Although a minority of colorectal cancers exhibit mismatch repair deficiency and associated sensitivity to immune checkpoint inhibitors (ICI), the vast majority of colorectal cancers arise in a tolerogenic microenvironment with mismatch repair proficiency, low tumor-intrinsic immunogenicity, and negligible immunotherapy responsiveness. Treatment strategies to augment tumor immunity with combination ICIs and chemotherapy have broadly failed in mismatch repair–proficient tumors. Similarly, although several small single-arm studies have shown that checkpoint blockade plus radiation or select tyrosine kinase inhibition may show improved outcomes compared with historical controls, this finding has not been clearly validated in randomized trials. An evolving next generation of intelligently engineered checkpoint inhibitors, bispecific T-cell engagers, and emerging CAR-T cell therapies may improve immunorecognition of colorectal tumors. Across these modalities, ongoing translational efforts to better define patient populations and biomarkers associated with immune response, as well as combine biologically sound and mutually amplifying therapies, show promise for a new era of immunotherapy in colorectal cancer.

Immune-modulating therapies have transformed the cancer treatment landscape. Immune-facilitated tumor responses are likely driven by two interacting disease features: (i) tumor-intrinsic foreignness derived from aberrant molecular alterations, and (ii) functionality of the tumor-extrinsic immune microenvironment (1–3). Highly immunogenic cancers, including non–small cell lung cancer (NSCLC), melanoma, and mismatch repair–deficient (MMRd) cancers, exhibit robust responses to immune checkpoint inhibition (ICI) of programmed cell death protein-1 (PD-1), programmed death ligand-1 (PD-L1), and cytotoxic T-lymphocyte–associated protein 4 (CTLA-4; refs. 4–7). However, this success is not seen in the vast majority of patients with mismatch repair–proficient (MMRp) colorectal cancers; immunotherapy is not approved for 95% of patients with colorectal cancer.

In this review, we analyze immunotherapy failures and promising signals to define an optimistic path for increasing immunotherapy benefit in colorectal cancer.

Increased immunogenicity of MMRd colorectal cancer

Immunotherapy efficacy requires recognition of molecular alterations that accompany cellular disorder. Tumor mutations can be translated into irregular peptides (neoantigens) which are presented for T-cell recognition via the MHC type I or type II (8–10). MMRd colorectal cancer is characterized by the loss of mismatch-repair protein(s) and the subsequent development of microsatellite instability, with accumulation of predominantly frameshift tumor mutations associated with high neoantigen burden. The landmark phase II clinical trial by Le and colleagues demonstrated that anti–PD-1 monotherapy provoked a 40% immune-related objective response rate (ORR) in heavily pretreated patients with MMRd cancers, including metastatic colorectal cancer (mCRC; ref. 11). The randomized, phase III KEYNOTE-177 trial demonstrated that first-line pembrolizumab (anti–PD-1) in MMRd mCRC led to a progression-free survival (PFS) benefit compared with first-line standard of care (16.5 months vs. 8.2 months; P = 0.0002; ref. 12). ORRs up to 65% were exhibited in a phase II study of nivolumab (anti–PD-1) and ipilimumab (anti–CTLA-4) in pretreated MMRd mCRC (13).

Early stage MMRd colorectal cancer tumors appear even more susceptible to ICIs. Complete response rates of 100% and 67% have been shown in phase II studies of stage II–III MMRd rectal or colon cancer, respectively (14–16). The explanation for this heightened efficacy is unknown, but may be related to favorable early microenvironmental features and/or a higher dependency of younger tumors on immune checkpoints for immune escape.

ICI in MMRp colorectal cancer

The outstanding success of ICI in MMRd colorectal cancer proves that sufficiently immunogenic colorectal cancer can be targeted by the immune system. However, for the 95% of patients with MMRp mCRC, ICI efficacy is poor.

MMRp mCRC tumor mutational burden (TMB) is on average half to 10-fold lower than tumors that exhibit high ICI response rates, such as melanoma, NSCLC and MMRd colorectal cancer (17, 18). No responses to anti–PD-1 monotherapy were witnessed in early studies of MMRp mCRC (11). An overall negative study of patients with MMRp mCRC randomized to combination durvalumab (anti–PD-L1) and tremelimumab (anti–CTLA-4) versus best supportive care showed that a subgroup of patients exhibited modestly increased survival in the ICI arm (6.6 months vs. 4.1 months; ref. 19). Notably, there was one objective response and no PFS difference in this small study.

One potential strategy to mitigate low MMRp mCRC immunogenicity is to select patients that exhibit tumor profiles akin to MMRd tumors, such as MMRd genomic signature patterns and/or especially high TMB. A retrospective analysis of 1,662 patients with advanced cancers, including colorectal cancer, determined that tumors in the top 20th TMB percentile per cancer type exhibited improved overall survival (OS) after ICI (17). Despite laudable efforts to define a predictive threshold to select ICI-appropriate cases, subsequent analyses have shown that this direct association between TMB and ICI-conferred OS disappears when only MMRp colorectal cancers are selected, with the rare exception of tumors harboring aberrant DNA polymerase δ and ε (POLD, POLE) that exhibit enormous mutational loads (1, 20, 21). TMB is a promising, but likely incomplete measurement of intrinsic tumor foreignness, and further controlled prospective studies are required to determine a threshold to predict ICI benefit in colorectal cancer. An increased focus on neoantigen qualities, such as peptide similarities to nonhuman infectious proteins and/or characteristics that increase the likelihood of HLA presentation and T-cell recognition, may better select disproportionately immunogenic tumors (22–24).

Tolerogenic colorectal cancer microenvironment

Immunotherapy success pan-cancer tends to disproportionately occur in tumors within a favorable “immune inflamed” microenvironment with infiltrating T cells and reduced stromal immunosuppressive signaling (2, 3). The colonic microenvironment is especially complex; functional digestion requires tolerance to commensal intestinal flora full of foreign antigens, swallowed material, and metabolic waste byproducts (25, 26). Higher microbial abundance and species diversity may enable especially pronounced immunotherapy responses in different cancer types (27, 28). The constellation of these external relationships integrated with intrinsic tumor aberrancy culminates in a diverse spectrum of overall colorectal cancer consensus molecular subtypes (CMS) from the functional, lymphocyte-dense MMRd signature (CMS1) to “T cell impaired” landscapes (CMS4; ref. 29). All colorectal cancer tumors and metastatic sites, however, may not exhibit the same degree of functional immunosuppression (30). Efforts to modify the local tumor-microenvironment interactions have been attempted to improve immunotherapy success in MMRp colorectal cancer.

Chemotherapy

The success of combination cytotoxic chemotherapy alongside ICI in multiple cancer types, including NSCLC and breast cancer, supports the growing hypothesis that chemotherapy may modulate the tumor environment and ICI-enabled disease control (31, 32). Cytotoxic chemotherapy, including oxaliplatin, may reduce immunosuppressive T-regulatory cells, induce cell surface death receptor density to facilitate immune-killing, and enhance neoantigen release in the microenvironment (33–35). This strategy has not been broadly successful in colorectal cancer (Fig. 1). First-line 5-FU, leucovorin, oxaliplatin, (FOLFOX) and bevacizumab with nivolumab provoked identical median PFS to chemotherapy alone, in a randomized phase II trial (36). However, the nivolumab arm exhibited higher response durability; 28% of patients in the chemo-ICI arm achieved 18-month PFS compared with 9% of patients with chemo, alone. Phase I/II single arm studies of first-line FOLFOX combined with immunotherapy, such as bevacizumab/ durvalumab/ oleclumab (COLUMBIA-1) or durvalumab/tremeliumumab (MEDITREME), have shown increases in response rates, but unclear PFS benefit compared with historical outcomes (37, 38). The phase II AtezoTRIBE study demonstrated that FOLFOXIRI/bevacizumab/atezolizumab modestly improves median PFS from 11.5 to 13.1 months (P = 0.018) compared with chemotherapy with no significant increase in response rate (38, 39).

Figure 1.

Overall response rates (ORR) of immunotherapy-based strategies in metastatic colorectal cancer in phase I–III clinical trials. Asterisk indicates that the study contained a control arm; in every instance, no statistical difference (NS) between ORRs was seen between the control and treatment arms. Pembro, pembrolizumab; Durva, durvalumab; Treme, tremelimumab; Nivo, nivolumab; Atezo, atezolizumab; Bev, bevacizumab; FOLFOX, 5-FU/ leucovorin/oxaliplatin; FOLFIRINOX, 5-FU/leucovorin/oxaliplatin/irinotecan; RT, radiotherapy; Ipi, ipilimumab; Cobi, cobimetinib; Rego, regorafenib; Lenva, lenvatinib; Cabo, cabozantinib; Enco, encorafenib; Cetux, cetuximab; TKI, tyrosine kinase inhibitor.

Figure 1.

Overall response rates (ORR) of immunotherapy-based strategies in metastatic colorectal cancer in phase I–III clinical trials. Asterisk indicates that the study contained a control arm; in every instance, no statistical difference (NS) between ORRs was seen between the control and treatment arms. Pembro, pembrolizumab; Durva, durvalumab; Treme, tremelimumab; Nivo, nivolumab; Atezo, atezolizumab; Bev, bevacizumab; FOLFOX, 5-FU/ leucovorin/oxaliplatin; FOLFIRINOX, 5-FU/leucovorin/oxaliplatin/irinotecan; RT, radiotherapy; Ipi, ipilimumab; Cobi, cobimetinib; Rego, regorafenib; Lenva, lenvatinib; Cabo, cabozantinib; Enco, encorafenib; Cetux, cetuximab; TKI, tyrosine kinase inhibitor.

Close modal

Radiotherapy

Many studies examine whether radiation improves immunotherapy responses. Rarely, an abscopal effect is documented where radiation of a single tumor site provokes a response in a non-radiated metastasis, supporting a hypothesis that radiation-facilitated tumor damage may lead to systemic immune response (40–43). A phase II study of durvalumab (anti–PD-L1) and tremelimumab (anti–CTLA-4) plus radiotherapy in chemo-refractory patients with MMRp mCRC did not meet its overall primary endpoint, yet responses were seen in distant non-radiated lesions in 8% (2/24) patients, accompanied by increased CD8 T cells, suggesting potential systemic immunomodulation (44). A similar phase II study of radiotherapy concurrent with ipilimumab (anti–CTLA-4) and nivolumab (anti–PD-1) defined a disease control rate in 10 of 40 (25%) of patients with mCRC in the intention to treat cohort (45). In the future, optimization of radiation timing, fractionation, and duration relative to immunotherapy may be warranted (43).

Signaling pathway inhibition

Tyrosine kinase inhibitors (TKI) and/or VEGF inhibition have been hypothesized to reduce immunosuppressive signaling by increasing dendritic cell maturation and priming (anti-VEGF, bevacizumab; ref. 46), enhancing pro-stimulatory interferon gamma responses (multi-TKI; lenvatinib; ref. 47), and decreasing secretion of pro-tumor cytokines (BRAF inhibitor; vemurafenib; refs. 33, 48).

In colorectal cancer, studies of co-TKI/ICI therapies demonstrate variable results (Table 1). Single arm studies of lenvatinib with pembrolizumab (49), and regorafenib with nivolumab (50) defined modest ORRs of 22% (7/32) and 7% (5/70), respectively. A separate phase I mCRC study of regorafenib, nivolumab and ipilimumab demonstrated an ORR of 31% (9/29) with a disease control rate of 65% (51). Response rates to single agent regorafenib, and single-agent or combination ICI in MMRp mCRC, are less than 1%, suggesting that these combinations may have synergistic activity (50, 52). A phase III study is ongoing to evaluate lenvatinib/pembrolizumab in MMRp mCRC (NCT04776148).

Table 1.

Combination ICI and tyrosine kinase pathway inhibitor therapies in MMRp metastatic colorectal cancer.

Study authors (Reference)Tyrosine kinase (Target)ICI (Target)Study designCohort sizeOverall response ratePFS (Months)OS (Months)
Eng et al (56Cobimetinib (MEK) Atezolizumab (PD-L1) Randomized phase III 183 3% 1.9 8.9 
Johnson et al (57Trametinib (MEK) Durvalumab (PD-1) Single-arm phase II 29 3% 3.2 6.9 
Barzi et al (96Regorafenib (multi-TKI) Pembrolizumab (PD-1) Single-arm phase I/II 73 0% 10.9 
Cousin et al (97Regorafenib (multi-TKI) Avelumab (PD-L1) Single-arm phase II 48 0% 3.6 10.8 
Fakih et al (50Regorafenib (multi-TKI) Nivolumab (PD-1) Single-arm phase II 70 7% 1.8 12 
Fakih et al (51Regorafenib (multi-TKI) Nivolumab (PD-1), Ipilimumab (CTLA-4) Single-arm phase I 29 31% 19.6 
Gomez-Roca et al (49Lenvatinib (multi-TKI) Pembrolizumab (PD-1) Single-arm phase II 32 22% 2.3 7.5 
Saeed et al (98Cabozantinib (multi-TKI) Durvalumab (PD-1) Single-arm phase II 36 28% 4.4 9.1 
Martinelli et al (99Cetuximab (EGFR) Nivolumab (PD-1) Single-arm phase II 71 8% 3.6 11.6 
Van Morris et al (54Cetuximab (EGFR), Encorafenib (BRAF) Nivolumab (PD-1) Single-arm phase I/II 26 45% 7.3 11.4 
Study authors (Reference)Tyrosine kinase (Target)ICI (Target)Study designCohort sizeOverall response ratePFS (Months)OS (Months)
Eng et al (56Cobimetinib (MEK) Atezolizumab (PD-L1) Randomized phase III 183 3% 1.9 8.9 
Johnson et al (57Trametinib (MEK) Durvalumab (PD-1) Single-arm phase II 29 3% 3.2 6.9 
Barzi et al (96Regorafenib (multi-TKI) Pembrolizumab (PD-1) Single-arm phase I/II 73 0% 10.9 
Cousin et al (97Regorafenib (multi-TKI) Avelumab (PD-L1) Single-arm phase II 48 0% 3.6 10.8 
Fakih et al (50Regorafenib (multi-TKI) Nivolumab (PD-1) Single-arm phase II 70 7% 1.8 12 
Fakih et al (51Regorafenib (multi-TKI) Nivolumab (PD-1), Ipilimumab (CTLA-4) Single-arm phase I 29 31% 19.6 
Gomez-Roca et al (49Lenvatinib (multi-TKI) Pembrolizumab (PD-1) Single-arm phase II 32 22% 2.3 7.5 
Saeed et al (98Cabozantinib (multi-TKI) Durvalumab (PD-1) Single-arm phase II 36 28% 4.4 9.1 
Martinelli et al (99Cetuximab (EGFR) Nivolumab (PD-1) Single-arm phase II 71 8% 3.6 11.6 
Van Morris et al (54Cetuximab (EGFR), Encorafenib (BRAF) Nivolumab (PD-1) Single-arm phase I/II 26 45% 7.3 11.4 

Colorectal cancers often exhibit mutations in MAPK signaling mediators. In vitro, inhibition of pathway modulators, including MEK1/2, can reprogram effector CD8 T cells into a regenerative, stem cell–like memory phenotype that may assist ICI (53). In patients who previously received anti-EGFR therapy, cetuximab (anti-EGFR) rechallenge with avelumab resulted in a relatively low ORR of 8.5% in a single-arm phase II study (39). For patients with BRAF V600E mutated MMRp mCRC, a single-arm phase I/II trial of encorafenib (BRAF inhibitor), cetuximab, and nivolumab demonstrated an ORR of 45%, higher than the conventional ORR of 20% seen in the TKI-combo, alone (54, 55). These results led to a randomized phase II trial (SWOG 2107) of encorafenib/cetuximab with or without nivolumab (NCT04017650). Similar efficacy with an ORR of 24% was seen with trametinib (MEK inhibitor), dabrafenib (BRAF inhibitor) plus PDR001 (anti–PD-1) in a phase II study (29).

Unfortunately, clinical activity of other MAPK/ICI combinations is limited. In the IMBlaze370 phase III trial, no OS advantage occurred with atezolizumab (anti–PD-L1) plus cobimetinib (MEK inhibitor) versus atezolizumab monotherapy or regorafenib (56). A phase II study of durvalumab plus trametinib in a similar population also failed: only one patient responded to therapy (57). Overall, the measured successes in single-arm studies paired with failures in multi-arm trials demonstrate that combination ICI plus conventional therapies appear unlikely to dramatically shift the landscape of success for colorectal cancer. Novel combinations and/or predictive biomarkers for patient selection are needed.

Expanded ICI

A next generation of potent ICI agents may improve T-cell recognition even in the setting of low, MMRp colorectal cancer immunogenicity (Fig. 2). Botensilimab, an Fc-enhanced next generation CTLA-4 antibody, is designed to promote intratumoral regulatory T-cell depletion via Fc gamma receptor signaling, activation of natural killer (NK) cells and macrophages, and T-cell enhancement via CTLA-4 blockade. Preliminary results from the phase IA/B study of botensilimab and balstilimab (anti–PD-1) have demonstrated an ORR of 24% (10/41) in patients with pretreated MMRp mCRC (58).

Figure 2.

Strategies to increase tumor immunorecognition. A, Next-generation combination ICIs. (B) Tumor neoantigen priming strategies. (C) Bispecific antibodies to facilitate tumor and T-cell engagement. (D) CAR-T cell therapies. Acronyms include antigen presenting cell (APC) and radiotherapy (RT). © 2023 Memorial Sloan-Kettering Cancer Center, Memorial Hospital for Cancer and Allied Diseases, and Sloan-Kettering Institute for Cancer Research, each in New York, NY. All rights reserved. Republished with permission.

Figure 2.

Strategies to increase tumor immunorecognition. A, Next-generation combination ICIs. (B) Tumor neoantigen priming strategies. (C) Bispecific antibodies to facilitate tumor and T-cell engagement. (D) CAR-T cell therapies. Acronyms include antigen presenting cell (APC) and radiotherapy (RT). © 2023 Memorial Sloan-Kettering Cancer Center, Memorial Hospital for Cancer and Allied Diseases, and Sloan-Kettering Institute for Cancer Research, each in New York, NY. All rights reserved. Republished with permission.

Close modal

Lymphocyte-activation gene 3 (LAG-3), an inhibitory immune checkpoint, is often co-expressed with immune checkpoint PD-1 on tumor-infiltrating lymphocytes; both contribute to T-cell exhaustion. LAG-3 inhibitors are approved in melanoma (59). In the dose confirmation phase of a multi-cohort, phase I study, favezelimab (anti–LAG-3) plus pembrolizumab or MK-4280A (favezelimab-pembrolizumab co-formulation), showed promising antitumor activity in MMRp colorectal cancer, compared with favezelimab alone, most notably in pts with PD-L1 CPS ≥ 1 tumors, with a 12-month survival rate of 50.6% compared with 29.5% in PD-L1 CPS low tumors (60). A randomized, phase III study of MK-4280A compared with standard of care in patients with PD-L1–positive colorectal cancer is underway (NCT05064059).

Vaccination to prime immunorecognition

Immunorecognition of the modest MMRp mCRC neoantigen load may be facilitated by vaccine priming towards common mCRC alterations. Phase I evaluation of BN-CV301, a poxvirus-engineered vaccine against MUC1 and CEA gastrointestinal antigens, induced T-cell responses against target antigens and provoked one partial response in mCRC (61). Similarly, a TP53 synthetic peptide vaccine induced T-cell specific anti-antigen responses in 9 of 10 treated patients with MMRp mCRC; in over half of the patients anti-TP53 activity was durable for over 6 months (62). Addition of the seven antigen PolyPEPI1018 vaccine to maintenance chemotherapy in patients with MMRp mCRC may facilitate immune-related tumor responses (63). However, despite promising preliminary data, cancer vaccines remain slow to translate into conventional cancer treatment. In a phase II randomized, placebo study of patients with mCRC given the anti-MUC1 tecemotide vaccine after liver resection, OS was not improved with the vaccine (64). In separate trials, FOLFIRI plus administration of anti-CEA or antitumor antigen 5T4 vaccines demonstrated promising initial antibody responses, yet limited durability (65, 66).

T-cell engagement with bispecific antibodies

An emerging strategy to redirect T cells involves the design of small molecules that create physical bridges between immune effector cells and tumor cell antigens. Bispecific antibodies are engineered to have separate binding sites directed against two different epitopes (67, 68). There are two major platforms of bispecific antibodies. IgG-like antibodies exhibit extended stability and solubility and possess Fc fragments which, depending on the design, may enable additional biological activity through antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity (69). Non-IgG-like bispecific antibodies are smaller due to absence of the Fc component (70), such as blinatumomab, which is approved for use in acute lymphoblastic leukemia (67, 71).

The evolution of bispecific antibody clinical design in colorectal cancer is ongoing with considerable variability in drug efficacy and tolerability. In a phase I study of 39 patients given MEDI-565, no objective responses were observed, high rates of antidrug antibodies were witnessed, and several patients developed severe inflammation-related adverse events (72).

Bispecific antibody re-optimization to reduce off-target inflammation and improve drug potency in later generations appears promising. The CEA-CD3 T-cell bispecific antibody cibisatamab (RO6958688) generated potent T-cell engagement and activation against CEA-positive tumor cells in vitro (73). Clinical evaluation of cibisatamab alone or in combination with atezolizumab (anti–PD-L1) in 31 patients with advanced colorectal cancer determined high rates of drug-assisted tumor inflammation (74). Three of 14 (21.5%) evaluable patients with colorectal cancer given the combination of cibisatamab and atezolizumab exhibited a partial response, and tumor bulk reduction in excess of 10% was seen in an additional 36% of patients (5/14). Additional evaluation of this drug combination in MMRp mCRC after obinutuzumab pretreatment is ongoing (NCT03866239).

Additional bispecific antibodies in development in mCRC include the anti-EGFR and the CD28 T-cell receptor engager (REGN7075) in combination with cemiplimab (anti–PD-1; NCT04626635); the guanylyl cyclase C (GUCY2C) and the CD3 T-cell receptor engager (PF-07062119) alone and in combination with sasanlimab (anti–PD-1) or anti-VEGF (NCT04171141); and, the fibroblast activation protein-a (FAP) and CD137/4–1BB T-cell and NK cell receptor engager RO7122290, in combination with cibisatamab after obinutuzumab pretreatment (NCT04826003), among others (Fig. 2).

Cellular therapies

Another modality to enable colorectal cancer immunorecognition includes the autologous transplant of T cells modified with chimeric antigen receptors (CAR) specific to an intrinsic colorectal cancer antigen (75). CAR T cells have been more successful in liquid versus solid cancers (76–78). Data for CAR T cells in colorectal cancer is limited. In a phase I mCRC study, anti-CEA CAR T-cell transplantation was well tolerated and enabled disease control in 4 of 10 patients (2 with tumor shrinkage; ref. 79). However, considerable challenges remain, as similar phase I investigations of CEA-CAM5-specific (80) and TAG-72 specific (81) CAR T cells demonstrated limited CAR T clonal longevity and minimal antitumor responses. GCC19CART, designed to overcome limitations of conventional CAR T cells in solid tumors, pairs CAR T cells that target guanylate cyclase-C (GCC) with CD19 targeting CAR T cells to amplify proliferation and activation of the solid tumor CAR T component. A phase I, investigator-initiated clinical trial in China reported results from two dose escalation cohorts, 1 × 106 or 2 × 106 CAR T cells/kg, demonstrating an ORR of 15.4% (2/13) and 50% (4/8), respectively (82). This therapy continues to be evaluated (NCT05319314).

Neoantigen induction

If therapies to improve intrinsic neoantigen recognition are insufficient, creative methods to: (i) induce increased native antigen expression and/or (ii) form new intrinsic antigens could transform MMRp mCRC into an MMRd molecular phenotype. Oncolytic viral-based platforms may be able to introduce new immunogenic tumor antigens into colorectal cancer to increase tumor immunogenicity (83, 84). Use of mutagenic agents, including temozolomide, may induce mutations and immune-related responses in a subset of patients with methylated methyl-guanine-methyl-transferase colorectal cancer in two phase II studies (85, 86).

Multiple studies across treatment modalities have shown that metastatic geography is an important feature for tumor immunoresponsiveness. In particular, across malignancy types, patients with liver metastases appear to exhibit worse immunotherapy outcomes (87–90). In the phase II study of patients with MMRp mCRC given combination regorafenib and nivolumab, 0 of 47 patients with liver metastases demonstrated a response, compared with 5 of 23 (22%) of patients with non-liver metastases (50). This was similarly observed in patients who received regorafenib with ipilimumab and nivolumab; 0 of 7 responses occurred in patients with liver metastases, compared with 9 of 22 (41%) of patients with non-liver metastases (47). Furthermore, in the combination botensilimab and balstilimab trial, an ORR of 42% (10/24) was seen patients without liver metastases, compared with 24% (10/41) in the overall population (26). These post hoc subgroup results provide optimism that future prospective studies enriched for patients without liver metastasis may show better ICI benefit. A randomized phase II study to compare botensilimab/balstilimab versus investigator's choice is underway in patients without liver metastasis (NCT05608044).

Explanations for these findings are under investigation. The liver is a tertiary immune organ with unique native antigen presenting cells which may create a tolerogenic microenvironment to prevent auto-immune reactions against filtered byproducts (87, 91–93). Notably, liver metastases confer a poor prognosis (94) and biological differences in the lower growth rates and reduced lethality of lung-predominant mCRC may confound observed trends.

Several broad patterns emerge from the history of immunotherapy in colorectal cancer that inform future efforts. MMRd colorectal cancer exhibit high TMB and sustained responses to ICI in early and metastatic settings. However, sustained antitumor responses are rarely observed in MMRp colorectal cancer across multiple trials of checkpoint inhibition, with or without the addition of chemotherapy, targeted therapies or radiotherapy. Tumor immunorecognition in MMRp colorectal cancer likely requires multi-modal activation and purposeful selection of complementary agents; several hopeful strategies have emerged.

T-cell redirecting bispecific antibodies can enable sustained T-cell engagement and heightened effector function within the colorectal cancer tumor microenvironment. Efforts to optimize T-cell redirecting therapies, and identify new surface targets and combination partners are especially promising. Next generation ICIs, such as the Fc-modified CTLA-4 inhibitor, botensilimab, and the LAG-3 inhibitor, favezelimab, have also shown preliminary signs of success, leading to randomized phase II and III studies

The mechanism of antigenic tolerance is not well defined, and heavy investment in immunobiology is needed to discover new targets. A shifting focus towards B-cell and Natural Killer therapies may help diversify the breadth of immune system activation to induce better immunological memory and decrease tolerogenic signals (95). In addition, close examination of MMRp mCRC immunotherapy responders may identify new biomarkers and facilitate a gradual transition to personalized immune-based regimens for especially immunogenic tumors. For all treatments, dosing and toxicity optimization will allow for more sustainable immune activation. Altogether, knowledge learned from prior immunotherapy failures and modest early successes encourages continued optimism for the future development of colorectal cancer immunotherapy.

G. Argilés reports personal fees from Gadeta BV and Amgen outside the submitted work. B. Rousseau reports grants and personal fees from Neophore during the conduct of the study; in addition, B. Rousseau has a patent for Methods and composition for cancer immunotherapy issued to US20230056846A1. N.H. Segal reports personal fees from Novartis, Puretech, Numab, AstraZeneca, GSK, ABL Bio, Revitope, Roche/Genentech, and Boehringer Ingelheim; grants from Roche/Genentech, Pfizer, Merck, BMS, AstraZeneca, Puretech, Immunocore, Regeneron, and Agenus; and other support from AstraZeneca and Regeneron outside the submitted work. No disclosures were reported by the other author.

MSK Cancer Center Support Grant/Core Grant (P30 CA008748).

1.
Rousseau
B
,
Foote
MB
,
Maron
SB
,
Diplas
BH
,
Lu
S
,
Argilés
G
, et al
.
The spectrum of benefit from checkpoint blockade in hypermutated tumors
.
N Engl J Med
2021
;
384
:
1168
70
.
2.
Trujillo
JA
,
Sweis
RF
,
Bao
R
,
Luke
JJ
.
T cell–inflamed versus Non-T cell–inflamed tumors: a conceptual framework for cancer immunotherapy drug development and combination therapy selection
.
Cancer Immunol Res
2018
;
6
:
990
1000
.
3.
Karasarides
M
,
Cogdill
AP
,
Robbins
PB
,
Bowden
M
,
Burton
EM
,
Butterfield
LH
, et al
.
Hallmarks of resistance to immune checkpoint inhibitors
.
Cancer Immunol Res
2022
;
10
:
372
83
.
4.
Wolchok
JD
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Rutkowski
P
,
Grob
J-J
,
Cowey
CL
, et al
.
Overall survival with combined nivolumab and ipilimumab in advanced melanoma
.
N Engl J Med
2017
;
377
:
1345
56
.
5.
Hodi
FS
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
J-J
,
Rutkowski
P
,
Cowey
CL
, et al
.
Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicenter, randomized, phase III trial
.
Lancet Oncol
2018
;
19
:
1480
92
.
6.
Hellmann
MD
,
Paz-Ares
L
,
Bernabe Caro
R
,
Zurawski
B
,
Kim
S-W
,
Carcereny Costa
E
, et al
.
Nivolumab plus ipilimumab in advanced non–small cell lung cancer
.
N Engl J Med
2019
;
381
:
2020
31
.
7.
Garon
EB
,
Rizvi
NA
,
Hui
R
,
Leighl
N
,
Balmanoukian
AS
,
Eder
JP
, et al
.
Pembrolizumab for the treatment of non–small cell lung cancer
.
N Engl J Med
2015
;
372
:
2018
28
.
8.
Segal
NH
,
Parsons
DW
,
Peggs
KS
,
Velculescu
V
,
Kinzler
KW
,
Vogelstein
B
, et al
.
Epitope landscape in breast and colorectal cancer
.
Cancer Res
2008
;
68
:
889
92
.
9.
Tran
E
,
Turcotte
S
,
Gros
A
,
Robbins
PF
,
Lu
Y-C
,
Dudley
ME
, et al
.
Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer
.
Science
2014
;
344
:
641
5
.
10.
Matsushita
H
,
Vesely
MD
,
Koboldt
DC
,
Rickert
CG
,
Uppaluri
R
,
Magrini
VJ
, et al
.
Cancer exome analysis reveals a T cell–dependent mechanism of cancer immunoediting
.
Nature
2012
;
482
:
400
4
.
11.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
.
PD-1 blockade in tumors with mismatch repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
12.
Andre
Y
,
Shiu
KK
,
Kim
TW
,
Jensen
BV
.
Pembrolizumab in microsatellite-instability–high advanced colorectal cancer
.
N Engl J Med
2020
;
383
:
2207
18
.
13.
André
T
,
Lonardi
S
,
Wong
KYM
,
Lenz
H-J
,
Gelsomino
F
,
Aglietta
M
, et al
.
Nivolumab plus low-dose ipilimumab in previously treated patients with microsatellite instability-high/mismatch repair–deficient metastatic colorectal cancer: 4-year follow-up from CheckMate 142
.
Ann Oncol
2022
;
33
:
1052
60
.
14.
Cercek
A
,
Lumish
M
,
Sinopoli
J
,
Weiss
J
,
Shia
J
,
Lamendola-Essel
M
, et al
.
PD-1 blockade in mismatch repair–deficient, locally advanced rectal cancer
.
N Engl J Med
2022
;
386
:
2363
76
.
15.
Chalabi
M
,
Fanchi
L
,
Dijkstra
K
.
Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers
.
Nat Med
2020
;
26
:
566
76
.
16.
Chalabi
M
,
Verschoor
Y
,
van den Berg
S
,
Beets
G
,
Lent
A
,
Grootscholten
M
, et al
.
Neoadjuvant immune checkpoint inhibition in locally advanced MMR-deficient colon cancer: the NICHE-2 study
.
Ann Oncol
2022
;
33
:
S808
69
.
17.
Samstein
RM
,
Lee
C-H
,
Shoushtari
AN
,
Hellmann
MD
,
Shen
R
,
Janjigian
YY
, et al
.
Tumor mutational load predicts survival after immunotherapy across multiple cancer types
.
Nat Genet
2019
;
51
:
202
6
.
18.
Chan
T
.
The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy
.
Nat Rev Cancer
2019
;
19
:
133
50
.
19.
Chen
EX
,
Jonker
DJ
,
Loree
JM
,
Kennecke
HF
,
Berry
SR
,
Couture
F
, 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
:
831
8
.
20.
McGrail
DJ
,
Pilié
PG
,
Rashid
NU
,
Voorwerk
L
,
Slagter
M
,
Kok
M
, et al
.
High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types
.
Ann Oncol
2021
;
32
:
661
72
.
21.
Rousseau
B
,
Bieche
I
,
Pasmant
E
,
Hamzaoui
N
,
Leulliot
N
,
Michon
L
, et al
.
PD-1 blockade in solid tumors with defects in polymerase epsilon
.
Cancer Discov
2022
;
12
:
1435
48
.
22.
Boesch
M
,
Baty
F
,
Rothschild
SI
,
Tamm
M
,
Joerger
M
,
Früh
M
, et al
.
Tumor neoantigen mimicry by microbial species in cancer immunotherapy
.
Br J Cancer
2021
;
125
:
313
23
.
23.
Turajilic
S
,
Litchfield
K
,
Xu
H
.
Insertion-and-deletion-derived tumor-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis
.
Lancet Oncology
2017
;
18
:
1009
21
.
24.
Łuksza
M
,
Riaz
N
,
Makarov
V
,
Balachandran
VP
,
Hellmann
MD
,
Solovyov
A
, et al
.
A neoantigen fitness model predicts tumor response to checkpoint blockade immunotherapy
.
Nature
2017
;
551
:
517
20
.
25.
Niño
JLG
,
Wu
H
,
LaCourse
KD
,
Kempchinsky
AG
,
Baryiames
A
,
Barber
B
, et al
.
Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer
.
Nature
2022
;
611
:
810
7
.
26.
Gopalakrishnan
V
,
Helmink
BA
,
Spencer
CN
,
Reuben
A
,
Wargo
JA
.
The influence of the gut microbiome on cancer, immunity, and cancer immunotherapy
.
Cancer Cell
2018
;
33
:
570
80
.
27.
Gopalakrishnan
V
,
Spencer
CN
,
Nezi
L
,
Reuben
A
,
Andrews
MC
,
Karpinets
TV
, et al
.
Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients
.
Science
2018
;
359
:
97
103
.
28.
Sivan
A
,
Corrales
L
,
Hubert
N
,
Williams
JB
,
Aquino-Michaels
K
,
Earley
ZM
, et al
.
Commensal bifidobacterium promotes antitumor immunity and facilitates anti–PD-L1 efficacy
.
Science
2015
;
350
:
1084
9
.
29.
Guinney
J
,
Dienstmann
R
,
Wang
X
,
de Reyniès
A
,
Schlicker
A
,
Soneson
C
, et al
.
The consensus molecular subtypes of colorectal cancer
.
Nat Med
2015
;
21
:
1350
6
.
30.
Angell
HK
,
Bruni
D
,
Barrett
JC
,
Herbst
R
,
Galon
J
.
The Immunoscore: Colon Cancer and Beyond
.
Clin Cancer Res
2020
;
26
:
332
9
.
31.
Cortes
J
,
Rugo
HS
,
Cescon
DW
,
Im
S-A
,
Yusof
MM
,
Gallardo
C
, et al
.
Pembrolizumab plus chemotherapy in advanced triple-negative breast cancer
.
N Engl J Med
2022
;
387
:
217
26
.
32.
Gandhi
L
,
Rodríguez-Abreu
D
,
Gadgeel
S
,
Esteban
E
,
Felip
E
,
De Angelis
F
, et al
.
Pembrolizumab plus chemotherapy in metastatic non–small cell lung cancer
.
N Engl J Med
2018
;
378
:
2078
92
.
33.
Vanneman
M
,
Dranoff
G
.
Combining immunotherapy and targeted therapies in cancer treatment
.
Nat Rev Cancer
2012
;
12
:
237
51
.
34.
Zitvogel
L
,
Kepp
O
,
Kroemer
G
.
Immune parameters affecting the efficacy of chemotherapeutic regimens
.
Nat Rev Clin Oncol
2011
;
8
:
151
60
.
35.
Suzuki
E
,
Kapoor
V
,
Jassar
AS
,
Kaiser
LR
,
Albelda
SM
.
Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity
.
Clin Cancer Res
2005
;
11
:
6713
21
.
36.
Lenz
H
,
Parikh
A
,
Spigel
D
,
Cohen
A
,
Yoshino
T
,
Kochenderfer
M
.
Nivolumab (NIVO) + 5-fluorouracil/leucovorin/oxaliplatin (mFOLFOX6)/bevacizumab (BEV) versus mFOLFOX6/BEV for first-line (1L) treatment of metastatic colorectal cancer (mCRC): phase II results from CheckMate 9×8
.
J Clin Oncol
40
:
S8
.
37.
Segal
N
,
Tie
J
,
Kopetz
S
,
Ducrex
M
,
Chen
E
,
Dienstmann
R
.
COLUMBIA-1: A phase Ib/II, open-label, randomized, multicenter study of durvalumab plus oleclumab in combination with chemotherapy and bevacizumab as first-line (1L) therapy in metastatic microsatellite-stable colorectal cancer (MSS-mCRC)
.
2022
;
16
:
S100272
.
38.
Ghiringhelli
F
,
Chibaudel
B
,
Taieb
J
,
Bennouna
J
,
Martin-Babau
F
.
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 Ib/II MEDETREME trial
.
J Clin Oncol
2020
;
38
:
3006
.
39.
Antoniotti
C
,
Rossini
D
,
Pietrantonio
F
,
Catteau
A
,
Salvatore
L
,
Lonardi
S
, et al
.
Upfront FOLFOXIRI plus bevacizumab with or without atezolizumab in the treatment of patients with metastatic colorectal cancer (AtezoTRIBE): a multicenter, open-label, randomized, controlled, phase II trial
.
Lancet Oncol
2022
;
23
:
876
87
.
40.
Ngwa
W
,
Irabor
OC
,
Schoenfeld
JD
,
Hesser
J
,
Demaria
S
,
Formenti
SC
.
Using immunotherapy to boost the abscopal effect
.
Nat Rev Cancer
2018
;
18
:
313
22
.
41.
Postow
MA
,
Callahan
MK
,
Barker
CA
,
Yamada
Y
,
Yuan
J
,
Kitano
S
, et al
.
Immunologic correlates of the abscopal effect in a patient with melanoma
.
N Engl J Med
2012
;
366
:
925
31
.
42.
Golden
EB
,
Chhabra
A
,
Chachoua
A
,
Adams
S
,
Donach
M
,
Fenton-Kerimian
M
, et al
.
Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumors: a proof-of-principle trial
.
Lancet Oncol
2015
;
16
:
795
803
.
43.
Barker
HE
,
Paget
JTE
,
Khan
AA
,
Harrington
KJ
.
The tumor microenvironment after radiotherapy: mechanisms of resistance and recurrence
.
Nat Rev Cancer
2015
;
15
:
409
25
.
44.
Segal
NH
,
Cercek
A
,
Ku
G
,
Wu
AJ
,
Rimner
A
,
Khalil
DN
, 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
:
2200
8
.
45.
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
:
1124
35
.
46.
Manzoni
M
,
Rovati
B
,
Ronzoni
M
,
Loupakis
F
,
Mariucci
S
,
Ricci
V
, et al
.
Immunological effects of bevacizumab-based treatment in metastatic colorectal cancer
.
Oncology
2010
;
79
:
187
96
.
47.
Kato
Y
,
Tabata
K
,
Kimura
T
,
Yachie-Kinoshita
A
,
Ozawa
Y
,
Yamada
K
, 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
:
e0212513
.
48.
Boni
A
,
Cogdill
AP
,
Dang
P
,
Udayakumar
D
,
Njauw
C-NJ
,
Sloss
CM
, et al
.
Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function
.
Cancer Res
2010
;
70
:
5213
9
.
49.
Gomez-Roca
C
,
Yanez
E
,
Seock-Ah
I
,
Alvarez
E
,
Senellart
H
,
Doherty
M
.
LEAP-005: A phase II 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
:
s94
.
50.
Fakih
M
,
Pratap
K
,
Raghav
S
,
Chang
D
,
Bendell
J
,
Larson
T
.
Single-arm, phase II study of regorafenib plus nivolumab in patients with mismatch repair–proficient (pMMR)/microsatellite stable (MSS) colorectal cancer (CRC)
.
J Clin Oncol
2021
;
39
:
S3560
.
51.
Fakih
M
,
Sandhu
J
,
Lim
D
,
Li
S
,
Wang
C
.
A phase I clinical trial of regorafenib, ipilimumab, and nivolumab (RIN) in chemotherapy resistant MSS metastatic colorectal cancer (mCRC)
.
Ann Oncol
2022
;
33
:
S136
.
52.
Grothey
A
,
Van Cutsem
E
,
Sobrero
A
,
Siena
S
,
Falcone
A
,
Ychou
M
, et al
.
Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase III trial
.
Lancet
2013
;
381
:
303
12
.
53.
Verma
V
,
Jafarzadeh
N
,
Boi
S
,
Kundu
S
,
Jiang
Z
,
Fan
Y
, et al
.
MEK inhibition reprograms CD8+ T lymphocytes into memory stem cells with potent antitumor effects
.
Nat Immunol
2021
;
22
:
53
66
.
54.
Morris
V
,
Parseghian
C
,
Escano
M
,
Johnson
B
,
Raghav
K
,
Dasari
A
.
Phase I/II trial of encorafenib, cetuximab, and nivolumab in patients with microsatellite stable (MSS), BRAFV600E metastatic colorectal cancer
.
J Clin Oncol
2022
;
40
.
55.
Kopetz
S
,
Grothey
A
,
Yaeger
R
,
Van Cutsem
E
,
Desai
J
,
Yoshino
T
, et al
.
Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer
.
N Engl J Med
2019
;
381
:
1632
43
.
56.
Eng
C
,
Kim
TW
,
Bendell
J
,
Argilés
G
,
Tebbutt
NC
,
Di Bartolomeo
M
, et al
.
Atezolizumab with or without cobimetinib versus regorafenib in previously treated metastatic colorectal cancer (IMblaze370): a multicenter, open-label, phase III, randomized, controlled trial
.
Lancet Oncol
2019
;
20
:
849
61
.
57.
Johnson
B
,
Haymaker
CL
,
Parra
ER
,
Soto
LMS
,
Wang
X
,
Thomas
JV
, et al
.
Phase II study of durvalumab (anti–PD-L1) and trametinib (MEKi) in microsatellite stable (MSS) metastatic colorectal cancer (mCRC)
.
J Immunother Cancer
2022
;
10
:
e005332
.
58.
Bullock
A
,
Grossman
J
,
Fakih
M
,
Lenz
H
,
Gardon
M
,
Margolin
K
.
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
.
59.
Tawbi
HA
,
Schadendorf
D
,
Lipson
EJ
,
Ascierto
PA
,
Matamala
L
,
Castillo Gutiérrez
E
, et al
.
Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma
.
N Engl J Med
2022
;
386
:
24
34
.
60.
Garralda
E
,
Sukari
A
,
Lakhani
N
,
Patnaik
A
,
Lou
Y
.
A phase I first-in-human study of the anti–LAG-3 antibody MK4280 (favezelimab) plus pembrolizumab in previously treated, advanced microsatellite stable colorectal cancer
.
J Clin Oncol
2021
;
39
:
3584
.
61.
Gatti-Mays
ME
,
Strauss
J
,
Donahue
RN
,
Palena
C
,
Del Rivero
J
,
Redman
JM
, et al
.
A 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
44
.
62.
Speetjens
FM
,
Kuppen
PJK
,
Welters
MJP
,
Essahsah
F
,
Voet van den Brink
A
,
Lantrua
MGK
, et al
.
Induction of p53-specific immunity by a p53 synthetic long peptide vaccine in patients treated for metastatic colorectal cancer
.
Clin Cancer Res
2009
;
15
:
1086
95
.
63.
Hubbard
JM
,
Tőke
ER
,
Moretto
R
,
Graham
RP
,
Youssoufian
H
,
Lőrincz
O
, et al
.
Safety and activity of PolyPEPI1018 combined with maintenance therapy in metastatic colorectal cancer: an open-label, multicenter, phase Ib study
.
Clin Cancer Res
2022
;
28
:
2818
29
.
64.
Schimanski
CC
,
Kasper
S
,
Hegewisch-Becker
S
,
Schröder
J
,
Overkamp
F
,
Kullmann
F
, et al
.
Adjuvant MUC vaccination with tecemotide after resection of colorectal liver metastases: a randomized, double-blind, placebo-controlled, multicenter AIO phase II trial (LICC)
.
Oncoimmunology
2020
;
9
:
1806680
.
65.
Kaufman
HL
,
Lenz
H-J
,
Marshall
J
,
Singh
D
,
Garett
C
,
Cripps
C
, et al
.
Combination chemotherapy and ALVAC-CEA/B7.1 vaccine in patients with metastatic colorectal cancer
.
Clin Cancer Res
2008
;
14
:
4843
9
.
66.
Harrop
R
,
Drury
N
,
Shingler
W
,
Chikoti
P
,
Redchenko
I
,
Carroll
MW
, et al
.
Vaccination of colorectal cancer patients with TroVax given alongside chemotherapy (5-fluorouracil, leukovorin and irinotecan) is safe and induces potent immune responses
.
Cancer Immunol Immunother
2008
;
57
:
977
86
.
67.
Blanco
B
,
Domínguez-Alonso
C
,
Alvarez-Vallina
L
.
Bispecific immunomodulatory antibodies for cancer immunotherapy
.
Clin Cancer Res
2021
;
27
:
5457
64
.
68.
Ma
J
,
Mo
Y
,
Tang
M
,
Shen
J
,
Qi
Y
,
Zhao
W
, et al
.
Bispecific antibodies: from research to clinical application
.
Front Immunol.
2021
;
12
:
626616
.
69.
Spiess
C
,
Zhai
Q
,
Carter
PJ
.
Alternative molecular formats and therapeutic applications for bispecific antibodies
.
Mol Immunol
2015
;
67
:
95
106
.
70.
Fan
G
,
Wang
Z
,
Hao
M
,
Li
J
.
Bispecific antibodies and their applications
.
J Hematol Oncol
2015
;
8
:
130
.
71.
Esfandiari
A
,
Cassidy
S
,
Webster
RM
.
Bispecific antibodies in oncology
.
Nat Rev Drug Discov
2022
;
21
:
411
2
.
72.
Pishvaian
M
,
Morse
MA
,
McDevitt
J
,
Norton
JD
,
Ren
S
,
Robbie
GJ
, et al
.
Phase I dose escalation study of MEDI-565, a bispecific T-cell engager that targets human carcinoembryonic antigen, in patients with advanced gastrointestinal adenocarcinomas
.
Clin Colorectal Cancer
2016
;
15
:
345
51
.
73.
Bacac
M
,
Fauti
T
,
Sam
J
,
Colombetti
S
,
Weinzierl
T
,
Ouaret
D
, et al
.
A novel carcinoembryonic antigen T-cell bispecific antibody (CEA TCB) for the treatment of solid tumors
.
Clin Cancer Res
2016
;
22
:
3286
97
.
74.
Segal
N
,
Saro
J
,
Melero
I
,
Ros
W
,
Argiles
G
,
Marabelle
A
.
Phase I studies of the novel carcinoembryonic antigen T-cell bispecific (CEA-CD3 TCB) antibody as a single agent and in combination with atezolizumab: Preliminary efficacy and safety in patients (pts) with metastatic colorectal cancer (mCRC)
.
Ann Oncol
2017
;
8
:
V134
.
75.
Fucà
G
,
Reppel
L
,
Landoni
E
,
Savoldo
B
,
Dotti
G
.
Enhancing chimeric antigen receptor T-cell efficacy in solid tumors
.
Clin Cancer Res
2020
;
26
:
2444
51
.
76.
Maude
SL
,
Frey
N
,
Shaw
PA
,
Aplenc
R
,
Barrett
DM
,
Bunin
NJ
, et al
.
Chimeric antigen receptor T cells for sustained remissions in leukemia
.
N Engl J Med
2014
;
371
:
1507
17
.
77.
Larson
RC
,
Maus
MV
.
Recent advances and discoveries in the mechanisms and functions of CAR T cells
.
Nat Rev Cancer
2021
;
21
:
145
61
.
78.
Shah
NN
,
Fry
TJ
.
Mechanisms of resistance to CAR T cell therapy
.
Nat Rev Clin Oncol
2019
;
16
:
372
85
.
79.
Zhang
C
,
Wang
Z
,
Yang
Z
,
Wang
M
,
Li
S
,
Li
Y
, et al
.
Phase I escalating-dose trial of CAR-T therapy targeting CEA+ metastatic colorectal cancers
.
Mol Ther
2017
;
25
:
1248
58
.
80.
Thistlethwaite
FC
,
Gilham
DE
,
Guest
RD
,
Rothwell
DG
,
Pillai
M
,
Burt
DJ
, et al
.
The clinical efficacy of first-generation carcinoembryonic antigen (CEACAM5)-specific CAR T cells is limited by poor persistence and transient pre-conditioning-dependent respiratory toxicity
.
Cancer Immunol Immunother
2017
;
66
:
1425
36
.
81.
Hege
KM
,
Bergsland
EK
,
Fisher
GA
,
Nemunaitis
JJ
,
Warren
RS
,
McArthur
JG
, et al
.
Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer
.
J Immunother Cancer
2017
;
5
:
22
.
82.
Cui
J
,
Chen
N
,
Chengfei
P
,
Zhao
L
,
Li
N
,
Wang
C
.
A phase I dose-escalation study of GCC19 CART a novel coupled CAR therapy for subjects with metastatic colorectal cancer
.
J Clin Oncol
2022
;
40
:
S3582
.
83.
Zhang
B
,
Huang
J
,
Tang
J
,
Hu
S
,
Luo
S
,
Luo
Z
, et al
.
Intratumoral OH2, an oncolytic herpes simplex virus 2, in patients with advanced solid tumors: a multicenter, phase I/II clinical trial
.
J Immunother Cancer
2021
;
9
:
e002224
.
84.
Geevarghese
SK
,
Geller
DA
,
de Haan
HA
,
Hörer
M
,
Knoll
AE
,
Mescheder
A
, et al
.
Phase I/II study of oncolytic herpes simplex virus NV1020 in patients with extensively pretreated refractory colorectal cancer metastatic to the liver
.
Hum Gene Ther
2010
;
21
:
1119
28
.
85.
Crisafulli
G
,
Sartore-Bianchi
A
,
Lazzari
L
,
Pietrantonio
F
,
Amatu
A
,
Macagno
M
, et al
.
Temozolomide treatment alters mismatch repair and boosts mutational burden in tumor and blood of colorectal cancer patients
.
Cancer Discov
2022
;
12
:
1656
75
.
86.
Morano
F
,
Raimondi
A
,
Pagani
F
,
Lonardi
S
,
Salvatore
L
,
Cremolini
C
, et al
.
Temozolomide followed by combination with low-dose ipilimumab and nivolumab in patients with microsatellite-stable, O6-methylguanine-DNA methyltransferase-silenced metastatic colorectal cancer: the MAYA trial
.
J Clin Oncol
2022
;
40
:
1562
73
.
87.
Lee
JC
,
Green
MD
,
Huppert
LA
,
Chow
C
,
Pierce
RH
,
Daud
AI
.
The liver-immunity nexus and cancer immunotherapy
.
Clin Cancer Res
2022
;
28
:
5
12
.
88.
Bilen
MA
,
Shabto
JM
,
Martini
DJ
,
Liu
Y
,
Lewis
C
,
Collins
H
, et al
.
Sites of metastasis and association with clinical outcome in advanced stage cancer patients treated with immunotherapy
.
BMC Cancer
2019
;
19
:
857
.
89.
Tumeh
PC
,
Hellmann
MD
,
Hamid
O
,
Tsai
KK
,
Loo
KL
,
Gubens
MA
, et al
.
Liver metastasis and treatment outcome with anti–PD-1 monoclonal antibody in patients with melanoma and NSCLC
.
Cancer Immunol Res
2017
;
5
:
417
24
.
90.
Sridhar
S
,
Paz-Ares
L
,
Liu
H
,
Shen
K
,
Morehouse
C
,
Rizvi
N
, et al
.
Prognostic significance of liver metastasis in durvalumab-treated lung cancer patients
.
Clin Lung Cancer
2019
;
20
:
e601
8
.
91.
Horst
AK
,
Neumann
K
,
Diehl
L
,
Tiegs
G
.
Modulation of liver tolerance by conventional and nonconventional antigen-presenting cells and regulatory immune cells
.
Cell Mol Immunol
2016
;
13
:
277
92
.
92.
Yu
J
,
Green
MD
,
Li
S
,
Sun
Y
,
Journey
SN
,
Choi
JE
, et al
.
Liver metastasis restrains immunotherapy efficacy via macrophage-mediated T cell elimination
.
Nat Med
2021
;
27
:
152
64
.
93.
Tiegs
G
,
Lohse
AW
.
Immune tolerance: what is unique about the liver
.
J Autoimmun
2010
;
34
:
1
6
.
94.
Engstrand
J
,
Nilsson
H
,
Strömberg
C
,
Jonas
E
,
Freedman
J
.
Colorectal cancer liver metastases: a population-based study on incidence, management and survival
.
BMC Cancer
2018
;
18
:
78
.
95.
Xiao
L
,
Cen
D
,
Gan
H
,
Sun
Y
,
Huang
N
,
Xiong
H
, et al
.
Adoptive transfer of NKG2D CAR mRNA-engineered natural killer cells in colorectal cancer patients
.
Mol Ther
2019
;
27
:
1114
25
.
96.
Barzi
A
,
Azad
NS
,
Yang
Y
,
Tsao-Wei
D
,
Rehmen
R
,
Fakih
M
.
Phase I/II study of regorafenib (rego) and pembrolizumab (pembro) in refractory microsatellite stable colorectal cancer (MSSCRC)
.
J Clin Oncol
40
:
S15
.
97.
Cousin
S
,
Cantarel
C
,
Guegan
J-P
,
Gomez-Roca
C
,
Metges
J-P
,
Adenis
A
, 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
:
2139
47
.
98.
Saeed
A
,
Park
R
,
Dai
J
,
Al-Rajabi
R
,
Kasi
A
,
Saeed
A
.
Phase II trial of cabozantinib (Cabo) plus durvalumab (Durva) in chemotherapy refractory patients with advanced mismatch repair–proficient/microsatellite stable (pMMR/MSS) colorectal cancer (CRC): CAMILLA CRC cohort results
.
J Clin Oncol
40
:
S135
.
99.
Martinelli
E
,
Martini
G
,
Famiglietti
V
,
Troiani
T
,
Napolitano
S
,
Pietrantonio
F
, et al
.
Cetuximab rechallenge plus avelumab in pretreated patients with RAS wild-type metastatic colorectal cancer: the phase II single-arm clinical CAVE trial
.
JAMA Oncol
2021
;
7
:
1529
35
.