opportunity to help us screen out patients with pMMR/MSS CRC who may benefit from immunotherapy. More evidence is needed to confirm whether immune score can be used as a predictor of the efficacy of ICIs in MSS CRC.
Gut Microbiota, etc
Gut microbiota plays an important role in the maintenance and development of host immune system, and the efficacy of ICIs in the treatment of tumor is related to gut microbiota.259,260 A study has shown that the ratio of Prevotella to Bacteroides in the gut of patients with gastrointestinal tumors is related to the efficacy of ICIs, and whether they can benefit from immunotherapy is likely to be related to the metabolites of the bacteria.76 Another study showed that a microbiome containing 11 types of bacteria collected from the human gut was able to induce CD8+T lymphocytes capable of producing interferon gamma in the mouse gut, thereby enhancing the anti-tumor efficacy of ICIs.261 Wang et al found that Clostridium in the baseline intestinal microflora could be used as a marker to predict the efficacy of ICIs.26 In addition, it has been reported that in non-colorectal cancer tumors treated with ICIs, antibiotic use may alter the number and function of immune cells in the gut, and alterations in the gut microbiota may negatively affect immunotherapy efficacy.262,263
In addition to the gut microbiota, the microbiota is also present within colorectal cancer tumor tissue. Among them, there are more studies on Fusobacterium nucleatum, a tumor resident bacteria in colorectal cancer. Related studies have shown that Fusobacterium nucleatum can activate interferon signaling pathway, induce PD-L1 expression and increase IFN-γ+CD8+TILs, thereby improving tumor sensitivity to ICIs. When there is a high level of Fusobacterium nucleatum in the tumor tissue, the treatment effect of ICIs in patients is good, and MSI status does not need to be detected,264 suggesting that Fusobacterium nucleatum can predict the efficacy of ICIs in colorectal cancer, including MSS. In the future, we should gain a deeper understanding of the CRC gut microbiome and the biological mechanisms that influence colorectal cancer response to ICIs, which may guide us to recommend or avoid certain antibiotics during ICIs treatment for a tumor microenvironment with a better immune response.
In addition to the above markers, circulating tumor DNA (ctDNA) can better monitor the efficacy of ICIs in MSS colorectal cancer. Patients with low ctDNA levels are stable during treatment, while patients with elevated ctDNA levels often experience rapid progression.265 In addition, a blood analysis of patients with chemotherapy-refractory mCRC receiving a combination of cetuximab and avelumab showed that a high neutrophil to lymphocyte ratio was a poor prognostic factor. Therefore, the ratio of neutrophils to lymphocytes can also be used as a predictive marker of immunotherapy efficacy.266
In summary, several studies are attempting to optimize or expand immune benefits population by screening ICIs efficacy predictor markers in MSS colorectal cancer, and some biomarkers have shown better potential and value. Among them, TMB and POLE/POLD1 genes have been shown to have a good predictive effect on ICIs efficacy in MSS colorectal cancer, but there is no consensus on the critical values of predictive markers such as TMB and TILs, while related markers such as PD-L1 expression, immune score and intestinal microbiota are being explored for predicting the efficacy of immunotherapy. The results of some studies are inconsistent, and the specific mechanisms and how to evaluate them need to be further explored. Considering that there are still few relevant studies to predict the efficacy of immunotherapy in colorectal cancer with MSS, more clinical studies are needed in the future to verify and explore high-value biomarkers in order to screen out patients who are more suitable for immunotherapy.
Summary and Outlook
Immunotherapy offers hope for long-term survival in patients with MSI-H colorectal cancer, but current studies have shown that immunotherapy, especially ICIs alone, is largely ineffective in treating patients with pMMR/MSS mCRC. How to improve the efficacy of immunotherapy in patients with MSS mCRC has always been one of the urgent problems to be solved. The key challenge is to transform the pMMR/MSS type of tumor into a highly immunogenic tumor. At present, the combination therapy strategy is a universal strategy to increase immunotherapy responsiveness.
From the perspective of mechanism, considering the possible synergistic effect of the combination of different treatment regiments, some ICIs combined with multi-target TKI, chemotherapy, radiotherapy, anti-VEGF /EGFR targeted therapy and other strategies have been carried out related clinical studies, and survival benefits have been initially seen. At the same time, new combined treatment strategies such as immunotherapy and cancer vaccines, BsAb, gut microbiota transplantation and targeting other targets are also being explored, which is expected to obtain more and better clinical data in the future, and increase the treatment options of pMMR/MSS colorectal cancer. At present, our preclinical studies are not deep enough, and the safety and effectiveness need to be verified by more prospective clinical trials with large samples. The immune mechanism is very complex, which makes pMMR/MSS immunotherapy for colorectal cancer face two problems: one is efficacy, and the other is safety. Considering that each treatment plan has its own adverse reactions, multiple treatments will inevitably bring about the superposition of adverse reactions, how to reduce the unnecessary toxic side effects caused by the combination of drugs and prolong the reaction time of immunotherapy is also the focus of clinical consideration. Therefore, we believe that the choice of regimen is very important in the clinical study of immune combination therapy for MSS colorectal cancer. More basic research should be conducted in the future to evaluate whether the combination regimen can activate the immune microenvironment of tumors to achieve synergies.
In addition, we need to find people who can really benefit from the combination immunotherapy strategy. This requires us to screen biomarkers and further refine the population to screen out patients who are more suitable for immunotherapy. In fact, the search for predictive biomarkers remains a major challenge in immunotherapy in pMMR/MSS colorectal cancer patients, not only to screen those who can benefit from immunotherapy, but also to avoid the unnecessary expense, hyperprogression and possible severe toxicity of treating non-responders. But unlike the genetic mutations of targeted drugs, the molecular markers of immunotherapy are more complex. It can be seen that some markers, such as TMB and POLE/POLD1 genes, have shown good potential and value in predicting ICIs efficacy in pMMR/MSS colorectal cancer patients. However, any single predictive biomarker has limitations, especially for highly heterogeneous colorectal cancer. It is often necessary to integrate a variety of different types of biomarkers for comprehensive judgment, so as to improve the ability to predict.
Through our study, we believe that for pMMR/MSS colorectal cancer patients, it is necessary to further elucidate the key mechanisms of immune escape and immune tolerance in the future, guide the development of more effective immunotherapy methods or drugs, and promote further basic and translational research to develop more accurate and effective combination therapy protocols. CONSIDERING that precision therapy based on molecular typing will inevitably become the premise and basis of future clinical practice and research design, it is expected to explore more dimensions of biomarkers and their combinations in the future, so as to more accurately enrich the benefit groups of immune therapy, and provide more accurate treatment for pMMR/MSS colorectal cancer patients. This will bring longer survival time and better quality of life for patients.
Data Sharing Statement
No additional data are available.
Author Contributions
The authors listed in this article have made significant contributions to the work of the report, whether in concept, research design, execution, data acquisition, analysis and interpretation, or in all of these areas, and meet the following criteria. All authors participated in drafting or writing, or substantially revised or critically reviewed the article, and have agreed on the journal in which the article will be submitted. Finally, all authors agree to take responsibility and be accountable for the contents of the article.
Funding
No funding was received for this study.
Disclosure
The authors declare that there are no conflicts of interest regarding the publication of this paper.
References
1. Wei SC, Levine JH, Cogdill AP, et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell. 2017;170(6):1120–1133. doi:10.1016/j.cell.2017.07.024
2. Sadeghi Rad H, Monkman J, Warkiani ME, et al. Understanding the tumor microenvironment for effective immunotherapy. Med Res Rev. 2021;41(3):1474–1498. doi:10.1002/med.21765
3. Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357(6349):409–413.
4. Cercek A, Dos Santos Fernandes G, Roxburgh CS, et al. Mismatch repair-deficient rectal cancer and resistance to neoadjuvant chemotherapy. Clin Cancer Res. 2020;26(13):3271–3279. doi:10.1158/1078-0432
5. Barbari C, Fontaine T, Parajuli P, et al. Immunotherapies and combination strategies for immuno-oncology. Int J Mol Sci. 2020;21(14):5009. doi:10.3390/ijms21145009
6. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–2520. doi:10.1056/NEJMoa1500596
7. Overman MJ, Kopetz S, McDermott RS, 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(Suppl 15):abstr3501. doi:10.1200/JCO.2016.34.15_suppl.3501
8. Morse MA, Overman MJ, Hartman L, et al. Safety of nivolumab plus low-dose ipilimumab in previously treated microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer. Oncologist. 2019;24:1453–1461. doi:10.1634/theoncologist.2019-0129
9. André T, Shiu KK, Kim TW, et al. Pembrolizumab in microsatellite-instability-high advanced colorectal cancer. N Engl J Med. 2020;383(23):2207–2218. doi:10.1056/NEJMoa2017699
10. Diaz LA Jr, Shiu KK, Kim TW, et al. 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(5):659–670. doi:10.1016/S1470-2045(22)00197-8
11. Gorzo A, Galos D, Volovat SR, et al. Landscape of immunotherapy options for colorectal cancer: current knowledge and future perspectives beyond immune checkpoint blockade. Life. 2022;12(2):229. doi:10.3390/life12020229
12. Zheng Y, Fu Y, Wang PP, Ding ZY. Neoantigen: a promising target for the immunotherapy of colorectal cancer. Dis Markers. 2022;15(2022):8270305. doi:10.1155/2022/8270305
13. Narayanan S, Kawaguchi T, Peng X, et al. Tumor infiltrating lymphocytes and macrophages improve survival in microsatellite unstable colorectal cancer. Sci Rep. 2019;9(1):13455. doi:10.1038/s41598-019-49878-4
14. Baraibar I, Mirallas O, Saoudi N, et al. Combined treatment with immunotherapy-based strategies for MSS metastatic colorectal cancer. Cancers. 2021;13(24):6311. doi:10.3390/cancers13246311
15. Liu YT, Sun ZJ. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics. 2021;11(11):5365–5386. doi:10.7150/thno.58390
16. Wang C, Fakih M. Targeting MSS colorectal cancer with immunotherapy: are we turning the corner? Expert Opin Biol Ther. 2021;21(10):1347–1357. doi:10.1080/14712598.2021.1933940
17. Yuan J, Li J, Gao C, Jiang C, Xiang Z, Wu J. Immunotherapies catering to the unmet medical need of cold colorectal cancer. Front Immunol. 2022;5(13):1022190. doi:10.3389/fimmu.2022.1022190
18. O’Neil BH, Wallmark JM, Lorente D, et al. Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced colorectal carcinoma. PLoS One. 2017;12(12):e0189848. doi:10.1371/journal.pone.0189848
19. Ganesh K, Stadler ZK, Cercek A, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat Rev Gastroenterol Hepatol. 2019;16(6):361–375. doi:10.1038/s41575-019-0126-x
20. Wang M, Wang S, Desai J, Trapani LA, Neeson PJ. Therapeutic strategies to remodel immunologically cold tumors. Clin Transl Immunology. 2020;9(12):e1226. doi:10.1002/cti2.1226
21. Ciardiello D, Vitiello PP, Cardone C, et al. Immunotherapy of colorectal cancer: challenges for therapeutic efficacy. Cancer Treat Rev. 2019;76:22–32. doi:10.1016/j.ctrv.2019.04.003
22. Fukuoka S, Hara H, Takahashi N, 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(18):2053–2061.
23. Fakih M, Raghav KPS, Chang DZ, et al. Regorafenib plus nivolumab in patients with mismatch repair-proficient/microsatellite stable metastatic colorectal cancer: a single-arm, open-label, multicentre phase 2 study. EClin Med. 2023;6(58):101917. doi:10.1016/j.eclinm.2023.101917
24. Cousin S, Bellera CA, Guégan JP, et al. REGOMUNE: a phase II study of regorafenib plus avelumab in solid tumors—results of the non-MSI-H metastatic colorectal cancer (mCRC) cohort. J Clin Oncol. 2020;38(15_suppl):4019. doi:10.1200/JCO.2020.38.15_suppl.4019
25. Cousin S, Cantarel C, Guegan JP, et al. Regorafenib-Avelumab Combination in Patients with Microsatellite Stable Colorectal Cancer (REGOMUNE): a single-arm, open-label, Phase II trial. Clin Cancer Res. 2021;27(8):2139–2147. doi:10.1158/1078-0432.CCR-20-3416
26. Wang F, He MM, Yao YC, et al. Regorafenib plus toripalimab in patients with metastatic colorectal cancer: a phase Ib/II clinical trial and gut microbiome analysis. Cell Rep Med. 2021;2(9):100383. doi:10.1016/j.xcrm.2021.100383
27. Gomez-Roca CA, Yanez E, Im SA, et al. LEAP-005: a phase 2 multicohort study of lenvatinib plus pembrolizumab in patients with previously treated selected solid tumors-results from the colorectal cancer cohort. J Clin Oncol. 2021;39(15_suppl). doi:10.1200/JCO.2021.39.15_suppl.3564
28. Guo Y, Zhang W, Ying J, et al. Phase 1b/2 trial of fruquintinib plus sintilimab in treating advanced solid tumours: the dose-escalation and metastatic colorectal cancer cohort in the dose-expansion phases. Eur J Cancer. 2023;181:26–37.
29. Chen EX, Jonker DJ, Loree JM, et al. Effect of combined immune checkpoint inhibition vs best supportive care alone in patients with advanced colorectal cancer: the Canadian Cancer Trials Group CO.26 Study. JAMA Oncol. 2020;6(6):831–838. doi:10.1001/jamaoncol.2020.0910
30. Fakih MG, Sandhu J, Lim D, Li SM, Wang C. 320MO A phase I clinical trial of regorafenib, ipilimumab, and nivolumab (RIN) in chemotherapy resistant MSS metastatic colorectal cancer (mCRC). Ann Oncol. 2022;33:S684.
31. Bullock A, j G, Fakih M, et al. LBA O-9 Botensilimab, a novel innate/adaptive immune activator, plus balstilimab (anti-PD-1) for metastatic heavily pretreated microsatellite stable colorectal cancer. Ann Oncol. 2022;33:S376.
32. Garralda E, Sukari A, Lakhani NJ, et al. A first-in-human study of the anti-LAG-3 antibody favezelimab plus pembrolizumab in previously treated, advanced microsatellite stable colorectal cancer. ESMO Open. 2022;7(6):100639. doi:10.1016/j.esmoop.2022.100639
33. Ree AH, Hamre H, Kersten C, et al. Repeat sequential oxaliplatin-based chemotherapy (FLOX) and nivolumab versus FLOX alone as first-line treatment of microsatellite-stable (MSS) metastatic colorectal cancer (mCRC): initial results from the randomized METIMMOX study. J Clin Oncol. 2021;39(15_suppl):3556. doi:10.1200/JCO.2021.39.15_suppl.3556
34. Kim RD, Tehfe M, Kavan P, et al. Pembrolizumab plus mFOLFOX7 or FOLFIRI for microsatellite stable/mismatch repair-proficient metastatic colorectal cancer: KEYNOTE-651 Cohorts B and D. Clin Colorectal Cancer. 2024;23(2):118–127.e6. doi:10.1016/j.clcc.2024.03.001
35. Pietrantonio F, Morano F, Lonardi S, et al.MAYA trial: temozolomide (TMZ) priming followed by combination with low-dose ipilimumab and nivolumab in patients with microsatellite stable (MSS), MGMT silenced metastatic colorectal cancer (mCRC). Ann Oncol. 2021;32(5_suppl):S530–S582.
36. Morano F, Raimondi A, Pagani F, et al. Temozolomide followed by combination with low-dose ipilimumab and nivolumab in patients with microsatellite-stable, O-Methylguanine-DNA methyltransferase-silenced metastatic colorectal cancer: the MAYA Trial. J Clin Oncol off J Am Soc Clin Oncol. 2022;40(14):1562–1573. doi:10.1200/JCO.21.02583
37. Mettu NB, Niedzwiecki D, Boland PM, et al. BACCI: a phase II randomized, double-blind, placebo-controlled study of capecitabine bevacizumab plus atezolizumab versus capecitabine bevacizumab plus placebo in patients with refractory metastatic colorectal cancer. J Clin Oncol. 2018;36:
38. G BA, Wang R, Behr S, et al. Phase II study of pembrolizumab plus capecitabine and bevacizumab in microsatellite stable (MSS) metastatic colorectal cancer (mCRC): interim analysis. J Clin Oncol. 2021;40:16. doi:10.1200/JCO.2022.40.16_suppl.3565
39. Grothey A, Tabernero J, Winter F, 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;29:714–715. doi:10.1093/annonc/mdy424.020
40. Tabernero J, Grothey A, Arnold D, de Gramont A, Ducreux M. O’Dwyer P, et al.MODUL cohort 2: an adaptable, randomized, signal-seeking trial of fluoropyrimidine plus bevacizumab with or without atezolizumab maintenance therapy for BRAF (wt) metastatic colorectal cancer. ESMO Open. 2022;7:100559. doi:10.1016/j.esmoop.2022.100559
41. Lenz HJ, Parikh A, Spigel DR, et al. Modified FOLFOX6 plus bevacizumab with and without nivolumab for first-line treatment of metastatic colorectal cancer: phase 2 results from the CheckMate 9X8 randomized clinical trial. J Immunother Cancer. 2024;12(3):e008409. doi:10.1136/jitc-2023-008409
42. Fang X, Zhu N, Zhong C, et al. Sintilimab plus bevacizumab, oxaliplatin and capecitabine as first-line therapy in RAS-mutant, microsatellite stable, unresectable metastatic colorectal cancer: an open-label, single-arm, phase II trial. EClinicalMedicine. 2023;62:102123. doi:10.1016/j.eclinm.2023.102123
43. Damato A, Bergamo F, Antonuzzo L, 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. doi:10.1200/JCO.2022.40.16_suppl.3509
44. Antoniotti C, Rossini D, Pietrantonio F, et al. Upfront FOLFOXIRI plus bevacizumab with or without atezolizumab in the treatment of patients with metastatic colorectal cancer (AtezoTRIBE): a multicentre, open-label, randomised, controlled, phase 2 trial. Lancet Oncol. 2022;23(7):876–887. doi:10.1016/S1470-2045(22)00274-1
45. Fumet JD, Isambert N, Hervieu A, et al. Phase Ib/II trial evaluating the safety, tolerability and immunological activity of durvalumab (MEDI4736) (anti-PD-L1) plus tremelimumab (anti-CTLA-4) combined with FOLFOX in patients with metastatic colorectal cancer. ESMO Open. 2018;3(4):e000375. doi:10.1136/esmoopen-2018-000375
46. Ghiringhelli F, Chibaudel B, Taieb J, et al. Durvalumab and tremelimumab in combination with FOLFOX in patients with RAS mutated, microsatellite stable, previously untreated metastatic colorectal cancer (MCRC): results of the first intermediate analysis of the phase I b /II MEDETREME tria. J Clin Oncol. 2020;38(15_suppl):3006. doi:10.1200/JCO.2020.38.15_suppl.3006
47. Thibaudin M, Fumet JD, Chibaudel B, et al. First-line durvalumab and tremelimumab with chemotherapy in RAS-mutated metastatic colorectal cancer: a phase 1b/2 trial. Nat Med. 2023;29(8):2087–2098. doi:10.1038/s41591-023-02497-z
48. Salvatore L, Bensi M, Corallo S, et al. Phase II study of preoperative (PREOP) chemoradiotherapy (CTRT) plus avelumab (AVE) in patients (PTS) with locally advanced rectal cancer (LARC): the AVANA study. J Clin Oncol. 2021;39:3511. doi:10.1200/JCO.2021.39.15_suppl.3511
49. Bando H, Tsukada Y, Inamori K, et al. Preoperative chemoradiotherapy plus nivolumab before surgery in patients with microsatellite stable and microsatellite instability-high locally advanced rectal cancer. Clin Cancer Res. 2022;28(6):1136–1146.
50. Parikh AR, Szabolcs A, Allen JN, Clark JW, Wo JY, Raabe M, et al.Radiation therapy enhances immunotherapy response in microsatellite stable colorectal and pancreatic adenocarcinoma in a phase II trial. Nat Cancer. 2021;2(11):1124–1135. doi:10.1038/s43018-021-00269-7
51. Segal NH, Cercek A, Ku G, et al. Phase II single-arm study of durvalumab and tremelimumab with concurrent radiotherapy in patients with mismatch repair-proficient metastatic colorectal cancer. Clin Cancer Res. 2021;27(8):2200–2208. doi:10.1158/1078-0432.CCR-20-2474
52. Monjazeb A, Giobbie-Hurder A, Lako A, et al. Analysis of colorectal cancer patients treated on ETCTN 10021: a multicenter randomized trial of combined PD-L1 and CTLA-4 inhibition with targeted low-dose or hypofractionated radiation. J Clin Oncol. 2019;37:
53. Martinelli E, Martini G, Famiglietti V, et al. Cetuximab rechallenge plus avelumab in pretreated patients with RAS wild-type metastatic colorectal cancer: the phase 2 single-arm clinical CAVE trial. JAMA Oncol. 2021;7(10):1529–1535. doi:10.1001/jamaoncol.2021.2915
54. Martinelli E, Martini G, Ciardiello D, et al. Evidence of therapeutic effectiveness of third-line cetuximab rechallenge in appropriately selected patients: findings from long-term follow-up of CRICKET and CAVE trials. Ann Oncol. 2022;33(suppl4):S381–s382.
55. Huyghe N, Cuyper AD, Sinapi I, et al. Interim analysis of the phase II AVETUXIRI trial: avelumab combined with cetuximab and irinotecan for treatment of refractory microsatellite stable (MSS) metastatic colorectal cancer (mCRC). J Clin Oncol. 2022;40(suppl16):3595. doi:10.1200/JCO.2022.40.16_suppl.3595
56. Xu X, Ai L, Hu K, et al. Tislelizumab plus cetuximab and irinotecan in refractory microsatellite stable and RAS wild-type metastatic colorectal cancer: a single-arm phase 2 study. Nat Commun. 2024;15(1):7255. doi:10.1038/s41467-024-51536-x
57. Stein A, Simnica D, Schultheiß C, et al. PD-L1 targeting and subclonal immune escape mediated by PD-L1 mutations in metastatic colorectal cancer. J Immunother Cancer. 2021;9(7):e002844. doi:10.1136/jitc-2021-002844
58. Lee MS, Loehrer PJ, Imanirad I, et al. Phase II study of ipilimumab, nivolumab, and panitumumab in patients with KRAS/NRAS/BRAF wild-type (WT) microsatellite stable (MSS) metastatic colorectal cancer (mCRC). J Clin Oncol. 2021;39(3 suppl):7. doi:10.1200/JCO.2021.39.3_suppl.7
59. Morris VK, M PC, Escano M, 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(4_suppl):12. doi:10.1200/JCO.2022.40.4_suppl.012
60. Bendell JC, Kim TW, Goh BC, et al. Clinical activity and safety of cobimetinib (cobi) and atezolizumab in colorectal cancer (CRC). J Clin Oncol. 2016;34(15_suppl):3502. doi:10.1200/JCO.2016.34.15_suppl.3502
61. Hellmann MD, Kim TW, LeeCB, et al. Phase Ib study of atezolizumab combined with cobimetinib in patients with solid tumors. Ann Oncol. 2019;30(7):1134–1142. doi:10.1093/annonc/mdz113
62. 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(6):849–861. doi:10.1016/S1470-2045(19)30027-0
63. Johnson B, Haymaker CL, Parra ER, 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(8):e005332. doi:10.1136/jitc-2022-005332
64. Lieu CH, Davis SL, Leong S, et al. Results from the safety lead in for a phase II study of pembrolizumab in combination with binimetinib and bevacizumab in patients with refractory metastatic colorectal cancer (mCRC). J Clin Oncol. 2020;38(15 suppl):4031. doi:10.1200/JCO.2020.38.15_suppl.4031
65. Tian J, Chen JH, Chao SX, et al. Combined PD-1, BRAF and MEK inhibition in BRAFV600E colorectal cancer: a phase 2 trial. Nat Med. 2023;29(2):458–466. doi:10.1038/s41591-022-02181-8
66. Español-Rego M, Fernández-Martos C, Elez E, et al. A Phase I-II multicenter trial with Avelumab plus autologous dendritic cell vaccine in pre-treated mismatch repair-proficient (MSS) metastatic colorectal cancer patients; GEMCAD 1602 Study. Cancer Immunol Immunother. 2023;72(4):827–840. doi:10.1007/s00262-022-03283-5
67. Tabernero J, Melero I, Ros W, et al. Phase Ia and Ib studies of the novel carcinoembryonic Antigen (CEA) T-cell bispecific (CEA CD3 TCB) antibody as a single agent and in combination with atezolizumab: preliminary efficacy and safety in patients with metastatic colorectal cancer (mCRC). J Clin Oncol. 2017;35(15 suppl):3002. doi:10.1200/JCO.2017.35.15_suppl.3002
68. Subbiah V, Solit D, Chan T, Kurzrock R. The FDA approval of pembrolizumab for adult and pediatric patients with tumor mutational burden (TMB) ≥10: a decision centered on empowering patients and their physicians. Ann Oncol. 2020;31:1115–1118. doi:10.1016/j.annonc.2020.07.002
69. Fabrizio DA, TJ G Jr, Dunne RF, et al. Beyond microsatellite testing: assessment of tumor mutational burden identifies subsets of colorectal cancer who may respond to immune checkpoint inhibition. J Gastrointest Oncol. 2018;9(4):610–617. doi:10.21037/jgo.2018.05.06
70. Domingo E, Freeman-Mills L, Rayner E, et al. Somatic POLE proofreading domain mutation, immune response, and prognosis in colorectal cancer: a retrospective, pooled biomarker study. Lancet Gastroenterol Hepatol. 2016;1(3):207–216. doi:10.1016/S2468-1253(16)30014-0
71. Wang F, Zhao Q, Wang YN, et al. Evaluation of POLE and POLD1 mutations as biomarkers for immunotherapy outcomes across multiple cancer types. JAMA Oncol. 2019;5(10):1504–1506. doi:10.1001/jamaoncol
72. Lee LH, Cavalcanti MS, Segal NH, et al. Patterns and prognostic relevance of PD-1 and PD-L1 expression in colorectal carcinoma. Mod Pathol. 2016;29:1433–1442. doi:10.1038/modpathol.2016.139
73. Giannakis M, Mu XJ, Shukla SA, et al. Genomic correlates of immune-cell infiltrates in colorectal carcinoma. Cell Rep. 2016;15(4):857–865. doi:10.1016/j.celrep.2016.03.075
74. Pagès F, Mlecnik B, Marliot F, et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet. 2018;391(10135):2128–2139. doi:10.1016/S0140-6736(18)30789-X
75. Malka D, Lièvre A, André T, et al. Immune scores in colorectal cancer: where are we? Eur J Cancer. 2020;140:105–118. doi:10.1016/j.ejca.2020.08.024
76. Peng Z, Cheng S, Kou Y, et al. The gut microbiome is associated with clinical response to anti-PD-1/PD-L1 immunotherapy in gastrointestinal cancer. Cancer Immunol Res. 2020;8(10):1251–1261. doi:10.1158/2326-6066.CIR-19-1014
77. Chen F, Zhuang X, Lin L, et al. New horizons in tumor microenvironment biology: challenges and opportunities. BMC Med. 2015;5(13):45. doi:10.1186/s12916-015-0278-7
78. Roma-Rodrigues C, Mendes R, Baptista PV, Fernandes AR. Targeting tumor microenvironment for cancer therapy. Int J Mol Sci. 2019;20(4):840. doi:10.3390/ijms20040840
79. Yamaoka T, Kusumoto S, Ando K, Ohba M, Ohmori T. Receptor tyrosine kinase-targeted cancer therapy. Int J Mol Sci. 2018;19(11):3491. doi:10.3390/ijms19113491
80. Zwick E, Bange J, Ullrich A. Receptor tyrosine kinase signalling as a target for cancer intervention strategies. Endocr Relat Cancer. 2001;8(3):161–173. doi:10.1677/erc.0.0080161
81. Lin Z, Zhang Q, Luo W. Angiogenesis inhibitors as therapeutic agents in cancer: challenges and future directions. Eur J Pharmacol. 2016;15(793):76–81. doi:10.1016/j.ejphar.2016.10.039
82. Fukumura D, Kloepper J, Amoozgar Z, Duda DG, Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat Rev Clin Oncol. 2018;15(5):325–340. doi:10.1038/nrclinonc.2018.29
83. Yasuda S, Sho M, Yamato I, et al. Simultaneous blockade of programmed death 1 and vascular endothelial growth factor receptor 2 (VEGFR2) induces synergistic anti-tumour effect in vivo. Clin Exp Immunol. 2013;172(3):500–506. doi:10.1111/cei.12069
84. Wilhelm SM, Dumas J, Adnane L, Lynch M, Carter CA. Schütz G, et al. Regorafenib (BAY 73-4506): a new oral multikinase inhibitor of angiogenic, stromal and oncogenic receptor tyrosine kinases with potent preclinical antitumor activity. Int J Cancer. 2011;129(1):245–255. doi:10.1002/ijc.25864
85. Ettrich TJ. Seufferlein T.Regorafenib. Recent Results Cancer Res. 2018;211:45–56. doi:10.1007/978-3-319-91442-8_3
86. Shigeta K, Matsui A, Kikuchi H, et al. Regorafenib combined with PD1 blockade increases CD8 T-cell infiltration by inducing CXCL10 expression in hepatocellular carcinoma. J Immunother Cancer. 2020;8(2):e001435. doi:10.1136/jitc-2020-001435
87. Zhai L, Spranger S, Binder DC, et al. Molecular pathways: targeting IDO1 and other tryptophan dioxygenases for cancer immunotherapy. Clin Cancer Res. 2015;21(24):5427–5433. doi:10.1158/1078-0432.CCR-15-0420
88. Li F, Zhang R, Li S, Liu J. IDO1: an important immunotherapy target in cancer treatment. Int Immunopharmacol. 2017;70–77. doi:10.1016/j.intimp.2017.03.024
89. Wu RY, Kong PF, Xia LP, et al. Regorafenib promotes antitumor immunity via inhibiting PD-L1 and IDO1 expression in melanoma. Clin Cancer Res. 2019;25(14):4530–4541. doi:10.1158/1078-0432.CCR-18-2840
90. Doleschel D, Hoff S, Koletnik S, Rix A, Zopf D, Kiessling F. Regorafenib enhances anti-PD1 immunotherapy efficacy in murine colorectal cancers and their combination prevents tumor regrowth. J Exp Clin Cancer Res. 2021;40(1):288. doi:10.1186/s13046-021-02043-0
91. Fakih M, Raghav KPS, Chang DZ, et al. Exploratory biomarker analyses of the single-arm, phase 2 study of regorafenib plus nivolumab in patients (pts) with mismatch repair-proficient (pMMR)/microsatellite stable (MSS) colorectal cancer (CRC). J Clin Oncol. 2022;40(4_suppl):89. doi:10.1200/JCO.2022.40.4_suppl.089
92. 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. Trial Lancet. 2013;381:303–312. doi:10.1016/S0140-6736(12)61900-X
93. Kato Y, Tabata K, Kimura T, et al. Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated macrophage and activation of the interferon pathway. PLoS One. 2019;14(2):e0212513. doi:10.1371/journal.pone.0212513
94. Sun Q, Zhou J, Zhang Z, et al. Discovery of fruquintinib, a potent and highly selective small molecule inhibitor of VEGFR 1, 2, 3 tyrosine kinases for cancer therapy. Cancer Biol Ther. 2014;15:1635–1645. doi:10.4161/15384047.2014.964087
95. Li J, Qin S, Xu RH, et al. Effect of fruquintinib vs placebo on overall survival in patients with previously treated metastatic colorectal cancer: the FRESCO randomized clinical trial. JAMA. 2018;319(24):2486–2496. doi:10.1001/jama.2018.7855
96. Mayer RJ, Van Cutsem E, Falcone A, et al.Randomized trial of Tas-102 for refractory metastatic colorectal cancer. N Engl J Med. 2015;372(20):1909–1919. doi:10.1056/NEJMoa1414325
97. Ruan H, Bu L, Hu Q, et al. Strategies of combination drug delivery for immune checkpoint blockades. Adv Healthc Mater. 2019;8(4):e1801099. doi:10.1002/adhm.201801099
98. Melero I, Berman DM, Aznar MA, et al. Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat Rev Cancer. 2015;15(8):457–472. doi:10.1038/nrc397
99. Das R, Verma R, Sznol M, et al. Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol. 2015;194(3):950–959. doi:10.4049/jimmunol.1401686
100. Boutros C, Tarhini A, Routier E, et al. Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat Rev Clin Oncol. 2016;13(8):473–486. doi:10.1038/nrclinonc.2016.58
101. Tawbi HA, Forsyth PA, Algazi A, et al. Combined nivolumab and ipilimumab in melanoma metastatic to the brain. N Engl J Med. 2018;379(8):722–730. doi:10.1056/NEJMoa1805453
102. Hellmann MD, Paz-Ares L, Bernabe Caro R, et al. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N Engl J Med. 2019;381(21):2020–2031. doi:10.1056/NEJMoa1910231
103. Ooki A, Shinozaki E, Yamaguchi K. Immunotherapy in colorectal cancer: current and future strategies. J Anus Rectum Colon. 2021;5:11–24. doi:10.23922/jarc.2020-064
104. Marie PK, Haymaker C, Parra ER, et al. Pilot clinical trial of perioperative durvalumab and tremelimumab in the treatment of resectable colorectal cancer liver metastases. Clin Cancer Res. 2021;27(11):3039–3049. doi:10.1158/1078-0432.CCR-21-0163
105. Datar I, Sanmamed MF, Wang J, et al. Expression analysis and significance of PD-1, LAG-3, and TIM-3 in human non–small cell lung cancer using spatially resolved and multiparametric single-cell analysis. Clin Cancer Res. 2019;25(15):4663–4673. doi:10.1158/1078-0432.CCR-18-4142
106. Blessin NC, Simon R, Kluth M, et al. Patterns of TIGIT expression in lymphatic tissue, inflammation, and cancer. Dis Markers. 2019;2019:5160565. doi:10.1155/2019/5160565
107. Kang CW, Dutta A, Chang LY, et al. Apoptosis of tumor infiltrating effector TIM-3+CD8+ T cells in colon cancer. Sci Rep. 2015;5(2015):15659. doi:10.1038/srep15659
108. Xie S, Huang J, Qiao Q, et al. Expression of the inhibitory B7 family molecule Vista in human colorectal carcinoma tumors. Cancer Immunol Immunother. 2018;67(11):1685–1694. doi:10.1007/s00262-018-2227-8
109. Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci USA. 2010;107(17):7875–7880. doi:10.1073/pnas.1003345107
110. Chauvin JM, Pagliano O, Fourcade J, et al. TIGIT and PD-1 impair tumor antigen-specific CD8⁺ T cells in melanoma patients. J. Clin Invest. 2015;125(5):2046–2058. doi:10.1172/JCI80445
111. Granier C, Vinatier E, Colin E, et al. Multiplexed immunofluorescence analysis and quantification of intratumoral PD-1+ Tim-3+ CD8+ T cells. J Vis Exp. 2018;8(132):56606. doi:10.3791/56606
112. Villarroel-Espindola F, Yu X, Datar I, et al. Spatially resolved and quantitative analysis of Vista/PD-1H as a novel immunotherapy target in human non-small cell lung cancer. Clin Cancer Res. 2018;24(7):1562–1573. doi:10.1158/1078-0432
113. Lee SJ, Jun S-Y, Lee IH, et al. CD274, LAG3, and IDO1 expressions in tumor-infiltrating immune cells as prognostic biomarker for patients with MSI-high colon cancer. J Cancer Res Clin Oncol. 2018;144:1005–1014. doi:10.1007/s00432-018-2620-x
114. Chen J, Chen Z. The effect of immune microenvironment on the progression and prognosis of colorectal cancer. Med Oncol. 2014;31(8):82. doi:10.1007/s12032-014-0082-9
115. Joller N, Lozano E, Burkett PR, et al. Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity. 2014;40(4):569–581. doi:10.1016/j.immuni.2014.02.012
116. Fuhrman CA, Yeh WI, Seay HR, et al. Divergent phenotypes of human regulatory T cells expressing the receptors TIGIT and CD226. J Immunol. 2015;195(1):145–155.
117. Harjunpää H, Guillerey C. TIGIT as an emerging immune checkpoint. Clin Exp Immunol. 2020;200(2):108–119. doi:10.1111/cei.13407
118. Chauvin JM, Zarour HM. TIGIT in cancer immunotherapy. J Immunother Cancer. 2020;8(2):e000957. doi:10.1136/jitc-2020-000957
119. Zhang Q, Bi J, Zheng X, et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat Immunol. 2018;19(7):723–732.
120. Johnston RJ, Comps-Agrar L, Hackney J, et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8+ T cell effector function. Cancer Cell. 2014;26(6):923–937. doi:10.1016/j.ccell.2014.10.018
121. Chiu DK, Yuen VW, Cheu JW, et al. Hepatocellular carcinoma cells up-regulate PVRL1, stabilizing PVR and inhibiting the cytotoxic T-Cell response via TIGIT to mediate tumor resistance to PD1 inhibitors in mice. Gastroenterology. 2020;159(2):609–623. doi:10.1053/j.gastro.2020.03.074
122. Banta KL, Xu X, Chitre AS, et al. Mechanistic convergence of the TIGIT and PD-1 inhibitory pathways necessitates co-blockade to optimize anti-tumor CD8(+) T cell responses. Immunity. 2022;55(3):512–526.e519. doi:10.1016/j.immuni.2022.02.005
123. Thibaudin M, Limagne E, Hampe L, Ballot E, Truntzer C, Ghiringhelli F. Targeting PD-L1 and TIGIT could restore intratumoral CD8 T cell function in human colorectal cancer. Cancer Immunol Immunother. 2022;71(10):2549–2563. doi:10.1007/s00262-022-03182-9
124. Kim TW, Bedard PL, LoRusso P, et al. Anti-TIGIT antibody tiragolumab alone or with atezolizumab in patients with advanced solid tumors: a Phase 1a/1b nonrandomized controlled trial. JAMA Oncol. 2023;9(11):1574–1582. doi:10.1001/jamaoncol.2023.3867
125. Niu J, Maurice-Dror C, Lee DH, et al. First-in-human phase 1 study of the anti-TIGIT antibody vibostolimab as monotherapy or with pembrolizumab for advanced solid tumors, including non-small-cell lung cancer. Ann Oncol. 2022;33(2):169–180. doi:10.1016/j.annonc.2021.11.002
126. Mettu NB, Ulahannan SV, Bendell JC, et al. A phase 1a/b open-label, dose-escalation study of etigilimab alone or in combination with nivolumab in patients with locally advanced or metastatic solid tumors. Clin Cancer Res. 2022;28(5):882–892. doi:10.1158/1078-0432.CCR-21-2780
127. Recondo G, Mezquita L. Tiragolumab and atezolizumab in patients with PD-L1 positive non-small-cell lung cancer. Lancet Oncol. 2022;23(6):695–697.
128. Du W, Yang M, Turner A, et al. TIM-3 as a target for cancer immunotherapy and mechanisms of action. Int J Mol. 2017;18(3):645. doi:10.3390/ijms18030645
129. Xu B, Yuan L, Gao Q, et al. Circulating and tumor-infiltrating Tim-3 in patients with colorectal cancer. Oncotarget. 2015;6(24):20592–20603. doi:10.18632/oncotarget.4112
130. Curigliano G, Gelderblom H, Mach N, et al. Abstract CT183: phase (Ph) I/II study of MBG453± spartalizumab (PDR001) in patients (pts) with advanced malignancies. Cancer Res. 2019;79(Suppl. 13):CT183.
131. Harding JJ, Patnaik A, Moreno V, et al. A phase Ia/Ib study of an anti-TIM-3 antibody (LY3321367) monotherapy or in combination with an anti-PD-L1 antibody (LY3300054): interim safety, efficacy, and pharmacokinetic findings in advanced cancers. J Clin Oncol. 2019;37(Suppl. 8):12. doi:10.1200/JCO.2019.37.8_suppl.12
132. Vanmeerbeek I, Sprooten J, De Ruysscher D, et al. Trial watch: chemotherapy-induced immunogenic cell death in immuno-oncology. Oncoimmunology. 2020. 9(1):
133. Zhang Z, Lu M, Qin Y, et al. Neoantigen: a new breakthrough in tumor immunotherapy. Front Immunol. 2021;12:672356. doi:10.3389/fimmu.2021.672356
134. Salas-Benito D, Pérez-Gracia L, Ponz-Sarvisé M, et al. Paradigms on immunotherapy combinations with chemotherapy. Cancer Discov. 2021;11(6):1353–1367. doi:10.1158/2159-8290.CD-20-1312
135. Cutsem EV, Nordlinger B, Cervantes A; ESMO Guidelines Working Group. Advanced colorectal cancer: ESMO clinical practice guidelines for treatment. Ann Oncol. 2010;:21:93–97. doi:10.1093/annonc/mdq222
136. Vincent J, Mignot G, Chalmin F, et al. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res. 2010:70,3052–3061. doi:10.1158/0008-5472.CAN-09-3690
137. Pfirschke P, Engblom C, Rickelt S, et al. Immunogenic chemotherapy sensitizes tumors to checkpoint blockade therapy. Immunity. 2016;44(2):343–54.doi. doi:10.1016/j.immuni.2015.11.024
138. Tesniere A, Schlemmer F, Boige V, et al. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene. 2010;29:482–491. doi:10.1038/onc.2009.356
139. Dosset M, Vargas TR, Lagrange A. PD-1/PD-L1 pathway: an adaptive immune resistance mechanism to immunogenic chemotherapy in colorectal cancer. Oncoimmunology. 2018;7(6):e1433981. doi:10.1080/2162402X.2018.1433981
140. Nifterik KAV, den Berg JV, der Meide WFV, et al. Absence of the MGMT protein as well as methylation of the MGMT promoter predict the sensitivity for temozolomide. Br J Cancer. 2010;103(1):29–35. doi:10.1038/sj.bjc.6605712
141. Newlands ES, Stevens MF, Wedge SR, Wheelhouse RT, Brock C. Temozolomide: a review of its discovery, chemical properties, pre-clinical development and clinical trials. Cancer Treat Rev. 1997;23(1):35–61. doi:10.1016/s0305-7372(97)90019-0
142. Esteller M, Herman JG. Generating mutations but providing chemosensitivity: the role of O6-methylguanine DNA methyltransferase in human cancer. Oncogene. 2004;23(1):1–8. doi:10.1038/sj.onc.1207316
143. Calegari MA, Inno A, Monterisi S, et al. A phase 2 study of temozolomide in pretreated metastatic colorectal cancer with MGMT promoter methylation. Br. J. Cancer. 2017;116(10):1279–1286. doi:10.1038/bjc.2017.109
144. Limagne E, Euvrard R, Thibaudin M, et al. Accumulation of MDSC and Th17 Cells in patients with metastatic colorectal cancer predicts the efficacy of a FOLFOX-bevacizumab drug treatment regimen. Cancer Res. 2016;76(18):5241–5252. doi:10.1158/0008-5472.CAN-15-3164
145. Lee WS, Yang H, Chon HJ, Kim C. Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity. Exp Mol Med. 2020;52(9):1475–1485. doi:10.1038/s12276-020-00500-y
146. 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. doi:10.3389/fimmu.2021.616837
147. Wallin J, Pishvaian MJ, Hernandez G, et al. Abstract 2651: clinical activity and immune correlates from a phase Ib study evaluating atezolizumab (anti-PDL1) in combination with FOLFOX and bevacizumab (anti-VEGF) in metastatic colorectal carcinoma. Cancer Res. 2016;76:2651. doi:10.1158/1538-7445.AM2016-2651
148. Wang Z, Peng J, Liang X, et al. First-line serplulimab in metastatic colorectal cancer: phase 2 results of a randomized, double-blind, phase 2/3 trial. Med. 2024:00213. doi:10.1016/j.medj.2024.05.009
149. Loupakis F, Cremolini C, Masi G, et al. Initial therapy with FOLFOXIRI and bevacizumab for metastatic colorectal cancer. N Engl J Med. 2014;371(17):1609–1618. doi:10.1056/NEJMoa1403108
150. Golden EB, Chhabra A, Chachoua A, et al. Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial. Lancet Oncol. 2015;16:795–803. doi:10.1016/S1470-2045(15)00054-6
151. Grassberger C, Ellsworth SG, Wilks MQ, et al.Assessing the interactions between radiotherapy and antitumour immunity. Nat Rev Clin Oncol. 2019;16(12):729–745. doi:10.1038/s41571-019-0238-9
152. Twyman-Saint Victor C, Rech AJ, Maity A, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015;520(2015):373–377. doi:10.1038/nature14292
153. Monjazeb AM, Schalper KA, Villarroel-Espindola F, et al.Effects of radiation on the tumor microenvironment. Semin Radiat Oncol. 2020;30(2):145. doi:10.1016/j.semradonc.2019.12.004
154. Zappasodi R, Merghoub T, Wolchok JD. Emerging concepts for immune checkpoint blockade-based combination therapies. Cancer Cell. 2018;33:581–598. doi:10.1016/j.ccell.2018.03.005
155. McLaughlin M, Patin EC, Pedersen M, et al. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat Rev Cancer. 2020;20(4):203–217. doi:10.1038/s41568-020-0246-1
156. Dovedi SJ, Lipowska-Bhalla G, Cheadle E, et al. Abstract 5034: the antitumor immune response generated by radiation therapy may be limited by tumor cell adaptive resistance and can be circumvented by PD-L1 blockade. Cancer Res. 2014;74:5034.
157. Deng L, Liang H, Burnette B, et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest. 2014;124(2):687–695. doi:10.1172/JCI67313
158. Dewan MZ, Galloway AE, Kawashima N, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res. 2009;15(17):5379–5388. doi:10.1158/1078-0432.CCR-09-0265
159. Floudas CS, Brar G, Mabry-Hrones D, et al. A pilot study of the PD-1 targeting agent AMP-224 used with low-dose cyclophosphamide and stereotactic body radiation therapy in patients with metastatic colorectal cancer. Clin Colorectal Cancer. 2019;18(4):e349–e360. doi:10.1016/j.clcc.2019.06.004
160. Inoue Y, Hazama S, Suzuki N, et al. Cetuximab strongly enhances immune cell infiltration into liver metastatic sites in colorectal cancer. Cancer Sci. 2017;108(3):455–460. doi:10.1111/cas.13162
161. Woolston A, Khan K, Spain G, Barber LJ, Griffiths B. et al. Genomic and transcriptomic determinants of therapy resistance and immune landscape evolution during anti-EGFR treatment in colorectal cancer. Cancer Cell. 2019;36(1):35–50.e9. doi:10.1016/j.ccell.2019.05.013
162. Pozzi C, Cuomo A, Spadoni I, et al. The EGFR-specific antibody cetuximab combined with chemotherapy triggers immunogenic cell death. Nat Med. 2016;22(6):624–631. doi:10.1038/nm.4078
163. Ferris RL, Lenz HJ, Trotta AM, et al. Rationale for combination of therapeutic antibodies targeting tumor cells and immune checkpoint receptors: harnessing innate and adaptive immunity through IgG1 isotype immune effector stimulation. Cancer Treat Rev. 2018;63. 48–60. doi:10.1016/j.ctrv.2017.11.008
164. Liu L, Mayes PA, Eastman S, et al. The BRAF and MEK inhibitors dabrafenib and trametinib: effects on immune function and in combination with immunomodulatory antibodies targeting PD-1, PD-L1, and CTLA-4. Clin Cancer Res. 2015;21(7):1639–1651. doi:10.1158/1078-0432.CCR-14-2339
165. Stefani C, Miricescu D, Stanescu-Spinu LL, Nica RL, Greabu M, Factors G. PI3K/AKT/mTOR and MAPK signaling pathways in colorectal cancer pathogenesis: where are we now?. Int J Mol Sci. 2021;22(19):10260. doi:10.3390/ijms2219102609
166. Sun Y, Liu WZ, Liu T, Feng X, Yang N, Zhou HF. Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. J Recept Signal Transduction Res. 2015;35(6):600–604. doi:10.3109/10799893.2015.1030412
167. Kopetz S, Desai J, Chan E, et al. Phase II pilot study of vemurafenib in patients with metastatic BRAF-mutated colorectal cancer. J Clin Oncol. 2015;33(34):4032–4038. doi:10.1200/JCO.2015.63.2497
168. Hyman DM, Puzanov L, Subbiah V, et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N Engl J Med. Med. 2015;373(8):726–736. doi:10.1056/NEJMoa1502309
169. Corcoran RB, Atreya CE, Falchook GS, et al. Combined BRAF and MEK inhibition with dabrafenib and trametinib in BRAF V600-mutant colorectal cancer. J Clin Oncol. 2015;33:4023–4031. doi:10.1200/JCO.2015.63.2471
170. Corcoran RB, André T, Atreya CE, et al. Combined BRAF, EGFR, and MEK inhibition in patients with BRAF(V600E)-mutant colorectal cancer. Cancer Discov. 2018;8:428–443. doi:10.1158/2159-8290.CD-17-1226
171. Kopetz S, Grothey A, Yaeger R, et al. Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer. N Engl J Med. 2019;381:1632–1643. doi:10.1056/NEJMoa1908075
172. 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):609–621. doi:10.1016/j.immuni.2016.01.024
173. Hirano H, Takashima A, Hamaguchi T, et al. Current status and perspectives of immune checkpoint inhibitors for colorectal cancer. Jpn J Clin Oncol. 2020;51(1):10–19. doi:10.1093/jjco/hyaa200
174. Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487(7407):330–337. doi:10.1038/nature11252
175. Prior IA, Hood FE, Hartley JL. The frequency of ras mutations in cancer. Cancer Res. 2020;80(14):2969–2974. doi:10.1158/0008-5472.CAN-19-3682
176. Pells S, Divjak M, Romanowski P, et al. Developmentally-regulated expression of murine K-ras isoforms. Oncogene. 1997;15(15):1781–1786. doi:10.1038/sj.onc.1201354
177. Drosten M, Dhawahir A, Sum EYM, et al. Genetic analysis of Ras signalling pathways in cell proliferation, migration and survival. EMBO J. 2010;29(6):1091–1104. doi:10.1038/emboj.2010.7
178. Zhu C, Guan X, Zhang X, et al. Targeting KRAS mutant cancers: from druggable therapy to drug resistance. Mol Cancer.2022;21(1):159. doi:10.1186/s12943-022-01629-2
179. Coelho MA, Carné Trécesson SD, Rana S, et al. Oncogenic RAS signaling promotes tumor immunoresistance by stabilizing PD-L1 mRNA. Immunity. 2017;47(6):1083–1099. doi:10.1016/j.immuni.2017.11.016
180. Canon J, Rex K, Saiki AY, et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019;575(7781):217–223. doi:10.1038/s41586-019-1694-1
181. Briere DM, Li S, Calinisan A, et al. The KRAS(G12C) inhibitor MRTX849 reconditions the tumor immune microenvironment and sensitizes tumors to checkpoint inhibitor therapy. Mol Cancer Ther. 2021;20:975–985. doi:10.1158/1535-7163.MCT-20-0462
182. Gupta M, Wahi A, Sharma P, et al. Recent advances in cancer vaccines: challenges, achievements, and futuristic prospects. Vaccines. 2022;10(12):2011. doi:10.3390/vaccines10122011
183. Picard E, Verschoor CP, Ma GW, Pawelec G. Relationships between immune landscapes, genetic subtypes and responses to immunotherapy in colorectal cancer. Front Immunol. 2020;6(11):369. doi:10.3389/fimmu.2020.00369
184. Keskin DB, Anandappa AJ, Sun J, et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature. 2019;565:234–239. doi:10.1038/s41586-018-0792-9
185. Kaufman HL, Lenz HJ, Marshall J, Singh D, Garett C, Cripps C. Combination chemotherapy and ALVAC-CEA/B7.1 vaccine in patients with metastatic colorectal cancer. Clin Cancer Res. 2008. 14:4843–4849. doi:10.1158/1078-0432.CCR-08-0276
186. Okuno K, Sugiura F, Hida JI, et al. Phase I clinical trial of a novel peptide vaccine in combination with UFT/LV for metastatic colorectal cancer. Exp Ther Med. 2011;2:73–79. doi:10.3892/etm.2010.182
187. Caballero-Baños M, Benitez-Ribas D, Tabera J, et al. Phase II randomised trial of autologous tumour lysate dendritic cell plus best supportive care compared with best supportive care in pre-treated advanced colorectal cancer patients. Eur J Cancer. 2016. 64:167–174. doi:10.1016/j.ejca.2016.06.008
188. Gatti-Mays ME, Strauss J, Donahue RN, et al. Phase I dose-escalation trial of BN-CV301, a recombinant poxviral vaccine targeting MUC1 and CEA with costimulatory molecules. Clin Cancer Res. 2019;25:4933–4944. doi:10.1158/1078-0432.CCR-19-0183
189. Ottensmeier CH, Delord JP, Lalanne A, et al. Safety and immunogenicity of TG4050: a personalized cancer vaccine in head and neck carcinoma. J Clin Oncol. 2023;41:6082. doi:10.1200/JCO.2023.41.16_suppl.6082
190. Gujar S, Pol JG, Kroemer G. Heating it up: oncolytic viruses make tumors ‘hot’ and suitable for checkpoint blockade immunotherapies. Oncoimmunology. 2018;7(8):
191. Ge Y, Wang H, Ren J, et al. Oncolytic vaccinia virus delivering tethered IL-12 enhances antitumor effects with improved safety. J Immunother Cancer. 2020;8(1):e000710.doi:10.1136/jitc–2020–000710.
192. Hemminki O. Oncolytic viruses for cancer immunotherapy. J Hematol Oncol. 2020;13(1):84. doi:10.1186/s13045-020-00922-1
193. Abd-Aziz N, Poh CL. Development of oncolytic viruses for cancer therapy. Transl Res. 2021;237:98–123. doi:10.1016/j.trsl.2021.04
194. Ripp J, Hentzen S, Saeed A.Oncolytic viruses as an adjunct to immune checkpoint inhibition. Front Biosci. 2022;27(5):151. doi:10.31083/j.fbl2705151
195. Liu Z, Ravindranathan R, Kalinski P, et al. Rational combination of oncolytic vaccinia virus and PD-L1 blockade works synergistically to enhance therapeutic efficacy. Nat Commun. 2017:14754. doi:10.1038/ncomms14754
196. Monge C, Xie C, Brar G, et al. A phase I/II study of JX-594 oncolytic virus in combination with immune checkpoint inhibition in refractory colorectal cancer. Eur J Cancer. 2020;138(2suppl):S57–S58. doi:10.1016/S0959-8049(20)31231-4
197. Li H, Saw PE, Song E. Challenges and strategies for next-generation bispecific antibody-based antitumor therapeutics. Cell Mol Immunol. 2020;17(5):451–461. doi:10.1038/s41423-020-0417-8
198. Wei J, Yang Y, Wang G, Liu M. Current landscape and future directions of bispecific antibodies in cancer immunotherapy. Front Immunol. 2022;28(13):1035276. doi:10.3389/fimmu.2022.1035276
199. Labrijn AF, Janmaat ML, Reichert JM, et al. Bispecific antibodies: a mechanistic review of the pipeline. Nat Rev Drug Discov. 2019;18(8):585–608.
200. Bacac M, Fauti T, Sam J, et al. A novel carcinoembryonic antigen T-Cell Bispecific Antibody (CEA TCB) for the treatment of solid tumors. Clin Cancer Res. 2016;22(13):3286–3297. doi:10.1158/1078-0432.CCR-15-1696
201. Osada T, Patel SP, Hammond SA, et al. CEA/CD3-bispecific T cell-engaging (BiTE) antibody-mediated T lymphocyte cytotoxicity maximized by inhibition of both PD1 and PD-L1. Cancer Immunol Immunother. 2015;64(6):677–688. doi:10.1007/s00262-015-1671-y
202. Moaaz M, Lotfy H, Motawea MA. Fadali G.The interplay of interleukin-17A and breast cancer tumor microenvironment as a novel immunotherapeutic approach to increase tumor immunogenicity. Immunobiology. 2021;226(2):152068. doi:10.1016/j.imbio.2021.152068
203. Li S, Na R, Li X, Zhang Y, Zheng T. Targeting interleukin-17 enhances tumor response to immune checkpoint inhibitors in colorectal cancer. Biochim Biophys Acta Rev Cancer. 2022;1877(4):188758. doi:10.1016/j.bbcan.2022.188758
204. Liu C, Liu R, Wang B, et al. Blocking IL-17A enhances tumor response to anti-PD-1 immunotherapy in microsatellite stable colorectal cancer. J Immunother Cancer. 2021;9(1):e001895. doi:10.1136/jitc-2020-001895
205. Metropulos AE, Munshi HG, Principe DR. The difficulty in translating the preclinical success of combined TGFβ and immune checkpoint inhibition to clinical trial. EBioMedicine. 2022:86:4380. doi:10.1016/j.ebiom.2022.104380
206. de Morales JMGR, Puig L, Daudén E, et al. Critical role of interleukin (IL)-17 in inflammatory and immune disorders: an updated review of the evidence focusing in controversies. Autoimmun Rev. 2020;19(1):102429. doi:10.1016/j.autrev.2019.102429
207. Welte T, Zhang XH. Interleukin-17 Could promote breast cancer progression at several stages of the disease. Mediators Inflamm. 2015;2015:804347. doi:10.1155/2015/804347
208. Chung AS, Wu X, Zhuang G, et al. An interleukin-17-mediated paracrine network promotes tumor resistance to anti-angiogenic therapy. Nat Med. 2013;19:1114. doi:10.1038/nm.3291
209. Hayata K, Iwahashi M, Ojima T, et al. Inhibition of IL-17A in tumor microenvironment augments cytotoxicity of tumor-infiltrating lymphocytes in tumor-bearing mice. PLoS One. 2013;8:e53131. doi:10.1371/journal.pone.0053131
210. Reppert S, Boross I, Koslowski M, et al. A role for T-bet-mediated tumour immune surveillance in anti-IL-17A treatment of lung cancer. Nat Commun. 2011;2:600. doi:10.1038/ncomms1609
211. Ciardiello D, Elez E, Tabernero J. Clinical development of therapies targeting TGFβ: current knowledge and future perspectives. Ann Oncol. 2020;31(10):1336–1349. doi:10.1016/j.annonc.2020.07.009
212. Derynck R, Turley SJ, Akhurst RJ. TGFβ biology in cancer progression and immunotherapy. Nat Rev Clin Oncol. 2021;18(1):9–34. doi:10.1038/s41571-020-0403-1
213. Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97.
214. Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103.
215. Matson V, Fessler J, Bao R, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–108.
216. Mager LF, Burkhard R, Pett N, et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science. 1481-1489;369(6510). doi:10.1126/science.abc3421
217. 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(9):1182–1191. doi:10.1016/S1470-2045(17)30422-9
218. Yarchoan M, Albacker LA, Hopkins AC, et al. PD-L1 expression and tumor mutational burden are independent biomarkers in most cancers. JCI Insight. 2019;4:e126908. doi:10.1172/jci.insight.126908
219. Fan A, Wang B, Wang X, et al. Immunotherapy in colorectal cancer: current achievements and future perspective. Int J Biol Sci. 2021;17:3837–3849. doi:10.7150/ijbs.64077
220. 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;30:1096–1103. doi:10.1093/annonc/mdz134
221. Marabelle A, Fakih M, Lopez J, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020;21(10):1353–1365. doi:10.1016/S1470-2045(20)30445-9
222. Goodman AM, Sokol ES, Frampton GM, Lippman SM, Kurzrock R. MicrosatelliteStable tumors with high mutational burden benefit from immunotherapy. Cancer Immunol Res. 2019;7(10):1570–1573. doi:10.1158/2326-6066.CIR-19-0149
223. Rousseau B, Foote MB, Maron SB, et al. The spectrum of benefit from checkpoint blockade in hypermutated tumors. N Engl J Med. 2021;384:1168–1170. doi:10.1056/NEJMc2031965
224. Heitzer E, Tomlinson I. Replicative DNA polymerase mutations in cancer. Curr Opin Genet Dev. 2014;24(100):107–113. doi:10.1016/j.gde.2013.12.005
225. Castellucci E, He T, Goldstein DY, Halmos B, Chuy J. DNA polymerase ε deficiency leading to an ultramutator phenotype: a novel clinically relevant entity. Oncologist. 2017;22:497–502. doi:10.1634/theoncologist.2017-0034
226. Mo S, Ma X, Li Y, et al. Somatic POLE exonuclease domain mutations elicit enhanced intratumoral immune responses in stage II colorectal cancer. J Immunother Cancer. 2020;8(2):e000881. doi:10.1136/jitc-2020-000881
227. Mehnert JM, Panda A, Zhong H, et al. Immune activation and response to pembrolizumab in POLE-mutant endometrial cancer. J Clin Invest. 2016;126(6):2334–2340. doi:10.1172/JCI84940
228. Gong J, Wang C, Lee PP, et al. Response to PD-1 blockade in microsatellite stable metastatic colorectal cancer harboring a POLE mutation. J Natl Compr Canc Netw. 2017;15(2):142–147. doi:10.6004/jnccn.2017.0016
229. Wang C, Gong J, Tu TY, et al. Immune profiling of microsatellite instability-high and polymerase ε (POLE)-mutated metastatic colorectal tumors identifies predictors of response to anti-PD-1 therapy. J Gastrointest Oncol. 2018;9(3):404–415. doi:10.21037/jgo.2018.01.09
230. Oh CR, Kim JE, Hong YS, et al. Phase II study of durvalumab monotherapy in patients with previously treated microsatellite instability-high/mismatch repair-deficient or POLE-mutated metastatic or unresectable colorectal cancer. Int J Cancer. 2022;150(12):2038–2045. doi:10.1002/ijc.33966
231. Kim JH, Kim SY, Baek JY, et al. Phase II study of avelumab monotherapy in patients with mismatch repair-deficient/microsatellite instability-high or POLE-mutated metastatic or unresectable colorectal cancer. Cancer Res Treat. 2020;52(4):1135–1144. doi:10.4143/crt.2020.218
232. Garmezy B, Gheeya J, Lin HY, et al. Clinical and molecular characterization of POLE mutations as predictive biomarkers of response to immune checkpoint inhibitors in advanced cancers. JCO Precis Oncol. 2022:e2100267. doi:10.1200/PO.21.00267
233. Rousseau B, Bieche I, Pasmant E, et al. PD-1 blockade in solid tumors with defects in polymerase epsilon. Cancer Discov. 2022;12:1435–1448. doi:10.1158/2159-8290.CD-21-0521
234. Gandini S, Massi D, Mandala M. PD-L1 expression in cancer patients receiving anti PD-1/PD-L1 antibodies: a systematic review and meta-analysis. Crit Rev Oncol Hematol. 2016;100:88–98. doi:10.1016/j.critrevonc.2016.02.001
235. Del Rio ML, Buhler L, Gibbons C, et al. PD-1/PD-L1, PD-1/PD-L2, and other co-inhibitory signaling pathways in transplantation. Transpl Int. 2008;21(11):1015–1028.
236. Kulangara K, Zhang N, Corigliano E, et al. Clinical utility of the combined positive score for programmed death ligand-1 expression and the approval of pembrolizumab for treatment of gastric cancer. Arch Pathol Labor Med. 2019;143(3):330–337. doi:10.5858/arpa.2018-0043-OA
237. Reck M, Rodriguez-Abreu D, G RA, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med. 2016;375(19):1823–1833. doi:10.1056/NEJMoa1606774
238. Takada K, Okamoto T, Toyokawa G, et al. The expression of PD-L1 protein as a prognostic factor in lung squamous cell carcinoma. Lung Cancer. 2017;104:7–15. doi:10.1016/j.lungcan
239. Baba Y, Nomoto D, Okadome K, et al. Tumor immune microenvironment and immune checkpoint inhibitors in esophageal squamous cell carcinoma. Cancer Sci. 2020;111(9):3132–3141. doi:10.1111/cas.14541
240. Overman MJ, Lonardi S, Leone F, et al. Nivolumab in patients with DNA mismatch repair deficient/microsatellite instability high metastatic colorectal cancer: update from CheckMate 142. J Clin Oncol. 2017;35(4_suppl):519. doi:10.1200/JCO.2017.35.4_suppl.519
241. Andre’T OM, Overman M, Lonardi S, et al. Analysis of tumor PDL1 expression and biomarkers in relation to clinical activity in patients (pts) with deficient DNA mismatch repair (dMMR)/high microsatellite instability (MSI-H) metastatic colorectal cancer (mCRC) treated with nivolumab (NIVO) ipilimumab (IPI): checkMate 142. Ann Oncol. 2017;28(Suppl_ 5):v158–v208. doi:10.1093/annonc/mdx393.011
242. Huang RR, Jalil J, Economou JS, et al. CTLA4 blockade induces frequent tumor infiltration by activated lymphocytes regardless of clinical responses in humans. Clinical Cancer Res. 2011;17(12):4101–4109. doi:10.1158/1078-0432.CCR-11-0407
243. Kvistborg P, Philips D, Kelderman S, et al. Anti-Ctla-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci Transl Med. 2014;6:ra128. doi:10.1126/scitranslmed.3008918
244. Robert L, Tsoi J, Wang X, et al. CTLA4 blockade broadens the peripheral T-cell receptor repertoire. Clin Cancer Res. 2014;20(9):2424–2432. doi:10.1158/1078-0432.CCR-13-2648
245. Huh JW, Lee JH, Kim HR. Prognostic significance of tumor-infiltrating lymphocytes for patients with colorectal cancer. Arch Surg. 2012;147(4):366–372. doi:10.1001/archsurg.2012.35
246. Masucci GV, Cesano A, Hawtin R, et al. Validation of biomarkers to predict response to immunotherapy in cancer: volume I-preanalytical and analytical validation. J Immunother Cancer. 2016;4:76. doi:10.1186/s40425-016-0178-1
247. Loupakis F, Depetris I, Biason P, et al. Prediction of benefit from checkpoint inhibitors in mismatch repair deficient metastatic colorectal cancer: role of tumor infiltrating lymphocytes. Oncologist. 2020:481–487. doi:10.1634/theoncologist.2019-0611
248. Goldstein JB, Wu W, Borras E, et al. Can microsatellite status of colorectal cancer be reliably assessed after neoadjuvant therapy? Clin Cancer Res. 2017;23:5246–5254. doi:10.1158/1078-0432.CCR-16-2994
249. Avallone A, Stefano AD, Pace U, et al.Neoadjuvant nivolumab in early stage colorectal cancer. Ann Oncol. 2020;31(Suppl 4):S449. doi:10.1016/j.annonc.2020.08.602
250. Oliveira AF, Bretes L, Furtado I. Review of PD-1/PD-L1 inhibitors in metastatic dMMR/MSI-H colorectal cancer. Front Oncol. 2019;396. doi:10.3389/fonc.2019.00396
251. Versluis JM, Long GV, Blank CU. Learning from clinical trials of neoadjuvant checkpoint blockade. Nat Med. 2020;26(4):475. doi:10.1038/s41591-020-0829-0
252. Zhang X, Wu T, Cai X, et al. Neoadjuvant immunotherapy for MSI-H/dMMR locally advanced colorectal cancer: new strategies and unveiled opportunities. Front Immunol. 2022;13:795972. doi:10.3389/fimmu.2022.795972
253. Nowicki TS, Akiyama R, Huang RR, et al. Infiltration of CD8 T cells and expression of PD-1 and PD-L1 in Synovial Sarcoma. Cancer Immunol Res. 2017;5(2):118–126. doi:10.1158/2326-6066.CIR-16-0148
254. Ogura A, Akiyoshi T, Yamamoto N, et al. Pattern of programmed cell death-ligand 1 expression and CD8-positive T-cell infiltration before and after chemoradiotherapy in rectal cancer. Eur J Cancer. 2018;91:11–20. doi:10.1016/j.ejca.2017.12.005
255. Llosa NJ, Luber B, Tam AJ, et al. Intratumoral adaptive immunosuppression and type 17 immunity in mismatch repair proficient colorectal tumors. Clin Cancer Res. 2019;25(17):5250–5259. doi:10.1158/1078-0432.CCR-19-0114
256. Galon J, Mlecnik B, Bindea G, et al. Towards the introduction of the ‘immunoscore’ in the classification of malignant tumours. J Pathol. 2014;232(2):199–209. doi:10.1002/path.4287
257. Mlecnik B, Tosolini M, Kirilovsky A, et al. Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction. J Clin Oncol. 2011;29:610–618. doi:10.1200/JCO.2010.30.5425
258. Angell HK, Bruni D, Barrett JC, et al. The immunoscore: colon cancer and beyond. Clin Cancer Res. 2020;26(2):332–339. doi:10.1158/1078-0432.CCR-18-1851
259. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol. 2009;9(5):313–323. doi:10.1038/nri2515
260. Caruso R, Lo BC, Núñez G. Host-microbiota interactions in inflammatory bowel disease. Nat Rev Immunol. 2020;20(7):411–426. doi:10.1038/s41577-019-0268-7
261. Tanoue T, Morita S, Plichta DR, et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature. 2019;565(7741):600–605. doi:10.1038/s41586-019-0878-z
262. Sun L, Zhang X, Zhang X, et al. Antibiotic-Induced disruption of gut microbiota alters local metabolomes and immune responses. Front Cell Infect Microbiol. 2019;24(9):99. doi:10.3389/fcimb.2019.00099
263. Cheng WY, Wu CY, Yu J. The role of gut microbiota in cancer treatment: friend or foe? Gut. 2020;69(10):1867–1876. doi:10.1136/gutjnl-2020-321153
264. Gao Y, Bi D, Xie R, et al. Fusobacterium nucleatum enhances the efficacy of PD-L1 blockade in colorectal cancer. Signal Transduct Target Ther. 2021;6(1):398. doi:10.1038/s41392-021-00795-x
265. Wang C, Chevalier D, Saluja J, et al. Regorafenib and nivolumab or pembrolizumab combination and circulating tumor DNA response assessment in refractory microsatellite stable colorectal cancer. Oncologist. 2020;25(8):e1188–e1194. doi:10.1634/theoncologist.2020-0161
266. Ciardiello D, Famiglietti V, Napolitano S, et al. Final results of the CAVE trial in RAS wild type metastatic colorectal cancer patients treated with cetuximab plus avelumab as rechallenge therapy: neutrophil to lymphocyte ratio predicts survival. Clin Colorectal Cancer. 2022;21. 141–148. doi:10.1016/j.clcc.2022.01.005
© 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.