Next Article in Journal
Ferroptosis Regulated by Hypoxia in Cells
Next Article in Special Issue
Zinc Finger Proteins in the War on Gastric Cancer: Molecular Mechanism and Clinical Potential
Previous Article in Journal
Tumor Microenvironmental Cytokines Drive NSCLC Cell Aggressiveness and Drug-Resistance via YAP-Mediated Autophagy
Previous Article in Special Issue
Hydrophobic Bile Salts Induce Pro-Fibrogenic Proliferation of Hepatic Stellate Cells through PI3K p110 Alpha Signaling
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Strategy to Treat Immunogenic Gastrointestinal Cancers: Perspectives for a New Era

1
Department of Gastrointestinal Oncology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo 135-0063, Japan
2
Department of Gastroenterology and Hepatology, Division of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
3
Department of Experimental Pathology, Cancer Institute of the Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
*
Author to whom correspondence should be addressed.
Cells 2023, 12(7), 1049; https://doi.org/10.3390/cells12071049
Submission received: 21 December 2022 / Revised: 15 March 2023 / Accepted: 29 March 2023 / Published: 30 March 2023

Abstract

:
Since pembrolizumab, an anti-programmed death-1 (PD-1) antibody, showed a dramatic response to immunogenic cancers with microsatellite instability-high (MSI-H) and/or deficient mismatch repair (dMMR) in the pilot clinical trial KEYNOTE-016, subsequent studies have confirmed durable responses of anti-PD-1 inhibitors for MSI-H/dMMR solid tumors. As immunotherapy is described as a “game changer,” the therapeutic landscape for MSI-H/dMMR solid tumors including gastrointestinal cancers has changed considerably in the last decade. An MSI/MMR status has been established as the predictive biomarker for immune checkpoint blockades, playing an indispensable role in the clinical practice of patients with MSI-H/dMMR tumors. Immunotherapy is also now investigated for locally advanced MSI-H/dMMR gastrointestinal cancers. Despite this great success, a few populations with MSI-H/dMMR gastrointestinal cancers do not respond to immunotherapy, possibly due to the existence of intrinsic or acquired resistance mechanisms. Clarifying the underlying mechanisms of resistance remains a future task, whereas attempts to overcome resistance and improve the efficacy of immunotherapy are currently ongoing. Herein, we review recent clinical trials with special attention to MSI-H/dMMR gastrointestinal cancers together with basic/translational findings, which provide their rationale, and discuss perspectives for the further therapeutic development of treatment in this field.

A microsatellite instability-high (MSI-H)/deficient mismatch repair (dMMR) tumor has an impaired mismatch repair machinery system, which induces hypermutations, mainly indel and/or deletion causing the potential mutation-generated neoantigens. This theoretical immunogenicity of the MSI-H/dMMR tumor has been proven by the successful results of clinical trials using immune checkpoint inhibitors (ICIs), now being recognized as the standard care treatment for metastatic disease. Immunotherapy has changed the therapeutic landscape for not only metastatic MSI-H/dMMR cancers but also locally advanced MSI-H/dMMR tumors. These promising results lead us to expect that the treatment goals for locally advanced MSI-H/dMMR cancers are shifting from “patient care” toward “cure” with or without organ preservation. Clarifying the underlying intrinsic or acquired resistant mechanisms is increasingly important for the further improvement of MSI-H/dMMR gastrointestinal (GI) cancers. Further, MSI-H/dMMR tumors comprise only a rare fraction of the entire GI cancers. Exploring the immunogenicity of GI cancers beyond MSI/MMR status is an important challenge to open the door to the new immune-oncology era. In this review, we look back on the history of immunotherapy for MSI-H/dMMR GI malignancies with special attention to the progress of clinical trials together with basic/translational findings, which provide perspectives for further therapeutic development for treatment in this field.

1. Results of Early-Phase Clinical Trials Were Disappointing for Colorectal Cancers

The first in-human clinical trial using an anti-programmed cell death-1 (PD-1) blockade was conducted between October 2006 and June 2009 [1]. Thirty-nine patients with advanced metastatic non-small-cell lung cancer (NSCLC), melanoma, castration-resistant prostate cancer, renal cell carcinoma (RCC), and metastatic colorectal cancer (mCRC) were enrolled in the phase I trial and received MDX-1106 (BMS-936558/ONO-4538), which is currently known as nivolumab, a fully humanized immunoglobulin G4 (IgG4) monoclonal antibody. This trial included not only cancers traditionally considered immunogenic such as melanoma and RCC but also certain malignancies such as NSCLC and CRC, which had been commonly viewed as refractory to immunotherapy or nonimmunogenic. This early-phase dose-expansion study identified the responders to an immune checkpoint blockade (three patients with melanoma, RCC, and mCRC). A 67-year-old man with mCRC had a complete response (CR) to MDX-1106 (3 mg/kg). Based on this finding, physicians expected this new agent to expand the boundary of immunotherapy to classical nonimmunogenic tumors [1]. However, the initial activity signal of MDX-1106 for patients with mCRC was not found reproducibly in a larger-scale clinical trial in which a total of 296 patients had been treated with BMS-936558 [2]. Of the 236 patients in the efficacy analysis cohort, approximately 25% of the patients with NSCLC (14 of 76), melanoma (26 of 94), and RCC (9 of 33) had an objective response, whereas no response was found in 19 patients with mCRC [2]. Further, the successful results in NSCLC accelerated the development of immunotherapy, and at present, NSCLC is the top-runner in the immune-oncology field. Subsequent developments of immunotherapy in solid tumors include RCC.
The first successful case of immunotherapy for mCRC in the phase I trial was found to be MSI-H, with tumor-infiltrating macrophages and lymphocytes surrounding the tumor cells, and durable CR for over 4 years was seen without any antineoplastic agents [3]. What was different between this successful case and the other non-responders was the key question that fostered opening the door of immunotherapy. Le et al. hypothesized that the MSI-H genotype contributed to the great response to the PD-1 blockade [4,5,6].

2. Opening the Door to the Immune-Oncology Field

MMR is one of the DNA repair processes that recognize and amend incorrect insertion, deletion, or misincorporation of bases [7,8,9]. The loss of MMR function results in changes in the number of short-tandem repeats termed microsatellites (Figure 1a) [10,11]. MSI-H refers to a high frequency of MSI, whereas MSI-low/microsatellite stable (MSI-L/MSS) refers to a low frequency or no MSI. The dMMR is thought to contribute to tumorigenesis and proliferation by accumulating mutations in genes involved in tumor suppression, cell proliferation, DNA repair, and apoptosis [12]. These mutation-generated abnormal proteins, so-called “neoantigens”, are taken up by antigen-presenting cells (APCs). MSI-H/dMMR cancers harbor numerous somatic mutations, which could encode diverse neoantigens, being represented as high tumor mutational burdens (Figure 1b) [13,14]. APCs stimulate cytotoxic T lymphocytes and attack tumor cells. Cancer cells, on the other hand, produce immune checkpoint molecules throughout this process, including programmed cell death ligand 1 (PD-L1), to evade T cells (Figure 1c) [15,16,17,18]. As a result, T cells can no longer target those cancer cells. Anti-PD-1 antibodies prevent PD-1/PD-L1 binding, allowing T cells to become active once more and fight the tumor [19]. Thus, MSI-H/dMMR tumors are more easily recognized by tumor antigen-specific T cells than those with MSI-L/MSS and/or proficient MMR (pMMR) tumors [20]. Indeed, Nicolas et al. revealed the pathological features of resected MSI-H/dMMR colon cancer in which activated T cells highly infiltrated the tumors (Figure 1d) [16]. Therefore, MSI-H tumors are presumed to be better candidates for ICI therapy.
Notably, this scenario has been successfully demonstrated in clinical trials. The KEYNOTE-016 trial was the first study to indicate the effectiveness of the PD-1 blockade for dMMR tumors irrelevant to the tumor origin and ineffectiveness of pMMR tumors. Pembrolizumab, a humanized IgG4 monoclonal antibody, demonstrated a clinically significant and long-lasting response for dMMR mCRC (cohort A: n = 11), with an objective response rate (ORR) of 40% (95% confidence interval [CI], 12–74), a disease control rate (DCR) of 90% (95% CI, 55–100), and the median overall survival (OS) and progression-free survival (PFS) were not reached (NR) at data cutoff, whereas pMMR mCRC (n = 21) did not receive clinical benefit from pembrolizumab [6]. Of note, 9 of 11 patients with dMMR mCRC were diagnosed with Lynch syndrome. Based on this promising result, KEYNOTE-164, a phase II multicohort study, was conducted to examine the efficacy of pembrolizumab in patients with MSI-H/dMMR mCRC who had received at least one treatment before enrollment [21]. The study reported the response of two groups: cohort A patients who had received two or more regimens before the induction of pembrolizumab (n = 61), and cohort B that had received one regimen before pembrolizumab administration (n = 63). In cohort A, pembrolizumab demonstrated an ORR of 33% (95% CI, 21–46), a median PFS of 2.3 months (2.1–8.1), and a median OS of 31.4 months (21.4–NR) (Table 1). Similarly, pembrolizumab showed clinically meaningful benefit in cohort B: the ORR, median PFS, and OS were 33% (22–46), 4.1 months (2.1–18.9), and NR (19.2–NR), respectively.
The success of immunotherapy for MSI-H/dMMR mCRC encouraged the investigation of the efficacy of pembrolizumab in different cancer subtypes with MSI-H/dMMR, resulting in the expansion of the cohort for MSI-H solid tumors in the KEYNOTE-016 [6,22]. Of the 86 patients finally enrolled in the study, CR was obtained in 18 (21%) patients, and the ORR was 53% (95% CI, 42–64). The PFS and OS were surprisingly prolonged (median PFS, 14.3–NR; median OS, NR–NR) [22]. The results of KEYNOTE-016 also built momentum for the development of immunotherapy for non-CRC MSI-H/dMMR tumors, leading to the commencement of the KEYNOTE-158 trial (details are described in a later section).
Table 1. Clinical trials for MSI-H/dMMR CRC.
Table 1. Clinical trials for MSI-H/dMMR CRC.
StudyKEYNOTE-164 [21]KEYNOTE-177 [23]CheckMate-142 [24,25,26]
Treatment line≥2nd line≥3rd line1st line≥2nd line≥2nd line1st line
PhaseIIIIIII
No. of patients63611531547411945
Primary endpointORRPFS and OSORR
RegimenPembrolizumab
200 mg every 3 weeks
Pembrolizumab
200 mg every 3 weeks
ChemotherapyNivolumab 3 mg/kg every 2 weeksNivolumab 3 mg/kg plus
ipilimumab 1 mg/kg every 3 weeks (4 cycles) followed by nivolumab 3 mg/kg every 2 weeks
Nivolumab 3 mg/kg every 2 weeks plus ipilimumab 1 mg/kg every 6 weeks
ORR33%33%43.8%33.1%32%49%69%
DCR57%51%64.7%75.3%64%79%84%
Median PFS4.1 months2.3 months16.5 months8.2 months14.3 monthsNRNR
12-month PFS rate41%34%55.3%37.3%50%71%76.4%
Median OSNR31.4 monthsNR36.7 monthsNRNRNR
12-month OS rate77%72%78%74%73%85%84.1%
Abbreviations: DCR, disease control rate; NR, not reached; ORR, objective response rate; OS, overall survival; PFS, progression-free survival.
This was the first moment when immunotherapy met MSI-H/dMMR tumors: a man’s success story expanded beyond the boundary of tumor origin. The resulting data support basket trials that have examined the effectiveness of immune checkpoint blockades in patients with advanced MSI-H/dMMR cancers. As described in detail in later sections, pembrolizumab first showed a dramatic response in the evaluation of 149 patients with advanced MSI-H/dMMR solid tumors (Table 2) [6,22,27], resulting in the first tumor-agnostic approval by the FDA (Figure 2) [28].

3. Current Status of Gastrointestinal (GI) Cancers with MSI-H/dMMR in Clinical Practice

3.1. CRC

Patients with MSI-H/dMMR mCRC account for approximately 3.5–6.5% of mCRC [22,24]; a majority of those are sporadic cases, whereas others have hereditary cases [4,30]. Among these cases, 35–43% of patients harbored BRAFV600E mutations [31]; however, the prognostic significance of the BRAFV600E mutation in MSI-H/dMMR mCRC has not been well explored. The prognosis of patients with MSI-H/dMMR mCRC has been reported as a median OS of 13.6–21.5 months, which was relatively poor when compared with MSS and/or pMMR mCRC [32]. Over the years, the standard of care for this population has remained cytotoxic chemotherapy, even if MSI-H/dMMR mCRC is generally thought to be less sensitive to conventional chemotherapy than MSS/pMMR mCRC.
Based on the results of KEYNOTE-016 [6] and KEYNOTE-164 [21], the attempts to offer the anti-PD-1 antibody to first-line treatment have progressed. KEYNOTE-177 was the randomized phase III trial that initially assessed the efficacy of pembrolizumab in comparison with standard-of-care chemotherapy for patients with previously untreated MSI-H/dMMR mCRC [23]. The co-primary outcomes were PFS and OS; two interim analyses and a final analysis were preplanned. In the second interim analysis, pembrolizumab demonstrated statistically significant improvement in PFS (median, 16.5 months; 95% CI, 5.4–32.4) compared with chemotherapy (median, 8.2 months; 95% CI, 6.1–10.1) (hazard ratio [HR], 0.60; 95% CI, 0.45–0.80; p = 0.0002), with fewer treatment-related adverse events than chemotherapy. Although OS data were immature at that time, the trial was considered positive. This led the FDA to approve pembrolizumab as the standard treatment for patients with MSI-H/dMMR mCRC as a first-line treatment in June 2020 [33]. In the final analysis, the median OS was NR in the pembrolizumab group (95% CI, 49.2–NR) and 36.7 months in the standard chemotherapy group (95% CI, 37.6–NR), with HR of 0.74 (95% CI, 0.53–1.03; p = 0.036), with a p-value exceeding the predetermined significance level (p = 0.0246) [34]. Additionally, a predetermined sensitivity analysis to account for the effect of crossover treatment revealed an HR for OS of 0.66 (95% CI, 0.42–1.04). Despite these positive results, pembrolizumab did not show a statistically significant reduction in the risk of death compared to standard chemotherapy, whereas only 140 events (45.6%) occurred at the time of the final analysis for OS [34]. The finding that 60% of the patients in the standard chemotherapy group had received anti-PD-1/PD-L1 antibody as a second-line treatment might also affect the results. Given the HR of 0.74 compared with a median OS of 36.7 months in the standard chemotherapy arm, which is much better than that previously reported, the results of this study support pembrolizumab as the standard first-line treatment for patients with MSI-H/dMMR mCRC (Table 2). The establishment of pembrolizumab as the first-line standard therapy is astonishing. The scientific rationale for the efficacy of the PD-1 blockade for MSI-H/dMMR tumors sustained further investigations and succeeded in establishing a new standard therapy.

3.2. Gastric/Gastroesophageal Junction Cancers (GC/GEJC)

The therapeutic development of ICIs for metastatic upper GI cancers followed different paths from that of mCRC. Seven patients with GC were included in the initial clinical trial conducted between 2009 and 2012; however, these patients were eliminated from the efficacy population [35]. The efficacy of ICIs in advanced GC/GEJC was evaluated in KEYNOTE-012 and CheckMate-032 [36,37]. KEYNOTE-012, a multicenter phase Ib trial, was conducted to investigate the safety and efficacy of pembrolizumab in PD-L1-positive advanced multiple malignancies. At that time, pembrolizumab demonstrated antitumor activity with durable responses in patients with NSCLC and high PD-L1 expression [36]. Given that previous studies have reported that approximately 40% of patients with GC/GEJC showed high PD-L1 expressions [38,39,40], patients with PD-L1-positive GC/GEJC had been selectively enrolled in KEYNOTE-012. In the GC/GEJC cohort (n = 39), tumor shrinkage was obtained in 22% of the patients [29]. KEYNOTE-059, a larger-scale, multicenter, multicohort phase II trial, was subsequently conducted to investigate the efficacy of pembrolizumab monotherapy for patients with previously treated GC/GEJC. Pembrolizumab showed favorable efficacy for patients with PD-L1-positive GC/GEJC and a manageable safety profile [41]. Therefore, the attempts to deliver immunotherapy to patients with GC/GEJC proceeded to focus on PD-L1 positivity. The eligibility criteria of KEYNOTE-061 and KEYNOTE-062 trials, which are randomized phase III trials, included PD-L1 positivity [42]. PD-L1 expression was not mandatory in clinical trials using nivolumab, although this did not mean that MSI-H/dMMR populations were unfocused. A multi-institutional phase I/II CheckMate-032 trial investigated the efficacy and safety of nivolumab and nivolumab plus ipilimumab, a fully human monoclonal antibody against cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), between 2013 and 2015 in multiple institutes in the United States and Europe [37]. Although tumor PD-L1 expression and MSI status were explored for potential biomarker analysis of nivolumab, PD-L1 positivity was not required in this study. A higher response rate was reported in nivolumab-treated patients with PD-L1-positive (n = 16, 19%) or MSI-H (n = 7, 29%) GC/GEJC compared with patients who had PD-L1-negative (n = 26, 12%) or MSS (n = 18, 11%) GC/GEJC. Given the small sample size, the trial did not draw conclusive results. ATTRACTION-2 was subsequently conducted in East Asian countries between 2014 and 2016 [43]. Nivolumab monotherapy demonstrated survival benefits for patients with heavily pretreated GC/GEJC, regardless of tumor PD-L1 expression: the median OS was 5.22 months in the nivolumab group and 3.83 months in the placebo group (HR, 0.51; 95% CI, 0.21–1.25) in patients with PD-L1 ≥ 1% GC/GEJC (n = 26) and 6.05 and 4.19 months (HR, 0.72; 95% CI, 0.49–1.05), respectively, in patients with PD-L1 < 1% GC/GEJC (n = 166). The results of ATTRACTION-2 were enough to approve nivolumab as the first ICI for GC/GEJC in 2017 in Japan.
Paradoxically, multiple observations indicate “immunologic cell death”. Certain types of cytotoxic anticancer agents induce a specific form of apoptosis in tumor cells; both innate and adaptive immune responses are activated and enhance the efficacy of immunotherapy [44,45,46,47,48,49]. Combination therapy with immune checkpoint blockade and classical cytotoxic agents has been investigated specifically for NSCLC, and its efficacy has been observed with reproducibility [50,51]. Therefore, the combination of immunotherapy and chemotherapy had also been applied to the front line in metastatic GC/GEJC. CheckMate-649 and ATTRACTION-4 recently demonstrated the superior efficacy of adding nivolumab to cytotoxic chemotherapy in the first-line treatment of HER2-negative advanced GC/GEJC [52,53,54]. In Japan, combination therapy with nivolumab and chemotherapy was approved for HER2-negative advanced GC/GEJC, irrespective of PD-L1 expression, whereas the administration of nivolumab plus chemotherapy as a first-line treatment was limited to PD-L1 expressing GC/GEJC in the United States and Europe. Reflecting these approval conditions, MSI testing is not routinely performed in clinical practice for patients with advanced GC/GEJC.
However, these attempts to broaden the indication of immunotherapy did not deny the potential role of MSI-H/dMMR as a predictive biomarker for immunotherapy in GC/GEJC. The integrated analyses of KEYNOTE-059, KEYNOTE-061, and KEYNOTE-062, which investigated the efficacy of pembrolizumab monotherapy for metastatic GC/GEJC in third-, second-, or first-line settings, respectively, were reported (Table 3) [42]. The enhanced efficacy of pembrolizumab in patients with MSI-H/dMMR was consistently reported in each treatment line. The number of MSI-H cases in each study was as follows: 7 of 174 (4.0%) in KEYNOTE-059, 27 of 514 (5.3%) in KEYNOTE-061, and 50 of 682 (7.3%) in KEYNOTE-062. The median OS in patients with MSI-H in KEYNOTE-059, KEYNOTE-061, and KEYNOTE-062 was NR (95% CI, 1.1–NR), NR (95% CI, 5.6–NR), and NR (95% CI, 10.7–NR), respectively. The median PFS periods for patients with MSI-H treated with pembrolizumab were NR in KEYNOTE-059 (95% CI, 1.1–NR), 17.8 months in KEYNOTE-061 (95% CI, 2.7–NR), and 11.2 months in KEYNOTE-062 (95% CI, 1.5–NR). The ORRs were 57.1% (95% CI, 18.4–90.1), 46.7% (95% CI, 21.3–73.4), and 57.1% (95% CI, 28.9–82.3), respectively. The post hoc analysis of CheckMate-649 also showed a clinically significant advantage of nivolumab plus chemotherapy over chemotherapy for MSI-H GC/GEJC in terms of OS (median, 38.7 vs. 12.3 months; HR 0.38; 95% CI, 0.17–0.84) and ORR (55% vs. 39%) [52]. Therefore, MSI-H/dMMR also has a positive predictive value to enrich the responder of immunotherapy with/without chemotherapy in the treatment of GC/GEJC. However, the incidence of MSI-H/dMMR cancers in metastatic/recurrent GC/GEJC is low (approximately 2–5%) [55,56,57]. Given the fact that patients with non-MSI-H/dMMR metastatic GC/GEJC responded to immunotherapy, it might not be reasonable to limit the use of nivolumab to MSI-H/dMMR alone.

3.3. Other GI Cancers

Far less limited data are available on MSI-H/dMMR GI cancers other than CRC and GC/GEJC. There are few reports on esophageal squamous cell carcinoma, suggesting its low incidence of MSI-H [58,59]. Although small bowel adenocarcinoma (SBA) with MSI-H/dMMR is found in 5–45% of cases, SBA is classified into a rare fraction of malignancies [60,61]. The frequencies of MSI-H/dMMR pancreatic ductal adenocarcinoma (PDAC), biliary tract cancer (BTC), and hepatocellular carcinoma (HCC) are very low. Akagi et al. reported real-world data on the MSI status in Japan (n = 26,469); the incidence of MSI-H is 0.76% in PDAC (n = 2775), 2.1% in BTC/gallbladder cancer (n = 1236), and 1.11% in HCC (n = 452) [62].
The efficacy of the anti-PD-1 antibody is strictly limited in biomarker-unselected non-CRC/GC/GEJC. As for PDAC, the efficacy of pembrolizumab was evaluated in a multicohort phase Ib trial (KEYNOTE-028) in which 24 patients with locally advanced or metastatic PDAC were treated with pembrolizumab, and the potential biomarker of immunotherapy was explored [63]. However, the result was disappointing, with an ORR of 0%, a median PFS of 1.7 months, and a median OS of 3.9 months. Similarly, data from patients with BTC who had received pembrolizumab in KEYNOTE-158 (n = 104) and KEYNOTE-028 (n = 24) were reported [64]. The ORR, median PFS, and median OS were 5.8–13.0%, 1.8–2.0 months, and 5.7–7.4 months, respectively.
The efficacy of immunotherapy in MSI-H/dMMR non-CRC/GC/GEJC GI cancers was assessable in the basket trials. In KEYNOTE-158, 22 patients with BTC, 22 with PDAC, and 19 with SBA were enrolled [27]. A subgroup analysis showed the efficacy of pembrolizumab in BTC, with an ORR of 40.9% (95% CI, 20.7–63.6), a median PFS of 4.2 months (95% CI, 2.1–NR), and a median OS of 24.3 months (95% CI, 6.5–NR). Similarly, the ORR, median PFS, and median OS were 18.2% (5.2–40.3), 2.1 months (1.9–3.4), and 4.0 months (2.1–9.8) in PDAC and 42.1% (20.3–66.5), 9.2 months (2.3–NR), and NR (1.6–NR) in SBA, respectively. Notably, CR was achieved in 9.1% of BTC cases, 4.5% of PDAC, and 15.8% of SBA. By taking these results together, the enrichment of the MSI-H/dMMR population consistently enhances the efficacy of pembrolizumab and demonstrates a favorable response in non-CRC/GC/GEJC.
The efficacy of dostarlimab, a humanized PD-1 monoclonal antibody, was also investigated for MSI-H/dMMR cancers in a nonrandomized, multicenter, multicohort study (GARNET trial: NCT02715284) [65,66]. Cohort F of the GARNET trial enrolled patients with dMMR or POLE-mutant nonendometrial solid tumors (n = 106); 37 patients with non-CRCs were included in the cohort. Sixteen patients (43.2%) had a confirmed ORR, 4 of 12 patients with SBA had a confirmed ORR, and 2 of 2 patients with BTC had a CR [65]. Dostarlimab has been also approved for dMMR advanced solid tumors in the United States since 2021.
The National Cancer Institute Molecular Analysis for Therapy Choice (NCI-MATCH) trial tested the efficacy of nivolumab monotherapy for dMMR solid tumors and reproduced the antitumor activity of immunotherapy for this population [67]. This indicates that ICIs are efficacious to MSI-H/dMMR tumors including rare tumor origins, suggesting that MSI testing for other GI malignancies is important in clinical practice. Several guidelines in the United States, Europe, and Asia recommend ICI therapy for MSI-H/dMMR GI cancers refractory to front-line standard therapy. Given its rare fraction, a randomized controlled study comparing immunotherapy and standard chemotherapy would not be feasible.
Doors for immunotherapy are also opened for neuroendocrine neoplasms (NEN). However, a detailed investigation of the incidence of MSI-H in gastroenteropancreatic NEN has still not been performed. The aforementioned nationwide screening project in Japan reported that 3.42% of patients with neuroendocrine carcinoma (NEC) harbored MSI-H tumors (n = 117) and 2.33% (n = 43) in pancreatic neuroendocrine tumors (NET). The potential efficacy of immunotherapy for locally advanced or metastatic pancreatic NET was reported in the multicohort KEYNOTE-028 study for patients whose tumor had a PD-L1 expression: the ORR was 6.3% (95% CI, 0.2–30.2) [68]. In KEYNOTE-158, patients with NET of the lung, appendix, small intestine, colon, rectum, or pancreas were treated with pembrolizumab. Overall, the ORR was 3.7% (95% CI, 1.0–9.3), whereas the efficacy measures according to tumor types were not described in detail [69]. Studies from Asia have suggested the potential benefit of anti-PD-1 antibodies [70,71]. The inter-trial differences might be explained by the regional differences between Asia and Europe, the proportion of NET and NEC enrolled in the trials, and the sensitivity to immunotherapy of pancreatic and other GI NETs. The MSI/MMR status was not reported in these trials. At present, we should recognize that the MSI-H tumor is present even in rare tumors, and immunotherapy may be beneficial in cases with MSI-H/dMMR. For these patients with rare cancer, the attempt to improve the efficacy of immunotherapy is underway (NCT02923934) [72].

4. Next Challenge for Further Improvement in Patients with MSI-H/dMMR

An MSI-H and/or dMMR status strongly predicts the efficacy of immunotherapy, whereas a certain proportion of patients with MSI-H/dMMR GI cancer show primary or acquired resistance to immunotherapy. Figure 3 highlights the possible mechanisms underlying the immunotherapy resistance in MSI-H/dMMR cancers. The detailed analysis of genetic mechanisms or tumor microenvironment in solid tumors irrespective of MSI/MMR status might give us a hint to resolve the questionable issues [73]. For example, JAK-STAT pathway alterations compromise the immune response and negatively regulate PD-L1 expression in tumor cells [74]. Mutations in beta 2 microglobulin, a component of MHC class I, could impair the presentation of neoantigens to immune cells [75]. The expression of inhibitory immune checkpoints, including CTLA-4, LAG-3, and TIM-3, may induce MSI-H/dMMR tumors to lose the ability to be immunogenic. Immune-evasive oncogenic signaling such as TGF-beta also leads to the differentiation of regulatory T cells and the suppression of NK cell function in the tumor microenvironment [76,77]. High activation of Wnt signaling and upregulation of the interferon–gamma pathway are associated with immune escape mechanisms in MSI-H/dMMR tumors [74,78,79]. The PTEN loss of function induced by PTEN mutations in MSI-H/dMMR tumors is associated with an immunosuppressive tumor microenvironment with depletion of CD8+ T cells and an abundance of tumor-associated macrophages [80]. These mechanisms are intricately involved and interact with each other, whereas the detailed evaluations with a special focus on MSI-H/dMMR tumors have not been fully investigated so far. Whether these assumed mechanisms of resistance for immunotherapy could be also applied in MSI-H/dMMR cancers or not should be investigated in future studies.
An inspection of the aforementioned clinical trials could give us beneficial insight into the resistance mechanism from the clinical aspect. In a subgroup analysis of KEYNOTE-177, patients with KRAS/NRAS mutant mCRC with MSI-H/dMMR did not show the favorable PFS benefit with pembrolizumab compared to chemotherapy (HR, 1.19; 95% CI, 0.68–2.07), whereas BRAFV600E mutant mCRC showed survival benefit from pembrolizumab (HR, 0.48; 95% CI, 0.27–0.86). Immunogenomic analysis in the KEYNOTE-177 also revealed that responsive tumors were rich in PD-1-positive CD8+ T cells interacting with PD-L1-expressed antigen-presenting macrophages [78]. These findings have not been fully investigated in preclinical analysis or validated in further clinical trials; however, it is important to understand the intrinsic or acquired mechanisms of resistance in MSI-H/dMMR cancers, and the attempt to overcome them are moving forward [81].
Pembrolizumab monotherapy was established as the first-line standard treatment in MSI-H/dMMR mCRC, which is a milestone in this field. However, the response rate of immunotherapy for MSI-H/dMMR mCRC remains <50%, suggesting the need for further improvement in this population [21,23,24]. The result of KEYNOTE-177 indicated the primary resistant cases to pembrolizumab, and Kaplan–Meier curves of PFS in each arm crossed at 6 months. This may be one of the next challenges to rescuing these early progressors.
An approach to resolve the problem is to add one agent to a PD-1 blockade. The frontrunner of candidates is an anti-CTLA-4 blockade. CLTA-4 is an immune checkpoint molecule that downregulates T-cell activation pathways via competitively binding to CD80/CD86 against CD28 [82]. An anti-CTLA-4 antibody blocks the binding of CTLA-4 and CD80/CD86; thus, the CD80/CD86-CD28 costimulatory binding strongly promotes T-cell activation. CheckMate-142 is a multicohort, nonrandomized phase II study of nivolumab-based therapies in patients with MSI-H mCRC [24]. The first report of CheckMate-142 showed the promising results of nivolumab monotherapy and nivolumab (3 mg/kg) and low-dose ipilimumab (1 mg/kg) for patients with previously treated MSI-H/dMMR mCRC. Notably, nivolumab plus low-dose ipilimumab showed a higher ORR (54.6%; 95% CI, 45.2–63.8) with manageable toxicity compared to nivolumab monotherapy (31.1%; 95% CI, 20.8–42.9) [24,25]. Although not statistically compared, nivolumab plus low-dose ipilimumab could provide more improved efficacy than nivolumab monotherapy for patients who were previously treated for MSI-H/dMMR mCRC. Recently, nivolumab plus low-dose ipilimumab as a first-line treatment in CheckMate-142 was reported [26]. Of note, this demonstrated promising results: the ORR and DCR increased to 69% (95% CI, 53–82) and 84% (95% CI, 70.5–93.5), respectively. This result led us to expect that the combination therapy could overcome the early progression observed with anti-PD-1 antibody monotherapy. The median PFS and OS were NR at the data cutoff; the 24-month PFS and OS rates were 74% and 79%, respectively. To evaluate the addition of low-dose ipilimumab to nivolumab versus chemotherapy as the first-line treatment of patients with MSI-H/dMMR mCRC, an ongoing confirmatory randomized phase III CheckMate-8HW trial (NCT04008030) is underway [83].
Following the success of nivolumab plus low-dose ipilimumab combination therapy in MSI-H/dMMR mCRC, the NO LIMIT study (WJOG13320G/CA209-7W7), a single-arm phase II trial, to evaluate the efficacy and safety of the nivolumab and low-dose ipilimumab as the first-line setting for MSI-H metastatic GC/GEJC (JapicCTI-205400) is ongoing [84]. Nivolumab plus ipilimumab (nivolumab 1 mg/kg plus ipilimumab 3 mg/kg every 3 weeks for four cycles, then nivolumab 240 mg every 2 weeks) adopted in CheckMate-649 resulted in an early discontinuation recommendation by the data monitoring committee because of unacceptable toxicities [53]. Despite the extremely limited number of patients, the CheckMate-649 regimen showed promising efficacy over chemotherapy in terms of OS (median, NR vs. 10.0 months; HR 0.28; 95% CI, 0.08–0.92) and ORR (70% vs. 57%) [53]. The modified low-dose regimen (nivolumab 3 mg/kg every 2 weeks and ipilimumab 1 mg/kg every 6 weeks until disease progression or unacceptable toxicity) is adopted in the NO LIMIT trial. The result of the NO LIMIT study could offer a novel treatment option to patients with MSI-H/dMMR metastatic GC/GEJC.
As described above, one of the attempts to strengthen the efficacy of the PD-1 blockade is the combination of immunotherapy with cytotoxic agents. The NRG GI004/SWOG1610 trial (COMMIT study) is an ongoing phase III trial that randomizes patients with MSI-H/dMMR mCRC to either atezolizumab, an anti-PD-L1 antibody, alone or in combination with mFOLFOX6 plus bevacizumab (NCT02997228) [85]. These two ongoing clinical trials might answer the question of which would be a better partner for PD-1 antibodies in the treatment of patients with MSI-H/dMMR mCRC: cytotoxic chemotherapy or a CTLA-4 antibody.
The investigation of the underlying mechanism of resistance is another fantastic approach. In the consensus molecular subtyping (CMS), the majority of patients with MSI-H/dMMR mCRC were categorized into CMS1, whereas some patients were classified as CMS4 (mesenchymal subtype) in which transforming growth factor-β (TGFβ) signaling plays an important role [86]. CMS has been constructed using data from patients with resected CRC. Interestingly, Lenz et al. reported that 24% of MSI-H/dMMR mCRC cases were classified into CMS4 in a post hoc analysis of the CALGB/SWOB80405 trial [87]. Some studies have indicated that activated TGFβ signaling prevents the response to ICIs [79,88,89,90]. The inhibition of TGFβ promoted the immunosuppressive tumor microenvironment, resulting in the increased efficacy of cancer immunotherapy [91,92]. A phase Ib/IIa trial evaluating the combination of pembrolizumab and TEW-7197, a highly selective and potent inhibitor of TGFβ receptor type 1, is ongoing (NCT03724851) [93].
Modulating the nonimmunogenic tumor microenvironment, the so-called “cold tumor,” to immunogenic circumstance, a “hot tumor,” is also a “hot” topic. Upfront FOLFOXIRI plus bevacizumab with atezolizumab showed promising results in a multicenter, open-label, randomized phase II trial in which investigators hypothesized that an intensified chemotherapy could enhance immunologic cell death and improve the efficacy of immunotherapy [94]. In the subgroup analysis of 13 patients with MSI-H/dMMR mCRC, the addition of atezolizumab to the intensified triplet chemotherapy boosted the efficacy regarding PFS (HR, 0.10; 95% CI, 0.02–0.51); however, whether this dramatic benefit would be translated into OS is uncertain.
The understanding of angiogenesis in the tumor microenvironment may contribute to overcoming de novo resistance to anti-PD-1 antibodies in MSI-H/dMMR tumors. Vascular endothelial growth factor (VEGF) inhibitors could modify the tumor microenvironment by increasing infiltrated CD8+ T cells and PD-L1 expression in tumor cells [95]. Wallin et al. showed that the blockade of VEGF could increase the secretion of interferon-γ, leading to the upregulation of major histocompatibility complex (MHC) class I expression [96]. Supporting this hypothesis, cases with MSI-H/dMMR CRC benefit from bevacizumab were reported in the post hoc analysis of CALGB/SWOG80407 and NSABP C-08 [97,98]. Notably, the improved immunomodulation induced by VEGF inhibition might be applicable regardless of MSI/MMR status. Some types of antiangiogenic multiple receptor tyrosine kinase inhibitors (RTKis) including regorafenib and lenvatinib are considered to reduce tumor-associated macrophages and regulatory T cells in the tumor microenvironment by regulating tumor angiogenesis [99,100]. The combination of regorafenib plus nivolumab, and lenvatinib plus pembrolizumab, exhibited ideal tumor growth suppression in mCRC and metastatic GC/GEJC, regardless of the MSI status [101,102,103]. At present, a confirmatory phase III trial in metastatic GC/GEJC is ongoing (NCT04662710). By resolving certain toxicities, such as rash or immune-related adverse events induced by immunotherapy plus RTKi, this combination strategy might become one of the essential options for MSI-H/dMMR GI cancers.
A dual blockade of PD-1/PD-L1 and other immune checkpoint molecules such as TIM-3 and LAG-3 might also enhance the efficacy and overcome the resistance mechanism for immunotherapy in MSI-H/dMMR tumors. TIM-3, which is expressed on multiple immune cells such as cytotoxic T cells, regulatory T cells, NK cells, and dendritic cells, suppresses antitumor immune response by interacting with carcinoembryonic antigen-related cell adhesion molecule 1, high mobility group box 1, galectin-9, and phosphatidylserine. LAG-3 expresses on activated CD8+ T cells and regulatory T cells; the interaction between LAG-3 and MHC on APC downregulates T-cell response [104,105,106,107]. In MSI-H CRC, PD-L1, and LAG-3 overexpressions were associated with worse relapse-free survival [108]. Although this novel combination strategy of blocking multiple immune checkpoints focuses on MSI-H/dMMR solid tumors in the exploratory phase II trials [109,110], these attempts are expected to benefit beyond the MSI status [111,112,113].

5. An Avenue for Innovative Immunotherapy for MSI-H/dMMR GI Cancers: From Care to Cure

Based on excellent outcomes in patients with metastatic MSI-H/dMMR GI cancers, it was hypothesized that immunotherapy could be successfully offered in patients with localized MSI-H/dMMR GI cancers. Preclinical data suggests the evidence that the intact primary tumor provides high-quality antigens for immune priming and the expansion of activated tumor-specific T cells [114,115]. As expected, but not surprisingly, immunotherapy for MSI-H/dMMR GI cancers has revolutionized the field of locoregional cancers. Exploratory clinical trials have demonstrated the potential of evolving neoadjuvant immunotherapy to make the “paradigm shift” in cancer treatment (Table 4), which includes the possibility that MSI-H/dMMR cancers could be cured without operative management.

5.1. CRC

The efficacy of immune checkpoint blockade is more enhanced in early-stage melanoma, lung cancer, and bladder cancer than those in advanced cases, and this is because increased T-cell infiltration and less systemic immunosuppression have been noted in early-stage tumors compared with metastatic tumors [122,123,124]. These successes encourage the induction of neoadjuvant immunotherapy to locoregional CRCs.
Chalabi et al. conducted the exploratory NICHE study to investigate the safety, feasibility, and activity of nivolumab plus ipilimumab with or without celecoxib (cyclooxygenase-2 inhibitor) in patients with nonmetastatic CRCs. In the NICHE trial, 40 patients with locally advanced CRC (20 patients with dMMR, 19 patients with pMMR, and one patient with dMMR- and pMMR-CRC) were enrolled and received nivolumab monotherapy or combination treatment with nivolumab 3 mg/kg plus ipilimumab 1 mg/kg just one cycle before surgery [116]. Celecoxib, as an immune modulator, was added in eight patients with pMMR. Ultimately, a total of 37 patients (20 dMMR, 8 pMMR, and 7 pMMR with celecoxib) were evaluable for the efficacy and translational endpoints. Notably, 95% of the patients had a major pathological response (MPR) with ≤10% residual viable tumor and 60% (95% CI, 36–81) achieved pathological CR (pCR) [116]. The final efficacy data of the NICHE study was presented at the ASCO annual meeting in 2022. Thirty-two patients with dMMR colon cancer were included in the final analysis, and MPR was observed in 97% of the patients [125]. Although only 2 of 32 patients with dMMR received adjuvant chemotherapy after curative resection, recurrence was not observed at the final cutoff date (median follow-up, 32 months). In the first-line treatment of mCRC, the response rate to nivolumab plus ipilimumab was reported to be 69% (95% CI, 53–82) [26]. The increased efficacy was consistently confirmed in early-stage colon cancers. Subsequent trials (NICHE-2 study) have also evaluated the efficacy of one ipilimumab dose and two doses of nivolumab in a larger sample of patients with locally advanced dMMR CRC. Of the 112 patients treated with neoadjuvant immunotherapy, MPR of 95% and pCR of 67% were observed, with a low incidence of severe adverse events or only three patients having a delay in subsequent surgery [117]. The small sample size and insufficient follow-up period restricted providing conclusive evidence, whereas the amazing results led us to expect that locally advanced MSI-H/dMMR CRC could be cured without chemotherapy and that neoadjuvant immunotherapy will be a new standard of care for this population. Similar attempts using immunotherapy for locally advanced MSI-H/dMMR CRC are ongoing in phase II trials (NCT05371197 and NCT04082572 [126]). Although MSI-H/dMMR is a small subset (<5%) in mCRC, the proportion of MSI-H/dMMR CRC is known to increase to approximately 10% in stage III and 20% in stage II [4]. Additionally, they indicated that there were some responders to an anti-PD-1 blockade, even in pMMR CRCs. However, MSI-H/dMMR CRC is generally associated with a favorable prognosis in stage II/III CRC. The 5-year disease-free survival and OS were 84% and 86% in stage II and 65% and 72% in patients with stage III CRC treated by surgery alone [127]. Strong evidence would be required to change the clinical practice.
The cure for locally advanced rectal cancer involved multimodality therapy including chemotherapy, radiation, and surgery. However, bowel and bladder dysfunction, sexual dysfunction, infertility, and permanent/temporal stoma are serious life-altering consequences after radical resection. A growing interest in nonoperative management emerges for responders to the preoperative treatment. Recently, a promising result was revealed at the ASCO annual meeting in 2022 and immediately published in the New England Journal of Medicine [118]. Cercek et al. reported the preliminary result of an ongoing prospective single-arm study with dostarlimab for locally advanced rectal cancer with dMMR (NCT04165772). Dostarlimab was administered every 3 weeks for 6 months before surgery, and surprisingly, 14 consecutive patients achieved a clinical CR (100%), and all of them avoided surgical resection [118]. Durable responses were seen in four patients over 1 year after the completion of dostarlimab treatment. Given the small sample and short follow-up, this result was immature for efficacy evaluation to date. However, the amazing result led us to expect that preoperative chemotherapy or chemoradiotherapy and even surgery could be omitted by neoadjuvant immunotherapy. A low prevalence of dMMR in rectal cancer (5–10%) would be a hazard in establishing neoadjuvant immunotherapy as the standard of care.
A similar finding has been recently reported by Kaysia et al. in which 35 patients with localized unresectable or high-risk resectable MSI-H/dMMR cancers, including 32 harboring gastrointestinal cancers, were treated with neoadjuvant pembrolizumab 200 mg once every 3 weeks for 6 months followed by surgical resection [121]. Of 17 patients who underwent surgery, pCR was obtained in 65% with manageable toxicity. Of the 35 patients including 18 patients who did not receive surgery for any reason, 33 were evaluable for radiographic response, with a CR of 30% and a PR of 52%, whereas the association between pCR and radiographic CR was not observed. In this trial, 27 patients with CRC were enrolled, and 11 of the 14 patients who underwent surgical resection had a pCR (79%), which was consistent with the results of other clinical trials.
Another approach is the combination of immunotherapy and preoperative chemoradiotherapy. Bando et al. investigated the efficacy of chemoradiation therapy followed by consolidation nivolumab for T3–4 N0–2 rectal cancer for which they hypothesized that chemoradiotherapy could enhance the efficacy of immunotherapy by modifying the tumor immune microenvironment even for pMMR rectal cancers [128]. In the exploratory analysis of an MSI-H cohort, a total of five patients were enrolled, and 60% of them achieved a pCR with a long-lasting response. The confirmed pCR rate accounted for 30% (11/37) in the MSS cohort. In 24 patients with MSS with assessable clinical samples, the translation research indicated that patients whose tumor microenvironment had changed immunologically after chemoradiation experienced higher efficacy than those without a change in the tumor microenvironment. This result is in line with those of other clinical trials showing the immune-modulating effect by chemoradiation, and a proof-of-concept study for modifying “cold tumor” to “hot tumor” was attempted. Thus, neoadjuvant immunotherapy combined with chemoradiation could be an innovative therapeutic strategy for originally immune suppressive phenotypes, such as pMMR tumors.

5.2. GC/GEJC

As we have seen, the predictive value of MSI-H for immune checkpoint blockade seemed less significant for non-CRC GI malignancies than CRC in the metastatic setting. However, neoadjuvant immunotherapy is also underway in locally advanced MSI-H/dMMR GC/GEJC. Pietrantonio et al. reported that perioperative chemotherapy with cytotoxic chemotherapy offers questionable benefits or is even detrimental for patients with MSI-H/dMMR locally advanced GC/GEJC [129]. In the GERCOR NEONIPIGA phase II study (NCT04006262), 32 patients with MSI-H/dMMR locally advanced GC/GEJC received neoadjuvant nivolumab 480 mg/body plus ipilimumab 1 mg/kg for six cycles followed by adjuvant nivolumab demonstrated impressive efficacy: the pCR, the primary endpoint, was 58.6%, and 30 of the 31 patients who underwent curative surgery were alive without recurrence at a median follow-up of 12 months [119].
The noteworthy attempt to improve the power of neoadjuvant therapy for locally advanced MSI-H/dMMR GC/GEJC shed light on the combination of cytotoxic chemotherapy and immunotherapy. The DANTE trial is a randomized phase IIb trial investigating the addition of atezolizumab, an anti-PD-L1 antibody, to perioperative FLOT (5-FU, leucovorin, oxaliplatin, and docetaxel) (NCT03421288). A total of 23 patients with locally advanced MSI-H/dMMR GC/GEJC were enrolled (8 patients with FLOT plus atezolizumab arm and 15 with FLOT arm): the pCR rates by central assessment were 63% and 27%, respectively, suggesting the benefit of adding immunotherapy to chemotherapy [120]. The enhancement of adding atezolizumab to chemotherapy was not observed in all patients (25% in the FLOT plus atezolizumab group and 24% in the FLOT group), especially in patients with a low PD-L1 expression. Therefore, immunotherapy with or without chemotherapy is consistently efficacious for patients with MSI-H/dMMR GC/GEJC both in early and advanced stages. However, the encouraging preliminary result was reported in the ASCO annual meeting in 2022. Sun et al. initiated a phase II study evaluating perioperative FOLFOX plus pembrolizumab for resectable GC/GEJC irrelevant of the MSI status [130]. The pCR rate was 22%, which was higher than the pCR rates of previous studies using the doublet regimen and similar to the results reported in previous studies adopting the FLOT regimen [120]. These results indicated the potential efficacy of response to neoadjuvant immunotherapy for patients with resectable GC/GEJC whose tumors are non-MSI-H, such as PD-L1-high/MSS. Larger-scale randomized clinical trials evaluating the combination of immunotherapy and chemotherapy are ongoing beyond the MSI status [131,132]. As described in the aforementioned section, limiting the indication of immunotherapy to MSI-H/dMMR GC/GEJC does not appear to be a wise decision in resectable-stage GC/GEJC.
In total, the ideal goal of cancer treatment for locally advanced MSI-H/dMMR GI cancers is to improve the curability by reducing treatment-related adverse events, and neoadjuvant immunotherapy for rectal cancer potentially could achieve the goal without operative management. Overcoming the difficulties of conducting randomized phase III trials should be discussed more. The clinical trials for MSI-H/dMMR have highlighted the clinically meaningful effect of biomarker-driven therapy and substantiated precision medicine in the field of early-stage GI malignancy.

6. Future Perspectives on MSI-H/dMMR Cancers

In the early 2010s, MSI-H/dMMR tumors were the first type of tumors that led to our current perception of immunogenic tumors and, consequently, of immunotherapy in GI malignancy. In this section, we envisioned the next frontiers in the therapy of immunogenic GI cancers. An apparent direction for this is the identification of more precise and fundamental biomarkers beyond the MSI/MMR status, which is the surrogate predictive marker for ICIs. However, theoretically, neoantigens, tumor-infiltrating T-cell activation, and the immune reactive tumor microenvironment are part of the immune system; therefore, the status of these parameters would be enlightening for a more precise prediction of immunotherapy efficacy. The tumor mutation burden (TMB), as calculated based on the total number of nonsynonymous mutations per megabase of tumor DNA, is correlated with the magnitude of tumor-infiltrating T cells or the immune-related gene expression signatures in certain types of tumors, such as melanoma or NSCLC, and can predict the response to immunotherapy [133,134,135]. A high TMB was the subsequent FDA-approved tissue-agnostic biomarker for immunotherapy based on the results of KEYNOTE-158 [136].
However, it is worth noting that there is presumably considerable overlap between the TMB-high and MSI-H/dMMR populations in GI cancers and that the ultra-high mutation load caused by POLE/POLD1 aberrations is extremely rare [137]. Schrock et al. reported that patients with TMB-high (defined as ≥37–41 mut/Mb) and MSI-H mCRC responded to immunotherapy more efficiently than did those with TMB-low (<37–41 mut/Mb) and MSI-H mCRC [80,138,139]. A TMB-high status might better refine the efficacy of immunotherapy in MSI-H/dMMR GI cancers. However, it might not directly reflect the tumor microenvironment or the immune gene signatures in the majority of GI cancers, including MSS-CRC or MSS-GC/GECJ in which the magnitude of the TMB and tumor microenvironment are not closely associated with each other.
Not only the “quantity” but also the “quality” of tumor-specific neoantigens might be important in this context [140]. The diverse and high-quality neoantigens that can be presented to multiple APCs induce infiltration of activated T cells [141]. Currently, we only have a snapshot of tumor immunity projected through TMB or MSI/MMR status at a given moment. However, recent advances in neoantigen evaluation will help us to optimize the subjects of immunotherapy. Moreover, next-generation sequencing (NGS)-based technology could offer the MSI status and TMB with comprehensive genomic profiles [142,143,144,145]. In the near future, NGS-based technology will enable the provision of information about the quantity and quality of neoantigens in combination with transcriptome data. Comprehensive NGS-based evaluations will replace the conventional PCR-based MSI testing or IHC testing for MMR, adjourning the MSI/MMR status, and bring the value of immunotherapy to a new level.
Exploring the candidates for predicting the immunogenicity of tumors is also underway including POLE/POLD1 mutation, chromosomal instability (CIN), and homologous recombination deficiency (HRD) [146,147,148]. CIN along with MSI is the fundamental nature of all malignant tumors, including CRCs and GCs [55,149,150]. CIN is thought to be located on the opposite side of MSI-high in the carcinogenesis of CRCs [151], whereas the mechanism underlying the endogenous immune reaction in CIN tumors has not been fully elucidated. Thus, understanding the immunogenicity of GI tumors harboring CIN could expand the boundaries of immunotherapy. Expanding the knowledge of neoantigens from the comprehensive concept of genomic instability might help us deeply understand the association between each type of genomic instability and carcinogenesis, the generation of high-quality neoantigens, and the magnitude of immunogenicity, opening the door to precision immune oncology beyond the MSI status in GI malignancies.

7. Conclusions

Our review highlighted the clinical trials of immunotherapy for MSI-H/dMMR GI malignancy. The era of immunotherapy for MSI-H/dMMR tumors has originated from man’s success and has now expanded beyond the tumor origin. Finally, pembrolizumab was approved as the first tumor-agnostic agent in 2017 and established as the standard treatment in the initial-line therapy of mCRC in 2020. The dual immune checkpoint blockades or combination therapy with chemotherapy could overcome the primary and acquired resistance to immunotherapy in MSI-H/dMMR GI cancers. Followed by the successes in controlling the metastatic setting, the next therapeutic goal of immunotherapy in GI malignancy is to achieve a cure. An understanding of tumor immunity from a broader perspective is absolutely required to guide us to overcome these malignancies.
MSI-H/dMMR tumor has been at the center of immune oncology in GI malignancies for a decade. More precise and fundamental biomarkers beyond MSI status will be detected by the recent progress in immune oncology and technical development in the near future. The end of the “MSI era” will be the beginning of the next precision immune oncology.

Author Contributions

K.S. drew conceptual frameworks, searched the literature, and wrote the manuscript; I.N. and T.H. supervised and revised the manuscript; K.Y. supervised and administered the project. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Brahmer, J.R.; Drake, C.G.; Wollner, I.; Powderly, J.D.; Picus, J.; Sharfman, W.H.; Stankevich, E.; Pons, A.; Salay, T.M.; McMiller, T.L.; et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: Safety, clinical activity, pharmacodynamics, and immunologic correlates. J. Clin. Oncol. 2010, 28, 3167–3175. [Google Scholar] [CrossRef] [PubMed]
  2. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lipson, E.J.; Sharfman, W.H.; Drake, C.G.; Wollner, I.; Taube, J.M.; Anders, R.A.; Xu, H.; Yao, S.; Pons, A.; Chen, L.; et al. Durable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibody. Clin. Cancer Res. 2013, 19, 462–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Koopman, M.; Kortman, G.A.; Mekenkamp, L.; Ligtenberg, M.J.; Hoogerbrugge, N.; Antonini, N.F.; Punt, C.J.; van Krieken, J.H. Deficient mismatch repair system in patients with sporadic advanced colorectal cancer. Br. J. Cancer 2009, 100, 266–273. [Google Scholar] [CrossRef] [Green Version]
  5. Goldstein, J.; Tran, B.; Ensor, J.; Gibbs, P.; Wong, H.L.; Wong, S.F.; Vilar, E.; Tie, J.; Broaddus, R.; Kopetz, S.; et al. Multicenter retrospective analysis of metastatic colorectal cancer (CRC) with high-level microsatellite instability (MSI-H). Ann. Oncol. 2014, 25, 1032–1038. [Google Scholar] [CrossRef]
  6. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef] [Green Version]
  7. Li, G.M. Mechanisms and functions of DNA mismatch repair. Cell Res. 2008, 18, 85–98. [Google Scholar] [CrossRef] [Green Version]
  8. Kunkel, T.A.; Erie, D.A. DNA mismatch repair. Annu. Rev. Biochem. 2005, 74, 681–710. [Google Scholar] [CrossRef] [Green Version]
  9. Schroering, A.G.; Edelbrock, M.A.; Richards, T.J.; Williams, K.J. The cell cycle and DNA mismatch repair. Exp. Cell Res. 2007, 313, 292–304. [Google Scholar] [CrossRef]
  10. Aaltonen, L.A.; Peltomaki, P.; Leach, F.S.; Sistonen, P.; Pylkkanen, L.; Mecklin, J.P.; Jarvinen, H.; Powell, S.M.; Jen, J.; Hamilton, S.R.; et al. Clues to the pathogenesis of familial colorectal cancer. Science 1993, 260, 812–816. [Google Scholar] [CrossRef]
  11. Eso, Y.; Shimizu, T.; Takeda, H.; Takai, A.; Marusawa, H. Microsatellite instability and immune checkpoint inhibitors: Toward precision medicine against gastrointestinal and hepatobiliary cancers. J. Gastroenterol. 2020, 55, 15–26. [Google Scholar] [CrossRef] [Green Version]
  12. Lynch, H.T.; de la Chapelle, A. Genetic susceptibility to non-polyposis colorectal cancer. J. Med. Genet. 1999, 36, 801–818. [Google Scholar] [PubMed]
  13. Goodman, A.M.; Kato, S.; Bazhenova, L.; Patel, S.P.; Frampton, G.M.; Miller, V.; Stephens, P.J.; Daniels, G.A.; Kurzrock, R. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol. Cancer Ther. 2017, 16, 2598–2608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef] [PubMed]
  15. Taube, J.M.; Klein, A.; Brahmer, J.R.; Xu, H.; Pan, X.; Kim, J.H.; Chen, L.; Pardoll, D.M.; Topalian, S.L.; Anders, R.A. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin. Cancer Res. 2014, 20, 5064–5074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Llosa, N.J.; Cruise, M.; Tam, A.; Wicks, E.C.; Hechenbleikner, E.M.; Taube, J.M.; Blosser, R.L.; Fan, H.; Wang, H.; Luber, B.S.; et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015, 5, 43–51. [Google Scholar] [CrossRef] [Green Version]
  17. Lee, V.; Murphy, A.; Le, D.T.; Diaz, L.A., Jr. Mismatch Repair Deficiency and Response to Immune Checkpoint Blockade. Oncologist 2016, 21, 1200–1211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Dudley, J.C.; Lin, M.T.; Le, D.T.; Eshleman, J.R. Microsatellite Instability as a Biomarker for PD-1 Blockade. Clin. Cancer Res. 2016, 22, 813–820. [Google Scholar] [CrossRef] [Green Version]
  19. Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef] [Green Version]
  20. Saeterdal, I.; Bjorheim, J.; Lislerud, K.; Gjertsen, M.K.; Bukholm, I.K.; Olsen, O.C.; Nesland, J.M.; Eriksen, J.A.; Moller, M.; Lindblom, A.; et al. Frameshift-mutation-derived peptides as tumor-specific antigens in inherited and spontaneous colorectal cancer. Proc. Natl. Acad. Sci. USA 2001, 98, 13255–13260. [Google Scholar] [CrossRef] [Green Version]
  21. Le, D.T.; Kim, T.W.; Van Cutsem, E.; Geva, R.; Jager, D.; Hara, H.; Burge, M.; O’Neil, B.; Kavan, P.; Yoshino, T.; et al. Phase II Open-Label Study of Pembrolizumab in Treatment-Refractory, Microsatellite Instability-High/Mismatch Repair-Deficient Metastatic Colorectal Cancer: KEYNOTE-164. J. Clin. Oncol. 2020, 38, 11–19. [Google Scholar] [CrossRef]
  22. Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Andre, T.; Shiu, K.K.; Kim, T.W.; Jensen, B.V.; Jensen, L.H.; Punt, C.; Smith, D.; Garcia-Carbonero, R.; Benavides, M.; Gibbs, P.; et al. Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer. N. Engl. J. Med. 2020, 383, 2207–2218. [Google Scholar] [CrossRef]
  24. Overman, M.J.; McDermott, R.; Leach, J.L.; Lonardi, S.; Lenz, H.J.; Morse, M.A.; Desai, J.; Hill, A.; Axelson, M.; Moss, R.A.; et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): An open-label, multicentre, phase 2 study. Lancet Oncol. 2017, 18, 1182–1191. [Google Scholar] [CrossRef]
  25. Overman, M.J.; Lonardi, S.; Wong, K.Y.M.; Lenz, H.J.; Gelsomino, F.; Aglietta, M.; Morse, M.A.; Van Cutsem, E.; McDermott, R.; Hill, A.; et al. Durable Clinical Benefit With Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer. J. Clin. Oncol. 2018, 36, 773–779. [Google Scholar] [CrossRef]
  26. Lenz, H.J.; Van Cutsem, E.; Luisa Limon, M.; Wong, K.Y.M.; Hendlisz, A.; Aglietta, M.; Garcia-Alfonso, P.; Neyns, B.; Luppi, G.; Cardin, D.B.; et al. First-Line Nivolumab Plus Low-Dose Ipilimumab for Microsatellite Instability-High/Mismatch Repair-Deficient Metastatic Colorectal Cancer: The Phase II CheckMate 142 Study. J. Clin. Oncol. 2021, 40, 161–170. [Google Scholar] [CrossRef] [PubMed]
  27. Marabelle, A.; Le, D.T.; Ascierto, P.A.; Di Giacomo, A.M.; De Jesus-Acosta, A.; Delord, J.P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; et al. Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J. Clin. Oncol. 2020, 38, 1–10. [Google Scholar] [CrossRef] [PubMed]
  28. Marcus, L.; Lemery, S.J.; Keegan, P.; Pazdur, R. FDA Approval Summary: Pembrolizumab for the Treatment of Microsatellite Instability-High Solid Tumors. Clin. Cancer Res. 2019, 25, 3753–3758. [Google Scholar] [CrossRef] [Green Version]
  29. Muro, K.; Chung, H.C.; Shankaran, V.; Geva, R.; Catenacci, D.; Gupta, S.; Eder, J.P.; Golan, T.; Le, D.T.; Burtness, B.; et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): A multicentre, open-label, phase 1b trial. Lancet Oncol. 2016, 17, 717–726. [Google Scholar] [CrossRef] [PubMed]
  30. Hewish, M.; Lord, C.J.; Martin, S.A.; Cunningham, D.; Ashworth, A. Mismatch repair deficient colorectal cancer in the era of personalized treatment. Nat. Rev. Clin. Oncol. 2010, 7, 197–208. [Google Scholar] [CrossRef] [PubMed]
  31. Koinuma, K.; Shitoh, K.; Miyakura, Y.; Furukawa, T.; Yamashita, Y.; Ota, J.; Ohki, R.; Choi, Y.L.; Wada, T.; Konishi, F.; et al. Mutations of BRAF are associated with extensive hMLH1 promoter methylation in sporadic colorectal carcinomas. Int. J. Cancer 2004, 108, 237–242. [Google Scholar] [CrossRef] [PubMed]
  32. Tougeron, D.; Sueur, B.; Zaanan, A.; de la Fouchardiere, C.; Sefrioui, D.; Lecomte, T.; Aparicio, T.; Des Guetz, G.; Artru, P.; Hautefeuille, V.; et al. Prognosis and chemosensitivity of deficient MMR phenotype in patients with metastatic colorectal cancer: An AGEO retrospective multicenter study. Int. J. Cancer 2020, 147, 285–296. [Google Scholar] [CrossRef] [PubMed]
  33. Casak, S.J.; Marcus, L.; Fashoyin-Aje, L.; Mushti, S.L.; Cheng, J.; Shen, Y.L.; Pierce, W.F.; Her, L.; Goldberg, K.B.; Theoret, M.R.; et al. FDA Approval Summary: Pembrolizumab for the First-line Treatment of Patients with MSI-H/dMMR Advanced Unresectable or Metastatic Colorectal Carcinoma. Clin. Cancer Res. 2021, 27, 4680–4684. [Google Scholar] [CrossRef]
  34. Diaz, L.A., Jr.; Shiu, K.K.; Kim, T.W.; Jensen, B.V.; Jensen, L.H.; Punt, C.; Smith, D.; Garcia-Carbonero, R.; Benavides, M.; Gibbs, P.; 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, 659–670. [Google Scholar] [CrossRef] [PubMed]
  35. Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [Green Version]
  36. Garon, E.B.; Rizvi, N.A.; Hui, R.; Leighl, N.; Balmanoukian, A.S.; Eder, J.P.; Patnaik, A.; Aggarwal, C.; Gubens, M.; Horn, L.; et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 2015, 372, 2018–2028. [Google Scholar] [CrossRef] [PubMed]
  37. Janjigian, Y.Y.; Bendell, J.; Calvo, E.; Kim, J.W.; Ascierto, P.A.; Sharma, P.; Ott, P.A.; Peltola, K.; Jaeger, D.; Evans, J.; et al. CheckMate-032 Study: Efficacy and Safety of Nivolumab and Nivolumab Plus Ipilimumab in Patients With Metastatic Esophagogastric Cancer. J. Clin. Oncol. 2018, 36, 2836–2844. [Google Scholar] [CrossRef] [PubMed]
  38. Kim, J.W.; Nam, K.H.; Ahn, S.H.; Park, D.J.; Kim, H.H.; Kim, S.H.; Chang, H.; Lee, J.O.; Kim, Y.J.; Lee, H.S.; et al. Prognostic implications of immunosuppressive protein expression in tumors as well as immune cell infiltration within the tumor microenvironment in gastric cancer. Gastric Cancer 2016, 19, 42–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Qing, Y.; Li, Q.; Ren, T.; Xia, W.; Peng, Y.; Liu, G.L.; Luo, H.; Yang, Y.X.; Dai, X.Y.; Zhou, S.F.; et al. Upregulation of PD-L1 and APE1 is associated with tumorigenesis and poor prognosis of gastric cancer. Drug Des. Devel. Ther. 2015, 9, 901–909. [Google Scholar] [CrossRef] [Green Version]
  40. Sun, J.; Xu, K.; Wu, C.; Wang, Y.; Hu, Y.; Zhu, Y.; Chen, Y.; Shi, Q.; Yu, G.; Zhang, X. PD-L1 expression analysis in gastric carcinoma tissue and blocking of tumor-associated PD-L1 signaling by two functional monoclonal antibodies. Tissue Antigens 2007, 69, 19–27. [Google Scholar] [CrossRef]
  41. Fuchs, C.S.; Doi, T.; Jang, R.W.; Muro, K.; Satoh, T.; Machado, M.; Sun, W.; Jalal, S.I.; Shah, M.A.; Metges, J.P.; et al. Safety and Efficacy of Pembrolizumab Monotherapy in Patients with Previously Treated Advanced Gastric and Gastroesophageal Junction Cancer: Phase 2 Clinical KEYNOTE-059 Trial. JAMA Oncol. 2018, 4, e180013. [Google Scholar] [CrossRef]
  42. Chao, J.; Fuchs, C.S.; Shitara, K.; Tabernero, J.; Muro, K.; Van Cutsem, E.; Bang, Y.J.; De Vita, F.; Landers, G.; Yen, C.J.; et al. Assessment of Pembrolizumab Therapy for the Treatment of Microsatellite Instability-High Gastric or Gastroesophageal Junction Cancer Among Patients in the KEYNOTE-059, KEYNOTE-061, and KEYNOTE-062 Clinical Trials. JAMA Oncol. 2021, 7, 895–902. [Google Scholar] [CrossRef] [PubMed]
  43. Kang, Y.K.; Boku, N.; Satoh, T.; Ryu, M.H.; Chao, Y.; Kato, K.; Chung, H.C.; Chen, J.S.; Muro, K.; Kang, W.K.; et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 390, 2461–2471. [Google Scholar] [CrossRef] [PubMed]
  44. Lesterhuis, W.J.; Punt, C.J.; Hato, S.V.; Eleveld-Trancikova, D.; Jansen, B.J.; Nierkens, S.; Schreibelt, G.; de Boer, A.; Van Herpen, C.M.; Kaanders, J.H.; et al. Platinum-based drugs disrupt STAT6-mediated suppression of immune responses against cancer in humans and mice. J. Clin. Investig. 2011, 121, 3100–3108. [Google Scholar] [CrossRef]
  45. Roselli, M.; Cereda, V.; di Bari, M.G.; Formica, V.; Spila, A.; Jochems, C.; Farsaci, B.; Donahue, R.; Gulley, J.L.; Schlom, J.; et al. Effects of conventional therapeutic interventions on the number and function of regulatory T cells. Oncoimmunology 2013, 2, e27025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Bracci, L.; Schiavoni, G.; Sistigu, A.; Belardelli, F. Immune-based mechanisms of cytotoxic chemotherapy: Implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Differ. 2014, 21, 15–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Wang, Z.; Till, B.; Gao, Q. Chemotherapeutic agent-mediated elimination of myeloid-derived suppressor cells. Oncoimmunology 2017, 6, e1331807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Galluzzi, L.; Buque, A.; Kepp, O.; Zitvogel, L.; Kroemer, G. Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol. 2017, 17, 97–111. [Google Scholar] [CrossRef]
  49. Fucikova, J.; Kepp, O.; Kasikova, L.; Petroni, G.; Yamazaki, T.; Liu, P.; Zhao, L.; Spisek, R.; Kroemer, G.; Galluzzi, L. Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 2020, 11, 1013. [Google Scholar] [CrossRef]
  50. West, H.; McCleod, M.; Hussein, M.; Morabito, A.; Rittmeyer, A.; Conter, H.J.; Kopp, H.G.; Daniel, D.; McCune, S.; Mekhail, T.; et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 924–937. [Google Scholar] [CrossRef]
  51. Paz-Ares, L.; Ciuleanu, T.E.; Cobo, M.; Schenker, M.; Zurawski, B.; Menezes, J.; Richardet, E.; Bennouna, J.; Felip, E.; Juan-Vidal, O.; et al. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): An international, randomised, open-label, phase 3 trial. Lancet Oncol. 2021, 22, 198–211. [Google Scholar] [CrossRef] [PubMed]
  52. Janjigian, Y.Y.; Shitara, K.; Moehler, M.; Garrido, M.; Salman, P.; Shen, L.; Wyrwicz, L.; Yamaguchi, K.; Skoczylas, T.; Campos Bragagnoli, A.; et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): A randomised, open-label, phase 3 trial. Lancet 2021, 398, 27–40. [Google Scholar] [CrossRef] [PubMed]
  53. Shitara, K.; Ajani, J.A.; Moehler, M.; Garrido, M.; Gallardo, C.; Shen, L.; Yamaguchi, K.; Wyrwicz, L.; Skoczylas, T.; Bragagnoli, A.C.; et al. Nivolumab plus chemotherapy or ipilimumab in gastro-oesophageal cancer. Nature 2022, 603, 942–948. [Google Scholar] [CrossRef]
  54. Kang, Y.K.; Chen, L.T.; Ryu, M.H.; Oh, D.Y.; Oh, S.C.; Chung, H.C.; Lee, K.W.; Omori, T.; Shitara, K.; Sakuramoto, S.; et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): A randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2022, 23, 234–247. [Google Scholar] [CrossRef]
  55. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Cristescu, R.; Lee, J.; Nebozhyn, M.; Kim, K.M.; Ting, J.C.; Wong, S.S.; Liu, J.; Yue, Y.G.; Wang, J.; Yu, K.; et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat. Med. 2015, 21, 449–456. [Google Scholar] [CrossRef] [PubMed]
  57. Yaeger, R.; Chatila, W.K.; Lipsyc, M.D.; Hechtman, J.F.; Cercek, A.; Sanchez-Vega, F.; Jayakumaran, G.; Middha, S.; Zehir, A.; Donoghue, M.T.A.; et al. Clinical Sequencing Defines the Genomic Landscape of Metastatic Colorectal Cancer. Cancer Cell 2018, 33, 125–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Matsumoto, Y.; Nagasaka, T.; Kambara, T.; Hoshizima, N.; Murakami, J.; Sasamoto, H.; Hosokawa, M.; Naomoto, Y.; Isozaki, H.; Shimizu, K.; et al. Microsatellite instability and clinicopathological features in esophageal squamous cell cancer. Oncol. Rep. 2007, 18, 1123–1127. [Google Scholar]
  59. Campanella, N.C.; Lacerda, C.F.; Berardinelli, G.N.; Abrahao-Machado, L.F.; Cruvinel-Carloni, A.; De Oliveira, A.T.T.; Scapulatempo-Neto, C.; Crema, E.; Adad, S.J.; Rodrigues, M.A.M.; et al. Presence of microsatellite instability in esophageal squamous cell carcinoma associated with chagasic megaesophagus. Biomark Med. 2018, 12, 573–582. [Google Scholar] [CrossRef]
  60. Schulmann, K.; Brasch, F.E.; Kunstmann, E.; Engel, C.; Pagenstecher, C.; Vogelsang, H.; Kruger, S.; Vogel, T.; Knaebel, H.P.; Ruschoff, J.; et al. HNPCC-associated small bowel cancer: Clinical and molecular characteristics. Gastroenterology 2005, 128, 590–599. [Google Scholar] [CrossRef]
  61. Latham, A.; Srinivasan, P.; Kemel, Y.; Shia, J.; Bandlamudi, C.; Mandelker, D.; Middha, S.; Hechtman, J.; Zehir, A.; Dubard-Gault, M.; et al. Microsatellite Instability Is Associated With the Presence of Lynch Syndrome Pan-Cancer. J. Clin. Oncol. 2019, 37, 286–295. [Google Scholar] [CrossRef] [PubMed]
  62. Akagi, K.; Oki, E.; Taniguchi, H.; Nakatani, K.; Aoki, D.; Kuwata, T.; Yoshino, T. The real-world data on microsatellite instability status in various unresectable or metastatic solid tumors. Cancer Sci. 2021. online ahead of print. [Google Scholar] [CrossRef]
  63. Ott, P.A.; Bang, Y.J.; Piha-Paul, S.A.; Razak, A.R.A.; Bennouna, J.; Soria, J.C.; Rugo, H.S.; Cohen, R.B.; O’Neil, B.H.; Mehnert, J.M.; et al. T-Cell-Inflamed Gene-Expression Profile, Programmed Death Ligand 1 Expression, and Tumor Mutational Burden Predict Efficacy in Patients Treated With Pembrolizumab Across 20 Cancers: KEYNOTE-028. J. Clin. Oncol. 2019, 37, 318–327. [Google Scholar] [CrossRef]
  64. Piha-Paul, S.A.; Oh, D.Y.; Ueno, M.; Malka, D.; Chung, H.C.; Nagrial, A.; Kelley, R.K.; Ros, W.; Italiano, A.; Nakagawa, K.; et al. Efficacy and safety of pembrolizumab for the treatment of advanced biliary cancer: Results from the KEYNOTE-158 and KEYNOTE-028 studies. Int. J. Cancer 2020, 147, 2190–2198. [Google Scholar] [CrossRef] [PubMed]
  65. Andre, T.; Berton, D.; Curigliano, G.; Ellard, S.; Pérez, J.M.T.; Arkenau, H.-T.; Abdeddaim, C.; Moreno, V.; Guo, W.; Im, E.; et al. Safety and efficacy of anti–PD-1 antibody dostarlimab in patients (pts) with mismatch repair-deficient (dMMR) solid cancers: Results from GARNET study. J. Clin. Oncol. 2021, 39, 9. [Google Scholar] [CrossRef]
  66. Berton, D.; Banerjee, S.N.; Curigliano, G.; Cresta, S.; Arkenau, H.-T.; Abdeddaim, C.; Kristeleit, R.S.; Redondo, A.; Leath, C.A.; Torres, A.A.; et al. Antitumor activity of dostarlimab in patients with mismatch repair-deficient/microsatellite instability–high tumors: A combined analysis of two cohorts in the GARNET study. J. Clin. Oncol. 2021, 39, 2564. [Google Scholar] [CrossRef]
  67. Azad, N.S.; Gray, R.J.; Overman, M.J.; Schoenfeld, J.D.; Mitchell, E.P.; Zwiebel, J.A.; Sharon, E.; Streicher, H.; Li, S.; McShane, L.M.; et al. Nivolumab Is Effective in Mismatch Repair-Deficient Noncolorectal Cancers: Results From Arm Z1D-A Subprotocol of the NCI-MATCH (EAY131) Study. J. Clin. Oncol. 2020, 38, 214–222. [Google Scholar] [CrossRef] [PubMed]
  68. Mehnert, J.M.; Bergsland, E.; O’Neil, B.H.; Santoro, A.; Schellens, J.H.M.; Cohen, R.B.; Doi, T.; Ott, P.A.; Pishvaian, M.J.; Puzanov, I.; et al. Pembrolizumab for the treatment of programmed death-ligand 1-positive advanced carcinoid or pancreatic neuroendocrine tumors: Results from the KEYNOTE-028 study. Cancer 2020, 126, 3021–3030. [Google Scholar] [CrossRef]
  69. Strosberg, J.; Mizuno, N.; Doi, T.; Grande, E.; Delord, J.P.; Shapira-Frommer, R.; Bergsland, E.; Shah, M.; Fakih, M.; Takahashi, S.; et al. Efficacy and Safety of Pembrolizumab in Previously Treated Advanced Neuroendocrine Tumors: Results from the Phase II KEYNOTE-158 Study. Clin. Cancer Res. 2020, 26, 2124–2130. [Google Scholar] [CrossRef]
  70. Lu, M.; Zhang, P.; Zhang, Y.; Li, Z.; Gong, J.; Li, J.; Li, J.; Li, Y.; Zhang, X.; Lu, Z.; et al. Efficacy, Safety, and Biomarkers of Toripalimab in Patients with Recurrent or Metastatic Neuroendocrine Neoplasms: A Multiple-Center Phase Ib Trial. Clin. Cancer Res. 2020, 26, 2337–2345. [Google Scholar] [CrossRef] [Green Version]
  71. Jia, R.; Li, Y.; Xu, N.; Jiang, H.P.; Zhao, C.H.; Liu, R.R.; Shi, Y.; Zhang, Y.Y.; Wang, S.Y.; Zhou, H.; et al. Sintilimab in Patients with Previously Treated Metastatic Neuroendocrine Neoplasms. Oncologist 2022, 27, e625–e632. [Google Scholar] [CrossRef]
  72. Klein, O.; Kee, D.; Markman, B.; Lee, R.C.; Michael, M.; Mileshkin, L.R.; Scott, C.L.; Linklater, R.; Menon, S.; Tebbutt, N.C.; et al. A phase II clinical trial of ipilimumab/nivolumab combination immunotherapy in patients with rare upper gastrointestinal, neuroendocrine, and gynecological malignancies. J. Clin. Oncol. 2019, 37, 2570. [Google Scholar] [CrossRef]
  73. Amodio, V.; Mauri, G.; Reilly, N.M.; Sartore-Bianchi, A.; Siena, S.; Bardelli, A.; Germano, G. Mechanisms of Immune Escape and Resistance to Checkpoint Inhibitor Therapies in Mismatch Repair Deficient Metastatic Colorectal Cancers. Cancers 2021, 13, 2638. [Google Scholar] [CrossRef]
  74. Kalbasi, A.; Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 2020, 20, 25–39. [Google Scholar] [CrossRef] [PubMed]
  75. Dhatchinamoorthy, K.; Colbert, J.D.; Rock, K.L. Cancer Immune Evasion Through Loss of MHC Class I Antigen Presentation. Front Immunol. 2021, 12, 636568. [Google Scholar] [CrossRef] [PubMed]
  76. Batlle, E.; Massague, J. Transforming Growth Factor-beta Signaling in Immunity and Cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef] [PubMed]
  77. Yang, L.; Pang, Y.; Moses, H.L. TGF-beta and immune cells: An important regulatory axis in the tumor microenvironment and progression. Trends Immunol. 2010, 31, 220–227. [Google Scholar] [CrossRef] [Green Version]
  78. Bortolomeazzi, M.; Keddar, M.R.; Montorsi, L.; Acha-Sagredo, A.; Benedetti, L.; Temelkovski, D.; Choi, S.; Petrov, N.; Todd, K.; Wai, P.; et al. Immunogenomics of Colorectal Cancer Response to Checkpoint Blockade: Analysis of the KEYNOTE 177 Trial and Validation Cohorts. Gastroenterology 2021, 161, 1179–1193. [Google Scholar] [CrossRef]
  79. Endo, E.; Okayama, H.; Saito, K.; Nakajima, S.; Yamada, L.; Ujiie, D.; Kase, K.; Fujita, S.; Endo, H.; Sakamoto, W.; et al. A TGFbeta-Dependent Stromal Subset Underlies Immune Checkpoint Inhibitor Efficacy in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Colorectal Cancer. Mol. Cancer Res. 2020, 18, 1402–1413. [Google Scholar] [CrossRef]
  80. Chida, K.; Kawazoe, A.; Kawazu, M.; Suzuki, T.; Nakamura, Y.; Nakatsura, T.; Kuwata, T.; Ueno, T.; Kuboki, Y.; Kotani, D.; et al. A Low Tumor Mutational Burden and PTEN Mutations Are Predictors of a Negative Response to PD-1 Blockade in MSI-H/dMMR Gastrointestinal Tumors. Clin. Cancer Res. 2021, 27, 3714–3724. [Google Scholar] [CrossRef]
  81. Randrian, V.; Evrard, C.; Tougeron, D. Microsatellite Instability in Colorectal Cancers: Carcinogenesis, Neo-Antigens, Immuno-Resistance and Emerging Therapies. Cancers 2021, 13, 3063. [Google Scholar] [CrossRef] [PubMed]
  82. Melero, I.; Hervas-Stubbs, S.; Glennie, M.; Pardoll, D.M.; Chen, L. Immunostimulatory monoclonal antibodies for cancer therapy. Nat. Rev. Cancer 2007, 7, 95–106. [Google Scholar] [CrossRef]
  83. Abdullaev, S.; André, T.; Lei, M.; Lenz, H.-J.; Novotny, J.; Paulson, A.S.; Tejpar, S.; Yamazaki, K.; Ledeine, J.-M. A phase III study of nivolumab (NIVO), NIVO + ipilimumab (IPI), or chemotherapy (CT) for microsatellite instability-high (MSI-H)/mismatch repair-deficient (dMMR) metastatic colorectal cancer (mCRC): Checkmate 8HW. J. Clin. Oncol. 2020, 38, TPS266. [Google Scholar] [CrossRef]
  84. Kawakami, H.; Hironaka, S.; Esaki, T.; Chayama, K.; Tsuda, M.; Sugimoto, N.; Kadowaki, S.; Makiyama, A.; Machida, N.; Hirano, H.; et al. An Investigator-Initiated Phase 2 Study of Nivolumab Plus Low-Dose Ipilimumab as First-Line Therapy for Microsatellite Instability-High Advanced Gastric or Esophagogastric Junction Cancer (NO LIMIT, WJOG13320G/CA209-7W7). Cancers 2021, 13, 805. [Google Scholar] [CrossRef]
  85. Lima, C.M.S.P.R.; Yothers, G.; Jacobs, S.A.; Sanoff, H.K.; Cohen, D.J.; Guthrie, K.A.; Henry, N.L.; Ganz, P.A.; Kopetz, S.; Lucas, P.C.; et al. NRG-GI004/SWOG-S1610: Colorectal cancer metastatic dMMR immuno-therapy (COMMIT) study—A randomized phase III study of atezolizumab (atezo) monotherapy versus mFOLFOX6/bevacizumab/atezo in the first-line treatment of patients (pts) with deficient DNA mismatch repair (dMMR) or microsatellite instability high (MSI-H) metastatic colorectal cancer (mCRC). J. Clin. Oncol. 2022, 40, TPS232. [Google Scholar] [CrossRef]
  86. Guinney, J.; Dienstmann, R.; Wang, X.; de Reynies, A.; Schlicker, A.; Soneson, C.; Marisa, L.; Roepman, P.; Nyamundanda, G.; Angelino, P.; et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 2015, 21, 1350–1356. [Google Scholar] [CrossRef]
  87. Lenz, H.J.; Ou, F.S.; Venook, A.P.; Hochster, H.S.; Niedzwiecki, D.; Goldberg, R.M.; Mayer, R.J.; Bertagnolli, M.M.; Blanke, C.D.; Zemla, T.; et al. Impact of Consensus Molecular Subtype on Survival in Patients With Metastatic Colorectal Cancer: Results From CALGB/SWOG 80405 (Alliance). J. Clin. Oncol. 2019, 37, 1876–1885. [Google Scholar] [CrossRef]
  88. de Miranda, N.F.; van Dinther, M.; van den Akker, B.E.; van Wezel, T.; ten Dijke, P.; Morreau, H. Transforming Growth Factor beta Signaling in Colorectal Cancer Cells with Microsatellite Instability Despite Biallelic Mutations in TGFBR2. Gastroenterology 2015, 148, 1427–1437.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Xu, Y.; Pasche, B. TGF-beta signaling alterations and susceptibility to colorectal cancer. Hum. Mol. Genet. 2007, 16, R14–R20. [Google Scholar] [CrossRef] [Green Version]
  90. Itatani, Y.; Kawada, K.; Sakai, Y. Transforming Growth Factor-beta Signaling Pathway in Colorectal Cancer and Its Tumor Microenvironment. Int. J. Mol. Sci. 2019, 20, 5822. [Google Scholar] [CrossRef] [Green Version]
  91. Luo, J.; Chen, X.Q.; Li, P. The Role of TGF-beta and Its Receptors in Gastrointestinal Cancers. Transl. Oncol. 2019, 12, 475–484. [Google Scholar] [CrossRef] [PubMed]
  92. Colak, S.; Ten Dijke, P. Targeting TGF-beta Signaling in Cancer. Trends Cancer 2017, 3, 56–71. [Google Scholar] [CrossRef]
  93. Kim, T.W.; Lee, K.W.; Ahn, J.B.; Lee, J.; Ryu, J.; Oh, B.; Ock, C.-Y.; Hwang, S.; Hahm, K.B.; Kim, S.-J.; et al. Efficacy and safety of vactosertib and pembrolizumab combination in patients with previously treated microsatellite stable metastatic colorectal cancer. J. Clin. Oncol. 2021, 39, 3573. [Google Scholar] [CrossRef]
  94. Antoniotti, C.; Rossini, D.; Pietrantonio, F.; Catteau, A.; Salvatore, L.; Lonardi, S.; Boquet, I.; Tamberi, S.; Marmorino, F.; Moretto, R.; 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, 876–887. [Google Scholar] [CrossRef]
  95. Wallin, J.; Pishvaian, M.J.; Hernandez, G.; Yadav, M.; Jhunjhunwala, S.; Delamarre, L.; He, X.; Powderly, J.; Lieu, C.; Eckhardt, S.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. [Google Scholar] [CrossRef]
  96. Wallin, J.J.; Bendell, J.C.; Funke, R.; Sznol, M.; Korski, K.; Jones, S.; Hernandez, G.; Mier, J.; He, X.; Hodi, F.S.; et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat. Commun. 2016, 7, 12624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Innocenti, F.; Ou, F.S.; Qu, X.; Zemla, T.J.; Niedzwiecki, D.; Tam, R.; Mahajan, S.; Goldberg, R.M.; Bertagnolli, M.M.; Blanke, C.D.; et al. Mutational Analysis of Patients With Colorectal Cancer in CALGB/SWOG 80405 Identifies New Roles of Microsatellite Instability and Tumor Mutational Burden for Patient Outcome. J. Clin. Oncol. 2019, 37, 1217–1227. [Google Scholar] [CrossRef]
  98. Pogue-Geile, K.; Yothers, G.; Taniyama, Y.; Tanaka, N.; Gavin, P.; Colangelo, L.; Blackmon, N.; Lipchik, C.; Kim, S.R.; Sharif, S.; et al. Defective mismatch repair and benefit from bevacizumab for colon cancer: Findings from NSABP C-08. J. Natl. Cancer Inst. 2013, 105, 989–992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Hoff, S.; Grünewald, S.; Röse, L.; Zopf, D. Immunomodulation by regorafenib alone and in combination with anti PD1 antibody on murine models of colorectal cancer. Ann. Oncol. 2017, 28, v423. [Google Scholar] [CrossRef]
  100. Tada, Y.; Togashi, Y.; Kotani, D.; Kuwata, T.; Sato, E.; Kawazoe, A.; Doi, T.; Wada, H.; Nishikawa, H.; Shitara, K. Targeting VEGFR2 with Ramucirumab strongly impacts effector/ activated regulatory T cells and CD8(+) T cells in the tumor microenvironment. J. Immunother. Cancer 2018, 6, 106. [Google Scholar] [CrossRef]
  101. Fukuoka, S.; Hara, H.; Takahashi, N.; Kojima, T.; Kawazoe, A.; Asayama, M.; Yoshii, T.; Kotani, D.; Tamura, H.; Mikamoto, Y.; et al. Regorafenib Plus Nivolumab in Patients With Advanced Gastric or Colorectal Cancer: An Open-Label, Dose-Escalation, and Dose-Expansion Phase Ib Trial (REGONIVO, EPOC1603). J. Clin. Oncol. 2020, 38, 2053–2061. [Google Scholar] [CrossRef]
  102. Kawazoe, A.; Fukuoka, S.; Nakamura, Y.; Kuboki, Y.; Wakabayashi, M.; Nomura, S.; Mikamoto, Y.; Shima, H.; Fujishiro, N.; Higuchi, T.; et al. Lenvatinib plus pembrolizumab in patients with advanced gastric cancer in the first-line or second-line setting (EPOC1706): An open-label, single-arm, phase 2 trial. Lancet Oncol. 2020, 21, 1057–1065. [Google Scholar] [CrossRef] [PubMed]
  103. Kato, Y.; Tabata, K.; Kimura, T.; Yachie-Kinoshita, A.; Ozawa, Y.; Yamada, K.; Ito, J.; Tachino, S.; Hori, Y.; Matsuki, M.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Huang, C.T.; Workman, C.J.; Flies, D.; Pan, X.; Marson, A.L.; Zhou, G.; Hipkiss, E.L.; Ravi, S.; Kowalski, J.; Levitsky, H.I.; et al. Role of LAG-3 in regulatory T cells. Immunity 2004, 21, 503–513. [Google Scholar] [CrossRef] [Green Version]
  105. Baixeras, E.; Huard, B.; Miossec, C.; Jitsukawa, S.; Martin, M.; Hercend, T.; Auffray, C.; Triebel, F.; Piatier-Tonneau, D. Characterization of the lymphocyte activation gene 3-encoded protein. A new ligand for human leukocyte antigen class II antigens. J. Exp. Med. 1992, 176, 327–337. [Google Scholar] [CrossRef] [PubMed]
  106. Deng, W.W.; Mao, L.; Yu, G.T.; Bu, L.L.; Ma, S.R.; Liu, B.; Gutkind, J.S.; Kulkarni, A.B.; Zhang, W.F.; Sun, Z.J. LAG-3 confers poor prognosis and its blockade reshapes antitumor response in head and neck squamous cell carcinoma. Oncoimmunology 2016, 5, e1239005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Huard, B.; Prigent, P.; Tournier, M.; Bruniquel, D.; Triebel, F. CD4/major histocompatibility complex class II interaction analyzed with CD4- and lymphocyte activation gene-3 (LAG-3)-Ig fusion proteins. Eur. J. Immunol. 1995, 25, 2718–2721. [Google Scholar] [CrossRef]
  108. Garralda, E.; Sukari, A.; Lakhani, N.J.; Patnaik, A.; Lou, Y.; Im, S.-A.; Golan, T.; Geva, R.; Wermke, M.; Miguel, M.D.; et al. A phase 1 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. [Google Scholar] [CrossRef]
  109. Hollebecque, A.; Chung, H.C.; de Miguel, M.J.; Italiano, A.; Machiels, J.P.; Lin, C.C.; Dhani, N.C.; Peeters, M.; Moreno, V.; Su, W.C.; et al. Safety and Antitumor Activity of alpha-PD-L1 Antibody as Monotherapy or in Combination with alpha-TIM-3 Antibody in Patients with Microsatellite Instability-High/Mismatch Repair-Deficient Tumors. Clin. Cancer Res. 2021, 27, 6393–6404. [Google Scholar] [CrossRef]
  110. Bever, K.M.; Wang, H.; Durham, J.N.; Petrie, S.; Hoare, J.; Wilt, C.; Sharfman, W.H.; Azad, N.S.; Laheru, D.A.; Anders, R.A.; et al. Phase II study of nivolumab and relatlimab in advanced mismatch repair deficient (dMMR) cancers resistant to prior PD-(L)1 inhibition. J. Clin. Oncol. 2020, 38, TPS839. [Google Scholar] [CrossRef]
  111. Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Castillo Gutierrez, E.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef] [PubMed]
  112. Zhou, C.; He, Y.; Ren, S.; Li, W.; Zhu, J.; Yu, J.; Wang, L.; Xiong, A.; Xu, N.; Mao, C.; et al. Phase Ia/Ib dose-escalation study of IBI110 (anti-LAG-3 mAb) as a single agent and in combination with sintilimab (anti-PD-1 mAb) in patients (pts) with advanced solid tumors. J. Clin. Oncol. 2021, 39, 2589. [Google Scholar] [CrossRef]
  113. Schoffski, P.; Tan, D.S.W.; Martin, M.; Ochoa-de-Olza, M.; Sarantopoulos, J.; Carvajal, R.D.; Kyi, C.; Esaki, T.; Prawira, A.; Akerley, W.; et al. Phase I/II study of the LAG-3 inhibitor ieramilimab (LAG525) +/− anti-PD-1 spartalizumab (PDR001) in patients with advanced malignancies. J. Immunother. Cancer 2022, 10, e003776. [Google Scholar] [CrossRef] [PubMed]
  114. Germano, G.; Lamba, S.; Rospo, G.; Barault, L.; Magri, A.; Maione, F.; Russo, M.; Crisafulli, G.; Bartolini, A.; Lerda, G.; et al. Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth. Nature 2017, 552, 116–120. [Google Scholar] [CrossRef]
  115. Kanani, A.; Veen, T.; Soreide, K. Neoadjuvant immunotherapy in primary and metastatic colorectal cancer. Br. J. Surg. 2021, 108, 1417–1425. [Google Scholar] [CrossRef] [PubMed]
  116. Chalabi, M.; Fanchi, L.F.; Dijkstra, K.K.; Van den Berg, J.G.; Aalbers, A.G.; Sikorska, K.; Lopez-Yurda, M.; Grootscholten, C.; Beets, G.L.; Snaebjornsson, P.; et al. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat. Med. 2020, 26, 566–576. [Google Scholar] [CrossRef] [PubMed]
  117. Chalabi, M.; Verschoor, Y.L.; van den Berg, J.; Sikorska, K.; Beets, G.; Lent, A.V.; Grootscholten, M.C.; Aalbers, A.; Buller, N.; Marsman, H.; et al. LBA7 Neoadjuvant immune checkpoint inhibition in locally advanced MMR-deficient colon cancer: The NICHE-2 study. Ann. Oncol. 2022, 33, S1389. [Google Scholar] [CrossRef]
  118. Cercek, A.; Lumish, M.; Sinopoli, J.; Weiss, J.; Shia, J.; Lamendola-Essel, M.; El Dika, I.H.; Segal, N.; Shcherba, M.; Sugarman, R.; et al. PD-1 Blockade in Mismatch Repair-Deficient, Locally Advanced Rectal Cancer. N. Engl. J. Med. 2022, 386, 2363–2376. [Google Scholar] [CrossRef]
  119. André, T.; Tougeron, D.; Piessen, G.; Fouchardière, C.d.l.; Louvet, C.; Adenis, A.; Jary, M.; Tournigand, C.; Aparicio, T.; Desrame, J.; et al. Neoadjuvant Nivolumab Plus Ipilimumab and Adjuvant Nivolumab in Localized Deficient Mismatch Repair/Microsatellite Instability-High Gastric or Esophagogastric Junction Adenocarcinoma: The GERCOR NEONIPIGA Phase II Study. J. Clin. Oncol. 2022, 41, 255–265. [Google Scholar] [CrossRef]
  120. Al-Batran, S.-E.; Lorenzen, S.; Thuss-Patience, P.C.; Homann, N.; Schenk, M.; Lindig, U.; Heuer, V.; Kretzschmar, A.; Goekkurt, E.; Haag, G.M.; et al. Surgical and pathological outcome, and pathological regression, in patients receiving perioperative atezolizumab in combination with FLOT chemotherapy versus FLOT alone for resectable esophagogastric adenocarcinoma: Interim results from DANTE, a randomized, multicenter, phase IIb trial of the FLOT-AIO German Gastric Cancer Group and Swiss SAKK. J. Clin. Oncol. 2022, 40, 4003. [Google Scholar] [CrossRef]
  121. Ludford, K.; Ho, W.J.; Thomas, J.V.; Raghav, K.P.S.; Murphy, M.B.; Fleming, N.D.; Lee, M.S.; Smaglo, B.G.; You, Y.N.; Tillman, M.M.; et al. Neoadjuvant Pembrolizumab in Localized Microsatellite Instability High/Deficient Mismatch Repair Solid Tumors. J. Clin. Oncol. 2023, JCO2201351. [Google Scholar] [CrossRef] [PubMed]
  122. Huang, A.C.; Orlowski, R.J.; Xu, X.; Mick, R.; George, S.M.; Yan, P.K.; Manne, S.; Kraya, A.A.; Wubbenhorst, B.; Dorfman, L.; et al. A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat. Med. 2019, 25, 454–461. [Google Scholar] [CrossRef] [PubMed]
  123. Blank, C.U.; Rozeman, E.A.; Fanchi, L.F.; Sikorska, K.; van de Wiel, B.; Kvistborg, P.; Krijgsman, O.; van den Braber, M.; Philips, D.; Broeks, A.; et al. Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma. Nat. Med. 2018, 24, 1655–1661. [Google Scholar] [CrossRef] [PubMed]
  124. Amaria, R.N.; Reddy, S.M.; Tawbi, H.A.; Davies, M.A.; Ross, M.I.; Glitza, I.C.; Cormier, J.N.; Lewis, C.; Hwu, W.J.; Hanna, E.; et al. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat. Med. 2018, 24, 1649–1654. [Google Scholar] [CrossRef] [PubMed]
  125. Verschoor, Y.L.; Berg, J.v.d.; Beets, G.; Sikorska, K.; Aalbers, A.; Lent, A.v.; Grootscholten, C.; Huibregtse, I.; Marsman, H.; Oosterling, S.; et al. Neoadjuvant nivolumab, ipilimumab, and celecoxib in MMR-proficient and MMR-deficient colon cancers: Final clinical analysis of the NICHE study. J. Clin. Oncol. 2022, 40, 3511. [Google Scholar] [CrossRef]
  126. Ludford, K.; Raghav, K.P.S.; Murphy, M.A.B.; Fleming, N.D.; Nelson, D.A.; Lee, M.S.; Smaglo, B.G.; You, Y.N.; Tillman, M.M.; Kamiya-Matsuoka, C.; et al. Safety and efficacy of neoadjuvant pembrolizumab in mismatch repair deficient localized/locally advanced solid tumors. J. Clin. Oncol. 2021, 39, 2520. [Google Scholar] [CrossRef]
  127. Bertagnolli, M.M.; Redston, M.; Compton, C.C.; Niedzwiecki, D.; Mayer, R.J.; Goldberg, R.M.; Colacchio, T.A.; Saltz, L.B.; Warren, R.S. Microsatellite instability and loss of heterozygosity at chromosomal location 18q: Prospective evaluation of biomarkers for stages II and III colon cancer—A study of CALGB 9581 and 89803. J. Clin. Oncol. 2011, 29, 3153–3162. [Google Scholar] [CrossRef] [Green Version]
  128. Bando, H.; Tsukada, Y.; Inamori, K.; Togashi, Y.; Koyama, S.; Kotani, D.; Fukuoka, S.; Yuki, S.; Komatsu, Y.; Homma, S.; 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, 1136–1146. [Google Scholar] [CrossRef]
  129. Pietrantonio, F.; Miceli, R.; Raimondi, A.; Kim, Y.W.; Kang, W.K.; Langley, R.E.; Choi, Y.Y.; Kim, K.M.; Nankivell, M.G.; Morano, F.; et al. Individual Patient Data Meta-Analysis of the Value of Microsatellite Instability As a Biomarker in Gastric Cancer. J. Clin. Oncol. 2019, 37, 3392–3400. [Google Scholar] [CrossRef]
  130. Sun, W.; Saeed, A.; Al-Rajabi, R.M.d.T.; Kasi, A.; Veeramachaneni, N.K.; Al-Kasspooles, M.M.; Baranda, J.C.; Phadnis, M.; Godwin, A.K.; Olyaee, M.; et al. A phase II study of perioperative mFOLFOX chemotherapy plus pembrolizumab combination in patients with potentially resectable adenocarcinoma of the esophageal, gastroesophageal junction (GEJ), and stomach. J. Clin. Oncol. 2022, 40, 329. [Google Scholar] [CrossRef]
  131. Janjigian, Y.Y.; Cutsem, E.V.; Muro, K.; Wainberg, Z.A.; Al-Batran, S.-E.; Hyung, W.J.; Molena, D.; Evans, B.; Ruscica, D.; Robbins, S.H.; et al. MATTERHORN: Efficacy and safety of neoadjuvant-adjuvant durvalumab and FLOT chemotherapy in resectable gastric and gastroesophageal junction cancer—A randomized, double-blind, placebo-controlled, phase 3 study. J. Clin. Oncol. 2021, 39, TPS4151. [Google Scholar] [CrossRef]
  132. Bang, Y.J.; Van Cutsem, E.; Fuchs, C.S.; Ohtsu, A.; Tabernero, J.; Ilson, D.H.; Hyung, W.J.; Strong, V.E.; Goetze, T.O.; Yoshikawa, T.; et al. KEYNOTE-585: Phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Future Oncol. 2019, 15, 943–952. [Google Scholar] [CrossRef] [PubMed]
  133. Fancello, L.; Gandini, S.; Pelicci, P.G.; Mazzarella, L. Tumor mutational burden quantification from targeted gene panels: Major advancements and challenges. J. Immunother. Cancer 2019, 7, 183. [Google Scholar] [CrossRef] [Green Version]
  134. Stenzinger, A.; Allen, J.D.; Maas, J.; Stewart, M.D.; Merino, D.M.; Wempe, M.M.; Dietel, M. Tumor mutational burden standardization initiatives: Recommendations for consistent tumor mutational burden assessment in clinical samples to guide immunotherapy treatment decisions. Genes Chromosomes Cancer 2019, 58, 578–588. [Google Scholar] [CrossRef] [Green Version]
  135. Cristescu, R.; Mogg, R.; Ayers, M.; Albright, A.; Murphy, E.; Yearley, J.; Sher, X.; Liu, X.Q.; Lu, H.; Nebozhyn, M.; et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018, 362, eaar3593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Marabelle, A.; Fakih, M.; Lopez, J.; Shah, M.; Shapira-Frommer, R.; Nakagawa, K.; Chung, H.C.; Kindler, H.L.; Lopez-Martin, J.A.; Miller, W.H., Jr.; 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, 1353–1365. [Google Scholar] [CrossRef] [PubMed]
  137. Vanderwalde, A.; Spetzler, D.; Xiao, N.; Gatalica, Z.; Marshall, J. Microsatellite instability status determined by next-generation sequencing and compared with PD-L1 and tumor mutational burden in 11,348 patients. Cancer Med. 2018, 7, 746–756. [Google Scholar] [CrossRef] [Green Version]
  138. Schrock, A.B.; Ouyang, C.; Sandhu, J.; Sokol, E.; Jin, D.; Ross, J.S.; Miller, V.A.; Lim, D.; Amanam, I.; Chao, 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. [Google Scholar] [CrossRef] [PubMed]
  139. Sha, D.; Jin, Z.; Budczies, J.; Kluck, K.; Stenzinger, A.; Sinicrope, F.A. Tumor Mutational Burden as a Predictive Biomarker in Solid Tumors. Cancer Discov. 2020, 10, 1808–1825. [Google Scholar] [CrossRef]
  140. McGranahan, N.; Swanton, C. Neoantigen quality, not quantity. Sci. Transl. Med. 2019, 11, eaax7918. [Google Scholar] [CrossRef]
  141. Xiao, Y.; Freeman, G.J. The microsatellite instable subset of colorectal cancer is a particularly good candidate for checkpoint blockade immunotherapy. Cancer Discov. 2015, 5, 16–18. [Google Scholar] [CrossRef] [Green Version]
  142. Shimozaki, K.; Hayashi, H.; Tanishima, S.; Horie, S.; Chida, A.; Tsugaru, K.; Togasaki, K.; Kawasaki, K.; Aimono, E.; Hirata, K.; et al. Concordance analysis of microsatellite instability status between polymerase chain reaction based testing and next generation sequencing for solid tumors. Sci. Rep. 2021, 11, 20003. [Google Scholar] [CrossRef]
  143. Middha, S.; Zhang, L.; Nafa, K.; Jayakumaran, G.; Wong, D.; Kim, H.R.; Sadowska, J.; Berger, M.F.; Delair, D.F.; Shia, J.; et al. Reliable Pan-Cancer Microsatellite Instability Assessment by Using Targeted Next-Generation Sequencing Data. JCO Precis Oncol. 2017, 2017, PO.17.00084. [Google Scholar] [CrossRef] [PubMed]
  144. Salipante, S.J.; Scroggins, S.M.; Hampel, H.L.; Turner, E.H.; Pritchard, C.C. Microsatellite instability detection by next generation sequencing. Clin. Chem. 2014, 60, 1192–1199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Nakamura, Y.; Okamoto, W.; Denda, T.; Nishina, T.; Komatsu, Y.; Yuki, S.; Yasui, H.; Esaki, T.; Sunakawa, Y.; Ueno, M.; et al. Clinical Validity of Plasma-Based Genotyping for Microsatellite Instability Assessment in Advanced GI Cancers: SCRUM-Japan GOZILA Substudy. JCO Precis Oncol. 2022, 6, e2100383. [Google Scholar] [CrossRef] [PubMed]
  146. Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Borresen-Dale, A.L.; et al. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Howitt, B.E.; Shukla, S.A.; Sholl, L.M.; Ritterhouse, L.L.; Watkins, J.C.; Rodig, S.; Stover, E.; Strickland, K.C.; D’Andrea, A.D.; Wu, C.J.; et al. Association of Polymerase e-Mutated and Microsatellite-Instable Endometrial Cancers With Neoantigen Load, Number of Tumor-Infiltrating Lymphocytes, and Expression of PD-1 and PD-L1. JAMA Oncol. 2015, 1, 1319–1323. [Google Scholar] [CrossRef] [Green Version]
  148. Budczies, J.; Kluck, K.; Beck, S.; Ourailidis, I.; Allgauer, M.; Menzel, M.; Kazdal, D.; Perkhofer, L.; Kleger, A.; Schirmacher, P.; et al. Homologous recombination deficiency is inversely correlated with microsatellite instability and identifies immunologically cold tumors in most cancer types. J. Pathol. Clin. Res. 2022, 8, 371–382. [Google Scholar] [CrossRef] [PubMed]
  149. Lengauer, C.; Kinzler, K.W.; Vogelstein, B. Genetic instabilities in human cancers. Nature 1998, 396, 643–649. [Google Scholar] [CrossRef] [PubMed]
  150. Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 2012, 487, 330–337. [Google Scholar] [CrossRef] [Green Version]
  151. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. MSI-H/dMMR cancer and immunotherapy. Adapted from “Tumor microenvironment,” by BioRender.com (2022). Retrieved from https://app.biorender.com/biorender-templates (accessed on 28 October 2022). Created with BioRender.com. Used under BioRender’s academic license terms. (a) The deficiency in mismatch repair results in the impossibility of repairing DNA mismatches in microsatellites, determining the accumulation of mutations in different genomic codons. (b) Neoantigens, or abnormal proteins resulting from cancer gene mutations, are taken up by antigen-presenting cells (APCs). APCs stimulate cytotoxic T lymphocytes, which subsequently go toward the tumor site and destroy the cancer cells. Cancer produces immune checkpoint molecules throughout this process, including programmed death ligand 1 (PD-L1), to evade T-cell attack. As a result, T cells can no longer target the tumor cell. Anti-PD-1 antibodies prevent PD-1/PD-L1 binding, allowing T cells to be activated once more and fight the tumor. (c,d) In the tumor microenvironment of microsatellite instability-high and/or deficient mismatch repair cancer, hypermutated tumor cells produce several neoantigens, stimulating T-cell activation and tumor infiltration by various immune cells.
Figure 1. MSI-H/dMMR cancer and immunotherapy. Adapted from “Tumor microenvironment,” by BioRender.com (2022). Retrieved from https://app.biorender.com/biorender-templates (accessed on 28 October 2022). Created with BioRender.com. Used under BioRender’s academic license terms. (a) The deficiency in mismatch repair results in the impossibility of repairing DNA mismatches in microsatellites, determining the accumulation of mutations in different genomic codons. (b) Neoantigens, or abnormal proteins resulting from cancer gene mutations, are taken up by antigen-presenting cells (APCs). APCs stimulate cytotoxic T lymphocytes, which subsequently go toward the tumor site and destroy the cancer cells. Cancer produces immune checkpoint molecules throughout this process, including programmed death ligand 1 (PD-L1), to evade T-cell attack. As a result, T cells can no longer target the tumor cell. Anti-PD-1 antibodies prevent PD-1/PD-L1 binding, allowing T cells to be activated once more and fight the tumor. (c,d) In the tumor microenvironment of microsatellite instability-high and/or deficient mismatch repair cancer, hypermutated tumor cells produce several neoantigens, stimulating T-cell activation and tumor infiltration by various immune cells.
Cells 12 01049 g001
Figure 2. Timeline of events for immunotherapy MSI-H/dMMR application and clinical trials. Abbreviations: ipi, ipilimumab; mCRC, metastatic colorectal cancer; MSI-H, microsatellite instability-high; nivo, nivolumab; PD-1, programmed death-1; pembro, pembrolizumab.
Figure 2. Timeline of events for immunotherapy MSI-H/dMMR application and clinical trials. Abbreviations: ipi, ipilimumab; mCRC, metastatic colorectal cancer; MSI-H, microsatellite instability-high; nivo, nivolumab; PD-1, programmed death-1; pembro, pembrolizumab.
Cells 12 01049 g002
Figure 3. Mechanisms of resistance for immunotherapy in MSI-H/dMMR cancers. Adapted from “Drug Targeting in Cytokine Receptor Pathways,” by BioRender.com (2022). Retrieved from https://app.biorender.com/biorender-templates (accessed on 28 October 2022). Created with BioRender.com. Used under BioRender’s academic license terms. (a) JAK-STAT pathway alterations compromise the immune response and negatively regulate PD-L1 expression in tumor cells. (b) Mutations in beta 2 microglobulin, a component of MHC class I, could impair the opportunity to present neoantigens to immune cells. (c) The expression of inhibitory immune checkpoints (i.e., CTLA-4, LAG-3, and TIM-3) may induce MSI-H/dMMR tumors to lose their ability to be immunogenic. (d) Immune-evasive oncogenic signaling such as TGF-beta also leads to Treg differentiation and the suppression of NK cell function in the tumor microenvironment. (e) Tumor-associated macrophages promote angiogenesis. Abbreviations: B2M, beta-2 microglobulin; MHC, major histocompatibility complex; NK cell, natural killer cell; TAM, tumor-associated macrophage; TCR, T-cell receptor; Treg, regulatory T cell.
Figure 3. Mechanisms of resistance for immunotherapy in MSI-H/dMMR cancers. Adapted from “Drug Targeting in Cytokine Receptor Pathways,” by BioRender.com (2022). Retrieved from https://app.biorender.com/biorender-templates (accessed on 28 October 2022). Created with BioRender.com. Used under BioRender’s academic license terms. (a) JAK-STAT pathway alterations compromise the immune response and negatively regulate PD-L1 expression in tumor cells. (b) Mutations in beta 2 microglobulin, a component of MHC class I, could impair the opportunity to present neoantigens to immune cells. (c) The expression of inhibitory immune checkpoints (i.e., CTLA-4, LAG-3, and TIM-3) may induce MSI-H/dMMR tumors to lose their ability to be immunogenic. (d) Immune-evasive oncogenic signaling such as TGF-beta also leads to Treg differentiation and the suppression of NK cell function in the tumor microenvironment. (e) Tumor-associated macrophages promote angiogenesis. Abbreviations: B2M, beta-2 microglobulin; MHC, major histocompatibility complex; NK cell, natural killer cell; TAM, tumor-associated macrophage; TCR, T-cell receptor; Treg, regulatory T cell.
Cells 12 01049 g003
Table 2. Clinical trials for MSI-H/dMMR solid tumors upon which the FDA based approval for pembrolizumab.
Table 2. Clinical trials for MSI-H/dMMR solid tumors upon which the FDA based approval for pembrolizumab.
StudyPatient PopulationNo. of Patients with MSI-H/dMMR Solid TumorsMSI-H/dMMR TestingDosePrior Therapy
KEYNOTE-016 [6] NCT01867511CRC and other tumors28 CRC
30 non-CRC
PCR or IHC
prospectively evaluated
10 mg/kg
every 2 weeks
CRC: ≥2 prior regimens
Non-CRC: ≥1 prior regimen
KEYNOTE-164 [21]
NCT02460198
CRC61PCR or IHC
prospectively evaluated
200 mg every 3 weeksPrior fluoropyrimidine, oxaliplatin, and irinotecan +/− anti-VEGF/EGFR mAb
KEYNOTE-012 [29]
NCT01848834
PD-L1–positive gastric, bladder, or triple-negative breast cancer6PCR
retrospectively identified
10 mg/kg every 2 weeks≥1 prior regimen
KEYNOTE-028 [28]
NCT02054806
PD-L1–positive esophageal, biliary, breast, endometrial, or CRC5PCR
retrospectively identified
10 mg/kg every 2 weeks≥1 prior regimen
KEYNOTE-158 [27]
NCT02628067
MSI-H/dMMR non-CRC19PCR or IHC
prospectively evaluated
(retrospectively identified in specific rare tumors)
200 mg every 3 weeks≥1 prior regimen
Abbreviations: CRC, colorectal cancer; dMMR, deficient mismatch repair; EGFR epidermal growth factor receptor; IHC, immunohistochemistry; mAb, monoclonal antibody; MSI-H, microsatellite instability-high; PCR, polymerase chain reaction; PD-L1, programmed death ligand 1; VEGF, vascular endothelial growth factor.
Table 3. Clinical trials for MSI-H/dMMR metastatic gastric cancer/gastroesophageal junction cancer.
Table 3. Clinical trials for MSI-H/dMMR metastatic gastric cancer/gastroesophageal junction cancer.
StudyKEYNOTE-059 [42]KEYNOTE-061 [42]KEYNOTE-062 [42]CheckMate-649 [53]
Treatment line≥3rd line2nd line1st line1st line
PhaseIIIIIIIIIII
No. of MSI-H/dMMR patients7151214
CPS ≥ 1
17
CPS ≥ 1
19
CPS ≥ 1
231121
Primary endpoint OS and PFS in PD-L1CPS ≥ 1OS and PFS in PD-L1CPS ≥ 1PS and PFS in PD-L1 CPS ≥ 5
RegimenPembroPembroChemoPembroChemo + PembroChemoChemo + NivoNivo1 + Ipi3Chemo
ORR57.1%46.7%16.7%57.1%64.7%36.8%55%70%39%
DoRNRNRNR21.2NR7.0
Median PFSNR17.83.511.2NR6.6
HR for PFS
(95% CI)
0.72
(0.31–1.68)
0.45
(0.18–1.11)
Ref
12-month PFS rate43%56%28%
Median OSNRNR8.1NRNR8.538.7NR12.3
HR for OS
(95% CI)
0.42
(0.13–1.31)
Ref0.21
(0.06–0.83)
0.37
(0.14–0.97)
Ref0.38
(0.17–0.84)
0.28
(0.08–0.92)
Ref
12-month OS rate71%73%25%79%71%47%
Abbreviations: CI, confidence interval; CPS, combined positive score; DoR, duration of response; dMMR, deficient mismatch repair; HR, hazard ratio; MSI-H, microsatellite instability-high; NR, not reached; ORR, objective response rate; OS, overall survival; PFS, progression-free survival; Ref, reference.
Table 4. Neoadjuvant immunotherapy in MSI-H/dMMR gastrointestinal cancer.
Table 4. Neoadjuvant immunotherapy in MSI-H/dMMR gastrointestinal cancer.
StudyNICHE-2 [116,117]Cercek et al. [118]GERCOR NEONIPIGA [119]DANTE [120]Kaysia et al. [121]
NCT numberNCT03026140NCT04165772NCT04006262NCT03421288NCT04082572
Primary tumorColorectalRectalGastric or gastroesophageal junction cancerGastric or gastroesophageal cancerSolid tumors
PhasePhase IIPhase IIPhase IIRandomized phase IIPhase II
Number of patients with MSI-H/dMMR tumor11212 (ongoing)328
(8 patients with FLOT plus atezolizumab arm)
35
Clinical stageI–IIIII/IIIcT2–4, any NcT2–4, any NUnresectable or high-risk resectable
RegimenNivolumab plus ipilimumabDostarlimabNivolumab plus ipilimumabFLOT plus atezolizumabPembrolizumab
Primary endpoints
  • ✓ Safety
  • ✓ 3-year DFS
Clinical CR at 12 months follow-up
Pathological CR
Pathological CR rate
PFS
DFS
Safety
Pathological CR rate
Clinical CR
(95% CI)
Not reported100% (74–100)NANA30%
Pathological CR67%NA58.6%63%65%
(79% in CRC)
MPR95%NA72.4%75%NA
Adverse events of grade ≥313%0%25%Not reported6%
Abbreviations: CI, confidence interval; CR, complete response; DFS, disease-free survival; dMMR, deficient mismatch repair; MPR major pathological response; MSI-H, microsatellite instability-high; NA, not assessed; PFS, progression-free survival.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shimozaki, K.; Nakayama, I.; Hirota, T.; Yamaguchi, K. Current Strategy to Treat Immunogenic Gastrointestinal Cancers: Perspectives for a New Era. Cells 2023, 12, 1049. https://doi.org/10.3390/cells12071049

AMA Style

Shimozaki K, Nakayama I, Hirota T, Yamaguchi K. Current Strategy to Treat Immunogenic Gastrointestinal Cancers: Perspectives for a New Era. Cells. 2023; 12(7):1049. https://doi.org/10.3390/cells12071049

Chicago/Turabian Style

Shimozaki, Keitaro, Izuma Nakayama, Toru Hirota, and Kensei Yamaguchi. 2023. "Current Strategy to Treat Immunogenic Gastrointestinal Cancers: Perspectives for a New Era" Cells 12, no. 7: 1049. https://doi.org/10.3390/cells12071049

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop