Skip to main content

Oncological characteristics, treatments and prognostic outcomes in MMR-deficient colorectal cancer

Abstract

Colorectal cancer (CRC) ranks as the third most prevalent cancer globally. It’s recognized that the molecular subtype of CRC, characterized by mismatch repair deficiency (dMMR) or microsatellite instability-high (MSI-H), plays a critical role in determining appropriate treatment strategies. This review examines the current molecular classifications, focusing on dMMR/MSI-H CRC and its subtypes: Lynch syndrome (LS), Lynch-like syndrome (LLS), and sporadic cases. Despite advances in understanding of these genetic backgrounds, clinical trials have not conclusively differentiated the efficacy of immune checkpoint inhibitors among these subgroups. Therefore, while this review details the molecular characteristics and their general implications for treatment and prognosis, it also highlights the limitations and the need for more refined clinical studies to ascertain tailored therapeutic strategies for each subtype. Furthermore, this review summarizes completed and ongoing clinical studies, emphasizing the importance of developing treatments aligned more closely with molecular profiles. By discussing these aspects, the review seeks to provide a comprehensive analysis of oncological characteristics, presenting a detailed understanding of their implications for treatment and prognosis in dMMR/MSI-H CRC.

Introduction

Colorectal cancer (CRC) currently accounts for 9.7% of cancer incidences and 9.4% of cancer-related deaths worldwide [1, 2]. In China, the average 5-year survival rate for CRC is 56.9% [3], ranking second in global cancer-related mortality [4]. Recently, the latest statistical report on the incidence and mortality of CRC in the United States indicated that while the overall incidence and mortality rates of CRC are decreasing, the disease burden is rapidly shifting towards younger patients and a growing trend of more individuals progressing to advanced stages [5]. Neoadjuvant therapy, surgery, adjuvant treatment, radiation therapy, and chemotherapy are the main treatment methods for CRC patients, but they may lead to changes in the patient’s physical appearance, varying degrees of impairment in basic physical functions, and social functional impairments [6, 7].

Microsatellites, also known as short tandem repeats, are DNA sequences in the genome consisting of a small number of nucleotide repeats. In cases where there is a malfunction in the DNA mismatch repair (MMR) system, errors in the replication of microsatellites accumulate, leading to changes in the length or composition of the sequences, a condition known as microsatellite instability (MSI). This instability gives rise to a high mutation phenotype [8]. Deficient MMR (dMMR) in tumors is usually caused by pathogenic mutations in MMR genes such as mutL homolog 1 (MLH1), muts homolog 2 (MSH2), muts homolog 6 (MSH6), postmeiotic segregation increased 2 (PMS2), or in the epithelial cell adhesion molecule (EPCAM) gene. It can also be caused by high methylation of the MLH1 promoter region, leading to loss of MLH1 expression [9]. MSI is classified into three categories: high instability (MSI-H), low instability (MSI-L), and stable (MSS) [10]. The National Comprehensive Cancer Network (NCCN) recommends the use of the 2B3D NCI panel and 5 mononucleotide marker panel primarily based on the Promega Panel for MSI testing [11, 12]. In stage II CRC, approximately 20% of patients exhibit dMMR/MSI-H, while in stage III, this proportion is around 12%. The prevalence is lowest in stage IV, at approximately 4% [13]. The prevalence of dMMR/MSI-H CRC varies by ethnicity [14, 15]. Studies has reported higher rates of MSI-H CRC among Europeans (5-24%) and Caucasian Americans of European descent (8-20%), as well as African Americans (12-45%) and Egyptians (37%) [8, 16,17,18]. In contrast, MSI-H CRC prevalence is relatively lower in Asian countries, with rates of 4.5-15.0% in China and 3.8 − 20.0% in Japan [19, 20].

The diagnostic and treatment approaches for advanced CRC patients with pMMR/MSS and dMMR/MSI-H subtypes are completely different, leading to distinct therapeutic regimens and varying treatment outcomes. dMMR/MSI-H CRC commonly presents in three clinical types: Lynch syndrome (LS), Lynch-like syndrome (LLS) and sporadic CRC (Fig. 1A). This article aims to delineate the molecular characteristics and their general implications for treatment, and prognosis of these three types.

Fig. 1
figure 1

(A) LLS tumors exhibit MSI or MMR expression deficiency (without BRAF mutation and MLH1 methylation), with no germline MMR mutations. Patients with LLS can be categorized into those with a family history and those without a cancer history. LS tumors are caused by germline mutations in MMR genes, followed by a second hit on the wild-type copy, which may occur through LOH, methylation, or point mutations, often accompanied by KRAS mutation and a family history of CRC. Sporadic CRC typically lack a family history and exhibit CpG island methylator phenotype (CIMP) characteristics, leading to methylation of numerous gene promoters. When the MLH1 promoter is methylated, MMR activity is disrupted, leading to MSI, with BRAF mutations observed in the majority of sporadic colorectal tumors, but not in tumors of LS patients. (B) PD-L1 is a ligand of PD-1 and is associated with immune suppression. The expression of PD-L1 by tumor cells can lead to T cell evasion, enabling immune escape. Anti-CTLA-4 antibodies activate anti-tumor immunity, while LAG-3 inhibits activated T cells, terminating signal transduction after binding to class II MHC molecules and class I MHC molecules, leading to a decline in immune response

Molecular biological characteristics of dMMR/MSI-H

Molecular mechanism and genetic background of 3 types of dMMR/MSI-H CRC

Hereditary nonpolyposis colorectal cancer (HNPCC), also known as LS, is a common autosomal dominant hereditary tumor syndrome, accounting for approximately 2–4% of CRCs [21]. The Amsterdam I and II criteria, the Bethesda guidelines, and the revised Bethesda guidelines are all screening criteria specifically for LS. They are used to identify CRC patients who require dMMR or MSI testing.LS is mainly caused by germline mutations in the MLH1, MSH2, MSH6, or PMS2 genes, or by deletion of the EPCAM gene resulting in non-expression of MSH2 [22]. The “second hit” associated with these mutations leads to the inactivation of the other allele, resulting in tumor development. Tumors often present with mismatch repair deficiencies [23, 24].

Lynch-like syndrome (LLS) refers to patients who present with features resembling LS, such as DNA mismatch repair deficiency, without detectable mutations in the genes encoding MMR proteins. LLS CRC cases present with MSI and deletion of MLH1, MSH2, MSH6 or PMS2 expression, as detected by immunohistochemistry (IHC) [25,26,27]. Unlike sporadic CRC, LLS does not display epigenetic silencing of MLH1 or mutations in the B-Raf proto-oncogene, serine/threonine kinase (BRAF). Studies have suggested that patients with LLS may harbor germline mutations in non-DNA MMR genes associated with the molecular mechanisms of LS, such as in the POLD1/POLE DNA polymerase genes [28].

The MSI is observed in approximately 15% of all sporadic CRC. MSI tumors are more common in the elderly (≥ 70 years) and are often found in women [29, 30]. Somatic mutation and epigenetic hypermethylation of the MLH1 gene promoter region are the main causes of sporadic MSI-H CRC, leading to transcriptional silencing [31, 32], The presence of the BRAFV600E is found in sporadic MSI CRC, and BRAFV600E is an activating mutation that leads to sustained mitogen-activated protein kinase (MAPK) pathway signalling [33, 34]. The high CpG island methylator phenotype (CIMP-H) refers to the high methylation status of three or five specific genes and is associated with MLH1 promoter methylation and MSI-H. Although CIMP-H patients show no response to 5-fluorouracil chemotherapy, those with CIMP-H and MSI have a higher survival rate [35].

LLS patients tend to be younger at onset, and the family history of CRC incidence is intermediate between LS and sporadic CRC [28], Among LLS CRC patients, 50-60% exhibit biallelic somatic inactivation of genes [36, 37]. LLS patients can be divided into two groups (Fig. 1A): those with a family history (potentially involving unknown hereditary risk factors) and those without a history of cancer (possibly harboring biallelic somatic mutations in MMR genes), the latter being considered as sporadic CRC [38, 39]. According to the revised Bethesda guidelines, a positive family history alone does not necessarily indicate a hereditary case; however, if the type of family history meets the Amsterdam criteria, it may suggest a hereditary case. Currently, there is no consensus on whether LLS should be classified as a potential hereditary or sporadic condition, as LLS patients may represent a heterogeneous group [40]. Although the use of multi-gene testing in LLS has increased in recent years, the optimal diagnostic strategy for LLS patients has not yet been established. Therefore, expert consensus is needed to accurately classify these cases as either true sporadic or potentially hereditary [38, 41]. Family history, age of onset, and prognosis are key clinical features distinguishing LLS patients from those with sporadic MSI-H CRC. Therefore, LLS cases encompass both sporadic and hereditary cases, and it is necessary to define molecular tools to effectively distinguish between the two groups [36].

Distinguishing between sporadic CRC patients and LS or LLS patients can be achieved through the age of CRC onset, a history of cancer in the family, and the presence of BRAFV600E and/or MLH1 methylation. Additionally, despite the lesion location being similar to LS, the presence of MLH1 hypermethylation and BRAFV600E in patients with sessile serrated adenoma/polyps (SSA) suggests that SSA is not part of LS [42].

dMMR/MSI-H CRC detection techniques

Current techniques used for MSI detection include IHC, PCR, NGS, and b-MSI assays [43].

The IHC detection of MMR protein loss and PCR amplification of microsatellite repeat sequences are widely used for clinical MSI screening. IHC is a simple and cost-effective method that has been widely used to detect protein expression. However, it cannot reflect gene variations and its screening utility is comparable to PCR analysis, with a consistency of 90.4-99.6% [44, 45]. IHC demonstrates a sensitivity of 74% for predicting MLH1 mutations and 91% for MSH2 mutations, alongside specificities of 81% and 90%, respectively. It effectively predicts the microsatellite instability (MSI) status in 76% of cases, with a specificity of 100% [46]. The interpretation criteria include the detection of four known MMR proteins (MLH1, MSH2, MSH6, and PMS2), with positive expression localized in the cell nucleus. If all four MMR proteins show positive expression, it is classified as pMMR. Any loss of expression of a MMR protein indicates dMMR, generally equivalent to the MSI-H phenotype.

The polymerase chain reaction (PCR) detection of MSI was the first established method for identifying MSI status in CRC and remains the current gold standard for MSI testing. The Bethesda panel, recommended by the National Cancer Institute in 1997, includes BAT-25, BAT-26, D5S346, D2S123, and D17S250. By 2002, this panel was revised to the Pentaplex panel, which added NR21 and NR24 to enhance sensitivity and specificity. In 2006, the MSI analysis system from Promega replaced NR27 with MONO-27, further increasing detection sensitivity. This panel has since been recognized by the National Cancer Institute as the international gold standard for identifying MSI in human tumors [47]. If two or more single nucleotide repeat fragment sizes change by ≥ 3 bp, the result is classified as MSI-H. Conversely, if only one fragment size changes by ≥ 3 bp or if no fragment size changes by > 3 bp, the result is classified as ‘non-MSI-H.’ It is important to highlight that studies have indicated that, in populations with high ethnic admixture, changes in a single nucleotide marker may correlate with the hereditary genetic characteristics of the individuals analyzed [48,49,50,51]. Recent reports have highlighted the high mutation frequency of the T17 single nucleotide repeat sequence in the gene encoding the chaperone HSP110 (HT17) in MSI CRC [52, 53]. Deletion of the HT17 repeat sequence in tumor DNA leads to exon 9 skipping (HSP110DE9), resulting in increased synthesis of HSP110 isoforms. Consequently, HT17 has been demonstrated as an improved biomarker for diagnosing MSI in CRC [54,55,56]. Despite its confirmed sensitivity, HT17 requires further evaluation, particularly in cancers beyond CRC.

Next-generation sequencing (NGS), as a second-generation sequencing method, provides comprehensive insights into genomic alterations and MSI status. Various algorithms using targeted, whole exome, or whole genome sequencing data have been designed to determine MSI status in NGS analysis [57,58,59], The NCCN guidelines recommend using validated NGS Panels for MSI testing, such as the FDA-approved MSK-IMPACT and Foundation One CDx, which cover 1000 and 95 MSI loci, respectively. These are particularly suitable for metastatic CRC patients requiring RAS and BRAF gene typing [60].Despite multiple MSI algorithms, the lack of a unified standard has led to challenges in standardizing NGS MSI testing. Moreover, NGS testing is costly and time-consuming, and large PD(L)1 clinical trials tend to use PCR or IHC rather than NGS to assess MSI/MMR status [61].

A blood-based MSI (b-MSI) NGS algorithm based on circulating tumor DNA (ctDNA) has also emerged as a new option for MSI testing in advanced solid tumor patients with difficult or inadequate tumor tissue sampling [62]. However, it should not be routinely recommended at present and only serves as an alternative means to definitively determine MSI status in patients lacking tissue. The content covered by b-MSI-NGS testing includes b-MSI, blood-based tumor mutational burden (b-TMB), and multi-gene (including MMR) germline/somatic mutation status.

Previous studies have shown a discordance rate of 1–10% between MMR protein IHC and MSI testing [63,64,65,66,67]. The common inconsistency is IHC indicating MSH6 expression loss while MSI indicates MSS, which is attributed to functional overlap between MMR proteins. IHC results may be misinterpreted due to poor staining quality. Therefore, under standardized operations and error avoidance, any positive result from either test can serve as a basis for using immune checkpoint inhibitors in advanced cancer (Table 1) [68].

Table 1 Comparison of MSI detection techniques and their accuracy

dMMR/MSI-H CRC pathologic types

dMMR/MSI-H is most commonly found in stage II colon cancer. Studies have shown that CRC with dMMR/MSI-H has similar pathological features: (1) the primary tumor is often located in the right half of the colon; (2) low pathological stage; (3) presence of mucinous components; (4) tumor‑infiltrating lymphocytes; (5) absence of necrotic cellular debris and the presence of a Crohn‑like nodular infiltrate [69,70,71] .

It can be seen that patients with dMMR/MSI-H CRC exhibit diverse prognostic characteristics. In comparison to patients with pMMR/MSS tumors, those with dMMR/MSI-H CRC demonstrate longer disease-free survival (DFS) and overall survival (OS), possibly due to higher lymphocyte infiltration in dMMR/MSI-H tumors. dMMR/MSI-H CRC may have an inflamed tumor microenvironment (TME), which can increase sensitivity to immune checkpoint inhibitors (ICIs) [72]. Another important pathological characteristic of MSI-H CRC is the peritumoral lymphocytic reaction, manifested by the clustering of lymphocytes into follicles surrounded by stellate fibroblasts, with some forming germinal centers [73]. Studies show that sporadic MSI-H CRC and LS CRC differ in histology and morphology. LS often presents with lymphocyte infiltration, tumor cell dedifferentiation, and adenomas, while sporadic MSI-H CRC are characterized by increased cytoplasmic eosinophils, large vacuolated nuclei, mucin secretion, poor differentiation, high heterogeneity, Crohn’s-like reactions, and a higher prevalence of serrated and sawtooth-like polyps [74, 75].And the LS patients typically exhibit more pronounced local T-cell infiltration and even higher mutational burden compared to sporadic dMMR/MSI-H CRC, leading to a better prognosis [76, 77].

In summary, different MSI statuses in CRC have specific clinicopathological features. Patients with dMMR/MSI-H CRC exhibit both favorable and unfavorable prognostic characteristics. The distinct nature of dMMR/MSI-H CRC determines the need for personalized treatment.

Advances in the treatment of dMMR/MSI-H CRC

The clinical trials of dMMR/MSI-H CRC have not yet clearly distinguished the differences in the immunotherapy efficacy between LS, LLS, and sporadic patients, and lack unified classification criteria. Consequently, molecular-driven differentiated treatments cannot yet be provided, and treatment continues to follow the standard protocols for dMMR/MSI-H CRC. KRAS/BRAF, as independent prognostic indicators, guide the targeted therapy of CRC, but direct associations with immunotherapy have not been discovered. Preliminary research results indicate that the BRAFV600E mutation cannot predict the outcomes of dMMR/MSI-H metastatic CRC (mCRC) patients receiving ICI therapy [78]. Therefore, for mCRC patients with dMMR/MSI-H accompanied by BRAFV600E mutation, ICI should be considered standard first-line treatment [79].

For dMMR/MSI-H patients who are amenable to curative resection, phase II studies have shown that neoadjuvant immunotherapy can achieve an major pathological response (MPR) rate of 97-100% and a pathologic complete response (pCR) rate of 65%~88% [80,81,82]. In cases of partially T4b, M0 dMMR/MSI-H patients where curative resection cannot be achieved through combined organ excision, consideration may be given to the use of programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) monoclonal antibodies, with pembrolizumab being the preferred option, as per the KEYNOTE-177 study and domestic indications for use in transformative or palliative therapy [83]. For dMMR/MSI-H stage III patients after curative surgery, studies related to the adjuvant therapy of ICIs are currently underway (Fig. 1B).

Neoadjuvant therapy

In the CheckMate-142 study, combination immunotherapy achieved high objective response rates (ORRs) in both later-line and first-line treatment of advanced dMMR/MSI-H mCRC. This has prompted researchers to explore the use of immunotherapy for neoadjuvant treatment.

The NICHE study is the first clinical trial to explore the adjuvant treatment of early-stage colon cancer using dual immunotherapy. The study involved 20 dMMR and 20 pMMR patients. dMMR patients received combined treatment with nivolumab and ipilimumab, while pMMR patients were randomly assigned to receive dual immunotherapy with or without cetuximab. The study confirmed the safety and feasibility of PD-1 monoclonal antibody combined with low-dose cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) monoclonal antibody therapy [80]. The NICHE-2 study [84] expanded the sample size of dMMR colon cancer patients, including 115 patients, with 111 participating in the study. These patients received combined neoadjuvant therapy with ipilimumab and nivolumab. The study results indicated that safety endpoints were met, with a MPR of 95% and a pCR of 68%. After a median follow-up of 13.1 months, no patients experienced recurrence [85]. Additionally, the results of the NICHE-3 study, presented at the 2023 ESMO Annual Meeting, demonstrated that using PD-1 monoclonal antibodies in combination with lymphocyte activation gene-3 (LAG-3) inhibitors as a dual adjuvant therapy for locally advanced dMMR colon adenocarcinoma patients achieved a pCR rate of 79% and a 100% pathological response rate [86]. These findings offer significant hope for adjuvant treatment of MSI-H/dMMR colon cancer patients, potentially realizing the goal of organ preservation and even drug-induced cure in the future.

The PICC study is a single-center, randomized, phase II clinical trial for patients with dMMR/MSI-H locally advanced CRC (LACRC), aiming to evaluate the efficacy of Trifluridine and Tipiracil Monotherapy or in combination with the cyclooxygenase-2 (COX-2) inhibitor Celecoxib as neoadjuvant therapy. The study confirmed that neoadjuvant therapy with Trifluridine and Tipiracil Monotherapy or in combination with Celecoxib is well tolerated and results in a high pCR rate [81].

The MDACC study is a phase II clinical trial of neoadjuvant therapy using pembrolizumab for dMMR/MSI-H locally advanced solid tumors, and the results showed a postoperative pCR rate of 83% and an MPR rate of 92% in CRC patients.

Pembrolizumab significantly improved the clinical outcomes of dMMR/MSI-H solid tumors, although there is still insufficient research on neoadjuvant therapy. A phase II study targeting locally unresectable or high-risk resectable dMMR/MSI-H tumors found that neoadjuvant pembrolizumab therapy is safe and effective, leading to a high pathological, radiological, and endoscopic response, which is of significant importance for organ preservation strategies [87].

The MSKCC study is a phase II clinical trial that explores the efficacy of the PD-1 inhibitor dostarlimab as a monotherapy or in combination with radiotherapy and chemotherapy in patients with dMMR-type LACRC. The study included 16 patients with dMMR LACRC, all of whom received six months of dostarlimab monotherapy. Of the 12 patients who completed the treatment, all achieved a complete clinical response (cCR) and did not require radiotherapy, chemotherapy, or surgery. During the 6 to 25 months of follow-up, no disease progression or recurrence was observed, and there were no reports of adverse events of grade 3 or above [88]. The study suggests that following neoadjuvant immunotherapy, patients may consider a watch-and-wait strategy, avoiding potential negative impacts on fertility, sexual health, as well as bowel and bladder function that may result from radiotherapy, chemotherapy, or surgery. Given the increasing incidence of rectal cancer in young patients, this study provides a new treatment option for individuals of childbearing age [89].

The SYSUCC study is a phase II clinical trial evaluating the use of sintilimab in treating dMMR/MSI-H CRC. The study confirms that PD-1 monoclonal antibody immunotherapy can lead to cCR in patients with locally advanced dMMR/MSI-H CRC, thus avoiding the need for radiotherapy, chemotherapy, and surgery while preserving organ function, fundamentally altering the treatment strategy for this disease [82].

In a phase Ib, randomized, and open-label study, the combination therapy of IBI310 and sintilimab was applied as neoadjuvant treatment for operable CRC patients with dMMR/MSI-H characteristics, and significant therapeutic effects were achieved. The core objective of this study was to explore the potential of dual immunotherapy in improving OS in patients. The research data revealed that when IBI310 was used in combination with sintilimab, the pathological complete response rate reached 80%, which was twice the rate of the group treated with sintilimab alone [90]. These findings not only highlight the significant advantage of dual immunotherapy in efficacy but also provide a new treatment option for patients, which may spare them from surgery and thus have broad prospects for clinical application.

A study evaluated the efficacy of tislelizumab as neoadjuvant therapy in patients with locally advanced dMMR/MSI-H CRC [91]. The study enrolled 33 patients aged ≥ 18 years with stage II/III disease, Eastern Cooperative Oncology Group Performance Status 0–1, and eligible for complete resection. Patients received tislelizumab 200 mg intravenously every three weeks for 3 cycles, followed by surgical resection within 10 weeks after the first dose of tislelizumab. Among the 29 patients who underwent surgery, the MPR rate was 89.7% and the pCR rate was 62.1%. The study results support the importance of MSI and MMR testing in the treatment of CRC, and demonstrate that tislelizumab achieves high MPR and pCR rates in patients with dMMR/MSI-H CRC, indicating significant anti-tumor activity and favorable surgical outcomes (Table 2).

Table 2 Key clinical trials of neoadjuvant therapy in dMMR/MSI-H CRC

Adjuvant therapy

Patients with dMMR/MSI-H CRC may benefit from adjuvant therapy using oxaliplatin-based regimens following curative surgery. However, a pooled analysis of four phase III trials revealed that dMMR/MSI-H CRC patients had significantly poorer progression-free survival (PFS) () and OS after standard first-line chemotherapy compared to pMMR/MSS CRC patients. This can be attributed to the high mutational burden of dMMR/MSI-H CRC tumors, which makes them responsive to immunotherapy but less responsive to certain chemotherapeutic agents. As a result, according to NCCN and CSCO guidelines, the use of fluoropyrimidine monotherapy for adjuvant therapy in stage II MSI-H CRC patients is not recommended [92].

First-line treatment

In the era of immunotherapy, the use of immune treatment in the first-line, early neoadjuvant, and adjuvant therapy for dMMR/MSI-H CRC patients has expanded. In 2020, the FDA approved pembrolizumab as a first-line treatment for unresectable or mCRC with dMMR/MSI-H [93]. The KEYNOTE-177 study showed that compared to chemotherapy, the median PFS of the pembrolizumab treatment group was longer (16.5 months vs. 8.2 months) and the ORR increased by 10.7%, confirming its lasting anti-tumor effects and good safety [83], Given its high efficacy and low toxicity, pembrolizumab was approved by the FDA in June 2020 for first-line treatment of dMMR/MSI-H mCRC [94, 95].

In the phase II multi-cohort CheckMate142 study [96], nivolumab monotherapy or in combination with ipilimumab for the treatment of mCRC patients demonstrated good efficacy and safety. The study showed a single-agent ORR of 34% for second-line and above treatments, and a combined treatment ORR of 65%; for first-line monotherapy, the ORR was 69%, with a disease control rate (DCR) of 84%. The median duration of response (DOR), PFS, and OS were not reached, and the 2-year PFS rate and OS rate were 74% and 79% respectively. The results suggest that dMMR/MSI-H LACRC patients can benefit from dual immunotherapy across all lines [97,98,99].

In the randomized phase III CheckMate 8HW study, the combination of nivolumab and ipilimumab significantly improved PFS in first-line treatment of dMMR/MSI-H mCRC compared to chemotherapy. At a median follow-up of 24.3 months, the PFS for the nivolumab + ipilimumab group exceeded 38 months, with a 2-year PFS rate of 72%, while the median PFS for the chemotherapy group was 5.9 months. The risk of disease progression or death was reduced by 79%. The study results support nivolumab + ipilimumab as a new standard for first-line treatment of dMMR/MSI-H mCRC patients [98].

Second and subsequent line of treatments

The results of the KEYNOTE-016 study, presented at the 2015 ASCO Annual Meeting, demonstrated the benefit of single-agent pembrolizumab treatment in patients with dMMR mCRC, marking the beginning of the era of immunotherapy for CRC [100].

KEYNOTE-164 further confirmed the efficacy and manageable safety profile of pembrolizumab in dMMR/MSI-H CRC patients [101, 102]. In 2017, both pembrolizumab and nivolumab received FDA approval for use in dMMR/MSI-H mCRC. The recommendations for CRC immunotherapy mainly apply to dMMR/MSI-H patients. Updated results from the CheckMate142 study demonstrated a 12-month PFS of 77% and OS of 84% in first-line dual immunotherapy. Nivolumab provides durable relief and disease control in pretreated dMMR/MSI-H mCRC patients, potentially establishing a novel treatment avenue for this patient population [103].

PRODIGE54-SAMCO is a randomized phase II trial that assessed the efficacy and safety of avelumab as a second-line treatment for dMMR/MSI-H mCRC patients. The study found avelumab to be more effective than standard second-line therapies and confirmed that anti-PD-(L)1 monoclonal antibodies retain their efficacy even after first-line treatment, without prior use of immune checkpoint inhibitors. This implies that dMMR/MSI-H mCRC patients should receive ICI therapy as early as possible [104].

Currently approved PD-1/PD-L1 monoclonal antibodies lack phase III randomized controlled clinical trial data for advanced second-line or first-line CRC patients. However, based on the results of phase II clinical studies across various tumor types, some domestically produced PD-1/PD-L1 monoclonal antibodies have obtained domestic indications for the later-line treatment of dMMR/MSI-H solid tumors, such as domestic monoclonal antibodies including envafolimab, serplulimab, tislelizumab, and pucotenlimab [105, 106], as well as imported pembrolizumab and nivolumab (Table 3).

Table 3 Key clinical trials of immunotherapy in dMMR/MSI-H CRC

Other treatments

The Werner Syndrome ATP-dependent Helicase (WRN) enzyme is considered a potential synthetic lethality target for dMMR/MSI-H cancers, showing broad sensitivity, particularly in CRC [107]. A series of studies have elucidated the molecular mechanisms by which MSI-H cancer cells rely on WRN to maintain genomic stability, indicating that the synthetic lethality relationship with WRN arises from the consequences of MMR dysfunction rather than a defect in MMR itself. Research has shown that tumors with WRN mutations exhibit higher TMB, increased PD-L1 expression, and dMMR/MSI-H characteristics compared to wild-type tumors [108]. Although no drugs have yet been approved globally, several WRN inhibitors are in development. VVD-133,214 and HRO761 are two WRN inhibitors showing significant progress [109, 110]. These advancements suggest that WRN inhibitors could offer new therapeutic options for MSI-H cancer patients and may become a strategy in precision medicine [107].

Fecal microbiota transplantation (FMT) involves extracting beneficial microorganisms from the feces of healthy donors and transplanting them into a patient’s gut via endoscopy or capsules to restore microbial balance. A case report in 2018 preliminarily confirmed the potential efficacy of FMT in treating immune-related adverse events [111]. Recent studies have shown that combining FMT with anti-PD-1 therapy significantly improves the median PFS and OS in patients with MSS mCRC [112]. Research also indicates that ginsenoside Rh4 can inhibit CRC progression by modulating gut microbiota balance and bile acid metabolism [113]. Additionally, the presence of Fusobacterium nucleatum (Fn) is closely associated with various pathological features of CRC [114]. Detection of Fn DNA in colorectal tissues has revealed that Fn-positive patients have lower cancer-specific survival and OS, particularly those with Fn strain C2 [115], which dominates the TME and is closely linked to poorer prognosis. In summary, FMT not only plays a role in regulating gut microbiota balance but may also synergize with immunotherapy to enhance CRC treatment outcomes. Furthermore, gut microbiota members such as Fn may serve as biomarkers for prognosis and treatment response in CRC, providing new perspectives and strategies for precision treatment of the disease.

Prognostic factors

The latest study utilized Cox proportional hazards regression models to analyze the impact of KRAS exon 2 and BRAFV600E mutations on time to relapse (TTR), OS, and survival after relapse (SAR) in MSI-H CRC patients [116]. The results indicated that in dMMR/MSI-H patients, these mutations showed minimal differences in 5-year TTR and OS. However, regardless of MSI status, patients with these mutations had shorter SAR. Additionally, prognostic impact analysis of dMMR/MSI-H mCRC patients receiving immunotherapy revealed that in sporadic CRC and LS patients, LS patients showed a significantly improved PFS, while there was no statistically significant difference in OS improvement, suggesting that LS CRC patients may have a better OS rate [117].

Directions of future research

Currently, multiple studies are focused on enhancing the efficacy of anti-PD-1 therapy for dMMR/MSI-H mCRC through immune combination therapy. For instance, a ready-made cancer vaccine Nous-209, based on shared neoantigens, has been proven safe and has demonstrated preliminary anti-tumor effects when used in combination with pembrolizumab for dMMR/MSI-H CRC and gastric cancer patients (NCT04041310).

The introduction of immune checkpoint inhibitors has made immunotherapy more attractive and promising for dMMR patients compared to pMMR patients. However, for operable MSI-H CRC, whether neoadjuvant immunotherapy can replace surgery, and whether adjuvant immunotherapy is more beneficial for MSI-H patients than adjuvant chemotherapy, currently lacks definitive conclusions. The potential value of immunotherapy before and after surgery requires further confirmation through phase III clinical studies. Additionally, primary and acquired resistance has been observed in over 50% of dMMR/MSI-H CRC patients and identifying these patients and overcoming resistance will be an important challenge in the future [118]. Currently, there are currently no studies differentiating the treatment differences among LS, LLS, and sporadic CRC types, and the optimal diagnostic criteria for LLS patients have yet to be established. research and targeted therapeutic drug development for various subtypes, as well as the precise stratification of CRC and the innovation of the treatment layout for advanced colon cancer patients, are promising endeavors.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

CRC:

Colorectal cancer

pMMR:

Proficient mismatch repair

MSS:

Microsatellite stability

dMMR:

Mismatch repair deficiency

MSI-H:

Microsatellite instability high

LS:

Lynch syndrome

LLS:

Lynch-like syndrome

MMR:

Mismatch repair

MSI:

Microsatellite instability

MLH1:

Mutl homolog 1

hMLH1:

Human mutl homolog 1

MSH2:

Muts homolog 2

hMSH2:

Human muts homolog 2

MSH6:

Muts homolog 6

hMSH6:

Human muts homolog 6

PMS2:

Postmeiotic segregation increased 2

hPMS2:

Human postmeiotic segregation increased 2

EPCAM:

Epithelial cell adhesion molecule

MSI-L:

Low-level microsatellite instability

HNPCC:

Hereditary non-polyposis colorectal cancer

IHC:

Immunohistochemistry

BRAF:

B-Raf proto-oncogene, serine/threonine kinase

MAPK:

Mitogen-activated protein kinase

CIMP-H:

High CpG island methylator phenotype

SSA:

Sessile serrated adenoma/polyps

PCR:

Polymerase chain reaction

NCCN:

National Comprehensive Cancer Network

CSCO:

Chinese Society of Clinical Oncology

NGS:

Next-generation sequencing

b-MSI:

Blood-based MSI

ctDNA:

Circulating tumor DNA

b-TMB:

Blood-based tumor mutational burden

CLR:

Crohn’s-like lymphoid reaction

KRAS:

Kirsten rat sarcoma

RAS:

Rat sarcoma viral oncogene homolog

DFS:

Disease-free survival

OS:

Overall survival

CR:

Complete response

ICI:

Immune checkpoint inhibitors

pCR:

Pathologic complete response

MPR:

Major pathological response

PD-1:

Programmed cell death protein 1

PD-L1:

Programmed cell death-ligand 1

ORRs:

Objective response rates

mCRC:

Metastatic colorectal cancer

CTLA-4:

Cytotoxic T-lymphocyte-associated protein 4

ESMO:

European Society for Medical Oncology

LACRC:

Locally advanced colorectal cancer

COX-2:

Cyclooxygenase-2

cCR:

Complete clinical response

LAG-3:

Lymphocyte activation gene-3

DOR:

Duration of response

DCR:

Disease control rate

PFS:

Progression-free survival

SAEs:

Serious adverse events

WRN:

Werner syndrome ATP-dependent helicase

TME:

Tumor microenvironment

TTR:

Time to relapse

References

  1. Ladabaum U. What is Lynch-like syndrome and how should we manage it? Clinical gastroenterology and hepatology: the official clinical practice. J Am Gastroenterological Association. 2020;18(2):294–6.

    Google Scholar 

  2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. Cancer J Clin. 2021;71(3):209–49.

    Article  Google Scholar 

  3. Zeng H, Chen W, Zheng R, Zhang S, Ji JS, Zou X, Xia C, Sun K, Yang Z, Li H, Wang N, Han R, Liu S, Li H, Mu H, He Y, Xu Y, Fu Z, Zhou Y, Jiang J, Yang Y, Chen J, Wei K, Fan D, Wang J, Fu F, Zhao D, Song G, Chen J, Jiang C, Zhou X, Gu X, Jin F, Li Q, Li Y, Wu T, Yan C, Dong J, Hua Z, Baade P, Bray F, Jemal A, Yu XQ, He J. Changing cancer survival in China during 2003-15: a pooled analysis of 17 population-based cancer registries. The Lancet. Global health 2018, 6 (5), e555-e567.

  4. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer Stat 2023 CA: cancer J Clin. 2023;73(1):17–48.

    Google Scholar 

  5. Siegel RL, Wagle NS, Cercek A, Smith RA, Jemal A. Colorectal cancer statistics, 2023. Cancer J Clin. 2023;73(3):233–54.

    Article  Google Scholar 

  6. Baziliansky S, Cohen M. Post-treatment psychological distress among Colorectal Cancer survivors: relation to emotion regulation patterns and Personal resources. Int J Behav Med. 2021;28(5):591–601.

    Article  PubMed  Google Scholar 

  7. Global. Regional, and national burden of colorectal cancer and its risk factors, 1990–2019: a systematic analysis for the global burden of Disease Study 2019. Lancet Gastroenterol Hepatol. 2022;7(7):627–47.

    Article  Google Scholar 

  8. Vilar E, Gruber SB. Microsatellite instability in colorectal cancer-the stable evidence. Nat Rev Clin Oncol. 2010;7(3):153–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sinicrope FA, Sargent DJ. Molecular pathways: microsatellite instability in colorectal cancer: prognostic, predictive, and therapeutic implications. Clin cancer Research: Official J Am Association Cancer Res. 2012;18(6):1506–12.

    Article  CAS  Google Scholar 

  10. Gelsomino F, Barbolini M, Spallanzani A, Pugliese G, Cascinu S. The evolving role of microsatellite instability in colorectal cancer: a review. Cancer Treat Rev. 2016;51:19–26.

    Article  CAS  PubMed  Google Scholar 

  11. Provenzale D, Gupta S, Ahnen DJ, Bray T, Cannon JA, Cooper G, David DS, Early DS, Erwin D, Ford JM, Giardiello FM, Grady W, Halverson AL, Hamilton SR, Hampel H, Ismail MK, Klapman JB, Larson DW, Lazenby AJ, Lynch PM, Mayer RJ, Ness RM, Regenbogen SE, Samadder NJ, Shike M, Steinbach G, Weinberg D, Dwyer M, Darlow S. Genetic/Familial High-Risk Assessment: Colorectal Version 1.2016, NCCN Clinical Practice guidelines in Oncology. J Natl Compr Cancer Network: JNCCN. 2016;14(8):1010–30.

    Article  CAS  Google Scholar 

  12. Daly MB, Pal T, Berry MP, Buys SS, Dickson P, Domchek SM, Elkhanany A, Friedman S, Goggins M, Hutton ML, Karlan BY, Khan S, Klein C, Kohlmann W, Kurian AW, Laronga C, Litton JK, Mak JS, Menendez CS, Merajver SD, Norquist BS, Offit K, Pederson HJ, Reiser G, Senter-Jamieson L, Shannon KM, Shatsky R, Visvanathan K, Weitzel JN, Wick MJ, Wisinski KB, Yurgelun MB, Darlow SD, Dwyer MA. Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network: JNCCN 2021, 19 (1), 77–102.

  13. Al-Sohaily S, Biankin A, Leong R, Kohonen-Corish M, Warusavitarne J. Molecular pathways in colorectal cancer. J Gastroenterol Hepatol. 2012;27(9):1423–31.

    Article  CAS  PubMed  Google Scholar 

  14. Wong MCS, Huang J, Lok V, Wang J, Fung F, Ding H, Zheng ZJ. Differences in incidence and mortality trends of Colorectal Cancer Worldwide based on sex, Age, and anatomic location. Clin Gastroenterol Hepatology: Official Clin Pract J Am Gastroenterological Association. 2021;19(5):955–e96661.

    Article  Google Scholar 

  15. Wong MC, Ding H, Wang J, Chan PS, Huang J. Prevalence and risk factors of colorectal cancer in Asia. Intestinal Res. 2019;17(3):317–29.

    Article  Google Scholar 

  16. Soliman AS, Bondy ML, El-Badawy SA, Mokhtar N, Eissa S, Bayoumy S, Seifeldin IA, Houlihan PS, Lukish JR, Watanabe T, Chan AO, Zhu D, Amos CI, Levin B, Hamilton SR. Contrasting molecular pathology of colorectal carcinoma in Egyptian and western patients. Br J Cancer. 2001;85(7):1037–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Waller J, Gu L, De Hoedt AM, Freedland SJ, Wang T, Amonkar M, Aurora-Garg D, Liaw KL, Wehn A, Klaassen Z. DNA mismatch repair and microsatellite instability in colorectal tumors: an observational study in the Veterans Affairs Health Care System. Future Oncol (London England). 2022;18(6):649–60.

    Article  CAS  Google Scholar 

  18. Sougklakos I, Athanasiadis E, Boukovinas I, Karamouzis M, Koutras A, Papakotoulas P, Latsou D, Hatzikou M, Stamuli E, Balasopoulos A, Sideris A. Treatment pathways and associated costs of metastatic colorectal cancer in Greece. Cost Eff Resource Allocation: C/E. 2022;20(1):7.

    Article  Google Scholar 

  19. Akagi K, Oki E, Taniguchi H, Nakatani K, Aoki D, Kuwata T, Yoshino T. Real-world data on microsatellite instability status in various unresectable or metastatic solid tumors. Cancer Sci. 2021;112(3):1105–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhang C, Ding H, Sun S, Luan Z, Liu G, Li Z. Incidence and detection of high microsatellite instability in colorectal cancer in a Chinese population: a meta-analysis. J Gastrointest Oncol. 2020;11(6):1155–63.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Kanth P, Grimmett J, Champine M, Burt R, Samadder NJ. Hereditary Colorectal Polyposis and Cancer syndromes: a primer on diagnosis and management. Am J Gastroenterol. 2017;112(10):1509–25.

    Article  PubMed  Google Scholar 

  22. Pathak SJ, Mueller JL, Okamoto K, Das B, Hertecant J, Greenhalgh L, Cole T, Pinsk V, Yerushalmi B, Gurkan OE, Yourshaw M, Hernandez E, Oesterreicher S, Naik S, Sanderson IR, Axelsson I, Agardh D, Boland CR, Martin MG, Putnam CD, Sivagnanam M. EPCAM mutation update: variants associated with congenital tufting enteropathy and Lynch syndrome. Hum Mutat. 2019;40(2):142–61.

    Article  CAS  PubMed  Google Scholar 

  23. Vasen HF, Wijnen JT, Menko FH, Kleibeuker JH, Taal BG, Griffioen G, Nagengast FM, Meijers-Heijboer EH, Bertario L, Varesco L, Bisgaard ML, Mohr J, Fodde R, Khan PM. Cancer risk in families with hereditary nonpolyposis colorectal cancer diagnosed by mutation analysis. Gastroenterology. 1996;110(4):1020–7.

    Article  CAS  PubMed  Google Scholar 

  24. Hampel H, Stephens JA, Pukkala E, Sankila R, Aaltonen LA, Mecklin JP, de la Chapelle A. Cancer risk in hereditary nonpolyposis colorectal cancer syndrome: later age of onset. Gastroenterology. 2005;129(2):415–21.

    Article  PubMed  Google Scholar 

  25. Haraldsdottir S, Hampel H, Tomsic J, Frankel WL, Pearlman R, de la Chapelle A, Pritchard CC. Colon and endometrial cancers with mismatch repair deficiency can arise from somatic, rather than germline, mutations. Gastroenterology. 2014;147(6):1308–e13161.

    Article  PubMed  Google Scholar 

  26. Rodríguez-Soler M, Pérez-Carbonell L, Guarinos C, Zapater P, Castillejo A, Barberá VM, Juárez M, Bessa X, Xicola RM, Clofent J, Bujanda L, Balaguer F, Reñé JM, de-Castro L, Marín-Gabriel JC, Lanas A, Cubiella J, Nicolás-Pérez D, Brea-Fernández A, Castellví-Bel S, Alenda C, Ruiz-Ponte C, Carracedo A, Castells A, Andreu M, Llor X, Soto JL, Payá A, Jover R. Risk of cancer in cases of suspected lynch syndrome without germline mutation. Gastroenterology. 2013;144(5):926–32. .e1; quiz e13-4.

    Article  PubMed  Google Scholar 

  27. Geurts-Giele WR, Leenen CH, Dubbink HJ, Meijssen IC, Post E, Sleddens HF, Kuipers EJ, Goverde A, van den Ouweland AM, van Lier MG, Steyerberg EW, van Leerdam ME, Wagner A, Dinjens WN. Somatic aberrations of mismatch repair genes as a cause of microsatellite-unstable cancers. J Pathol. 2014;234(4):548–59.

    Article  CAS  PubMed  Google Scholar 

  28. Magrin L, Fanale D, Brando C, Fiorino A, Corsini LR, Sciacchitano R, Filorizzo C, Dimino A, Russo A, Bazan V. POLD1, and NTHL1: the last but not the least hereditary cancer-predisposing genes. Oncogene. 2021;40(40):5893–901.

    Article  CAS  PubMed  Google Scholar 

  29. Carethers JM, Murali B, Yang B, Doctolero RT, Tajima A, Basa R, Smith EJ, Lee M, Janke R, Ngo T, Tejada R, Ji M, Kinseth M, Cabrera BL, Miyai K, Keku TO, Martin CF, Galanko JA, Sandler RS, McGuire KL. Influence of race on microsatellite instability and CD8 + T cell infiltration in colon cancer. PLoS ONE 2014, 9 (6), e100461.

  30. Carethers JM, Stoffel EM. Lynch syndrome and Lynch syndrome mimics: the growing complex landscape of hereditary colon cancer. World J Gastroenterol. 2015;21(31):9253–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Miyakura Y, Sugano K, Konishi F, Ichikawa A, Maekawa M, Shitoh K, Igarashi S, Kotake K, Koyama Y, Nagai H. Extensive methylation of hMLH1 promoter region predominates in proximal colon cancer with microsatellite instability. Gastroenterology. 2001;121(6):1300–9.

    Article  CAS  PubMed  Google Scholar 

  32. Nakagawa H, Nuovo GJ, Zervos EE, Martin EW Jr., Salovaara R, Aaltonen LA, de la Chapelle A. Age-related hypermethylation of the 5’ region of MLH1 in normal colonic mucosa is associated with microsatellite-unstable colorectal cancer development. Cancer Res. 2001;61(19):6991–5.

    CAS  PubMed  Google Scholar 

  33. Stoffel EM, Kastrinos F. Familial colorectal cancer, beyond Lynch syndrome. Clin Gastroenterol Hepatology: Official Clin Pract J Am Gastroenterological Association. 2014;12(7):1059–68.

    Article  Google Scholar 

  34. Carethers JM. One colon lumen but two organs. Gastroenterology. 2011;141(2):411–2.

    Article  PubMed  Google Scholar 

  35. Meessen S, Currey N, Jahan Z, Parker HW, Jenkins MA, Buchanan DD, Hopper JL, Segelov E, Dahlstrom JE, Kohonen-Corish MRJ. Tetranucleotide and Low Microsatellite Instability Are Inversely Associated with the CpG Island Methylator phenotype in Colorectal Cancer. Cancers. 2021;13:14.

    Article  Google Scholar 

  36. Martínez-Roca A, Giner-Calabuig M, Murcia O, Castillejo A, Soto JL, García-Heredia A, Jover R. Lynch-like syndrome: potential mechanisms and management. Cancers 2022, 14 (5).

  37. Golubicki M, Díaz-Gay M, Bonjoch L, Franch-Expósito S, Muñoz J, Cuatrecasas M, Ocaña T, Iseas S, Mendez G, Carballido M, Robbio J, Cisterna D, Roca E, Castells A, Balaguer F, Castellví-Bel S, Antelo M. Comprehensive genomic characterization of Fifteen Early-Onset Lynch-Like Syndrome Colorectal cancers. Cancers. 2021;13:6.

    Article  Google Scholar 

  38. Picó MD, Sánchez-Heras AB, Castillejo A, Giner-Calabuig M, Alustiza M, Sánchez A, Moreira L, Pellise M, Castells A, Llort G, Yagüe C, Ramon YCT, Gisbert-Beamud A, Cubiella J, Rivas L, Herraiz M, Garau C, Salces I, Carrillo-Palau M, Bujanda L, López-Fernández A, Alvarez-Urturi C, López MJ, Alenda C, Zapater P, Lacueva FJ, Balaguer F, Soto JL, Murcia Ó, Jover R. Risk of Cancer in Family members of patients with Lynch-Like Syndrome. Cancers. 2020;12:8.

    Article  Google Scholar 

  39. Picó MD, Castillejo A, Murcia Ó, Giner-Calabuig M, Alustiza M, Sánchez A, Moreira L, Pellise M, Castells A, Carrillo-Palau M, Ramon YCT, Gisbert-Beamud A, Llort G, Yagüe C, López-Fernández A, Alvarez-Urturi C, Cubiella J, Rivas L, Rodríguez-Alcalde D, Herraiz M, Garau C, Dolz C, Bujanda L, Cid L, Povés C, Garzon M, Salces I, Ponce M, Hernández-Villalba L, Alenda C, Balaguer F, Soto JL, Jover R. Clinical and pathological characterization of Lynch-Like Syndrome. Clin Gastroenterol Hepatology: Official Clin Pract J Am Gastroenterological Association. 2020;18(2):368–e3741.

    Article  Google Scholar 

  40. Carethers JM. Differentiating Lynch-like from Lynch syndrome. Gastroenterology. 2014;146(3):602–4.

    Article  PubMed  Google Scholar 

  41. Pérez-Carbonell L, Ruiz-Ponte C, Guarinos C, Alenda C, Payá A, Brea A, Egoavil CM, Castillejo A, Barberá VM, Bessa X, Xicola RM, Rodríguez-Soler M, Sánchez-Fortún C, Acame N, Castellví-Bel S, Piñol V, Balaguer F, Bujanda L, De-Castro ML, Llor X, Andreu M, Carracedo A, Soto JL, Castells A, Jover R. Comparison between universal molecular screening for Lynch syndrome and revised Bethesda guidelines in a large population-based cohort of patients with colorectal cancer. Gut. 2012;61(6):865–72.

    Article  PubMed  Google Scholar 

  42. Leggett B, Whitehall V. Role of the serrated pathway in colorectal cancer pathogenesis. Gastroenterology. 2010;138(6):2088–100.

    Article  CAS  PubMed  Google Scholar 

  43. Bayle A, Belcaid L, Aldea M, Vasseur D, Peyraud F, Nicotra C, Geraud A, Sakkal M, Seknazi L, Cerbone L, Blanc-Durand F, Hadoux J, Mosele F, Tagliamento M, Bernard-Tessier A, Verret B, Smolenschi C, Clodion R, Auger N, Romano PM, Gazzah A, Camus MN, Micol J, Caron O, Hollebecque A, Loriot Y, Besse B, Lacroix L, Rouleau E, Ponce S, Soria JC, Barlesi F, Andre F, Italiano A. Clinical utility of circulating tumor DNA sequencing with a large panel: a National Center for Precision Medicine (PRISM) study. Annals Oncology: Official J Eur Soc Med Oncol. 2023;34(4):389–96.

    Article  CAS  Google Scholar 

  44. Cheah PL, Li J, Looi LM, Koh CC, Lau TP, Chang SW, Teoh KH, Mun KS, Nazarina AR. Screening for microsatellite instability in colorectal carcinoma: practical utility of immunohistochemistry and PCR with fragment analysis in a diagnostic histopathology setting. Malays J Pathol. 2019;41(2):91–100.

    CAS  PubMed  Google Scholar 

  45. How-Kit A, Daunay A, Buhard O, Meiller C, Sahbatou M, Collura A, Duval A, Deleuze JF. Major improvement in the detection of microsatellite instability in colorectal cancer using HSP110 T17 E-ice-COLD-PCR. Hum Mutat. 2018;39(3):441–53.

    Article  CAS  PubMed  Google Scholar 

  46. Huang MN, McPherson JR, Cutcutache I, Teh BT, Tan P, Rozen SG. MSIseq: Software for assessing microsatellite instability from catalogs of somatic mutations. Sci Rep. 2015;5:13321.

    Article  CAS  PubMed  Google Scholar 

  47. Umar A, Boland CR, Terdiman JP, Syngal S, de la Chapelle A, Rüschoff J, Fishel R, Lindor NM, Burgart LJ, Hamelin R, Hamilton SR, Hiatt RA, Jass J, Lindblom A, Lynch HT, Peltomaki P, Ramsey SD, Rodriguez-Bigas MA, Vasen HF, Hawk ET, Barrett JC, Freedman AN, Srivastava S. Revised Bethesda guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst. 2004;96(4):261–8.

    Article  CAS  PubMed  Google Scholar 

  48. Chang L, Chang M, Chang HM, Chang F. Microsatellite instability: a predictive biomarker for Cancer Immunotherapy. Appl Immunohistochem Mol Morphology: AIMM. 2018;26(2):e15–21.

    Article  CAS  Google Scholar 

  49. Latham A, Shia J, Patel Z, Reidy-Lagunes DL, Segal NH, Yaeger R, Ganesh K, Connell L, Kemeny NE, Kelsen DP, Hechtman JF, Nash GM, Paty PB, Zehir A, Tkachuk KA, Sheikh R, Markowitz AJ, Mandelker D, Offit K, Berger MF, Cercek A, Garcia-Aguilar J, Saltz LB, Weiser MR, Stadler ZK. Characterization and clinical outcomes of DNA mismatch repair-deficient small bowel adenocarcinoma. Clin cancer Research: Official J Am Association Cancer Res. 2021;27(5):1429–37.

    Article  Google Scholar 

  50. Roudko V, Bozkus CC, Orfanelli T, McClain CB, Carr C, O’Donnell T, Chakraborty L, Samstein R, Huang KL, Blank SV, Greenbaum B, Bhardwaj N. Shared Immunogenic poly-epitope frameshift mutations in microsatellite unstable tumors. Cell. 2020;183(6):1634–e164917.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Boland CR, Thibodeau SN, Hamilton SR, Sidransky D, Eshleman JR, Burt RW, Meltzer SJ, Rodriguez-Bigas MA, Fodde R, Ranzani GN, Srivastava S. A National Cancer Institute Workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res. 1998;58(22):5248–57.

    CAS  PubMed  Google Scholar 

  52. Dorard C, de Thonel A, Collura A, Marisa L, Svrcek M, Lagrange A, Jego G, Wanherdrick K, Joly AL, Buhard O, Gobbo J, Penard-Lacronique V, Zouali H, Tubacher E, Kirzin S, Selves J, Milano G, Etienne-Grimaldi MC, Bengrine-Lefèvre L, Louvet C, Tournigand C, Lefèvre JH, Parc Y, Tiret E, Fléjou JF, Gaub MP, Garrido C, Duval A. Expression of a mutant HSP110 sensitizes colorectal cancer cells to chemotherapy and improves disease prognosis. Nat Med. 2011;17(10):1283–9.

    Article  CAS  PubMed  Google Scholar 

  53. Collura A, Lagrange A, Svrcek M, Marisa L, Buhard O, Guilloux A, Wanherdrick K, Dorard C, Taieb A, Saget A, Loh M, Soong R, Zeps N, Platell C, Mews A, Iacopetta B, De Thonel A, Seigneuric R, Marcion G, Chapusot C, Lepage C, Bouvier AM, Gaub MP, Milano G, Selves J, Senet P, Delarue P, Arzouk H, Lacoste C, Coquelle A, Bengrine-Lefèvre L, Tournigand C, Lefèvre JH, Parc Y, Biard DS, Fléjou JF, Garrido C, Duval A. Patients with colorectal tumors with microsatellite instability and large deletions in HSP110 T17 have improved response to 5-fluorouracil–based chemotherapy. Gastroenterology. 2014;146(2):401–e111.

    Article  CAS  PubMed  Google Scholar 

  54. Buhard O, Lagrange A, Guilloux A, Colas C, Chouchène M, Wanherdrick K, Coulet F, Guillerm E, Dorard C, Marisa L, Bokhari A, Greene M, El-Murr N, Bodo S, Muleris M, Sourouille I, Svrcek M, Cervera P, Blanché H, Lefevre JH, Parc Y, Lepage C, Chapusot C, Bouvier AM, Gaub MP, Selves J, Garrett K, Iacopetta B, Soong R, Hamelin R, Garrido C, Lascols O, André T, Fléjou JF, Collura A, Duval A. HSP110 T17 simplifies and improves the microsatellite instability testing in patients with colorectal cancer. J Med Genet. 2016;53(6):377–84.

    Article  CAS  PubMed  Google Scholar 

  55. Berardinelli GN, Scapulatempo-Neto C, Durães R, de Antônio M, Guimarães D, Reis RM. Advantage of HSP110 (T17) marker inclusion for microsatellite instability (MSI) detection in colorectal cancer patients. Oncotarget. 2018;9(47):28691–701.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Cohen R, Hain E, Buhard O, Guilloux A, Bardier A, Kaci R, Bertheau P, Renaud F, Bibeau F, Fléjou JF, André T, Svrcek M, Duval A. Association of Primary Resistance to Immune checkpoint inhibitors in metastatic colorectal Cancer with misdiagnosis of microsatellite instability or Mismatch Repair Deficiency Status. JAMA Oncol. 2019;5(4):551–5.

    Article  PubMed  Google Scholar 

  57. Ruiz-Bañobre J, Goel A. DNA mismatch Repair Deficiency and Immune checkpoint inhibitors in gastrointestinal cancers. Gastroenterology. 2019;156(4):890–903.

    Article  PubMed  Google Scholar 

  58. Foltz SM, Liang WW, Xie M, Ding L. MIRMMR: binary classification of microsatellite instability using methylation and mutations. Bioinf (Oxford England). 2017;33(23):3799–801.

    CAS  Google Scholar 

  59. Hirotsu Y, Nagakubo Y, Amemiya K, Oyama T, Mochizuki H, Omata M. Microsatellite instability status is determined by targeted sequencing with MSIcall in 25 cancer types. Clin Chim Acta. 2020;502:207–13.

    Article  CAS  PubMed  Google Scholar 

  60. Network. NC. NCCN clinical practiceguidelines in oncology(NCCN Guidelines). https://www.nccn.org/patientresources/patient-resources/guidelines-for-patients (accessed 2023-01-25).

  61. Waalkes A, Smith N, Penewit K, Hempelmann J, Konnick EQ, Hause RJ, Pritchard CC, Salipante SJ. Accurate Pan-cancer Molecular diagnosis of microsatellite instability by single-molecule Molecular Inversion Probe capture and high-throughput sequencing. Clin Chem. 2018;64(6):950–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kasi PM, Bucheit LA, Liao J, Starr J, Barata P, Klempner SJ, Gandara D, Shergill A, da Madeira L, Weipert C, Zhang N, Pretz C, Hardin A, Kiedrowski LA, Odegaard JI. Pan-cancer prevalence of microsatellite instability-high (MSI-H) identified by circulating Tumor DNA and Associated Real-World Clinical outcomes. JCO Precision Oncol. 2023;7:e2300118.

    Article  Google Scholar 

  63. Hatch SB, Lightfoot HM Jr., Garwacki CP, Moore DT, Calvo BF, Woosley JT, Sciarrotta J, Funkhouser WK, Farber RA. Microsatellite instability testing in colorectal carcinoma: choice of markers affects sensitivity of detection of mismatch repair-deficient tumors. Clin cancer Research: Official J Am Association Cancer Res. 2005;11(6):2180–7.

    Article  CAS  Google Scholar 

  64. Piñol V, Castells A, Andreu M, Castellví-Bel S, Alenda C, Llor X, Xicola RM, Rodríguez-Moranta F, Payá A, Jover R, Bessa X. Accuracy of revised Bethesda guidelines, microsatellite instability, and immunohistochemistry for the identification of patients with hereditary nonpolyposis colorectal cancer. JAMA. 2005;293(16):1986–94.

    Article  PubMed  Google Scholar 

  65. Watson N, Grieu F, Morris M, Harvey J, Stewart C, Schofield L, Goldblatt J, Iacopetta B. Heterogeneous staining for mismatch repair proteins during population-based prescreening for hereditary nonpolyposis colorectal cancer. J Mol Diagnostics: JMD. 2007;9(4):472–8.

    Article  CAS  PubMed  Google Scholar 

  66. Lindor NM, Burgart LJ, Leontovich O, Goldberg RM, Cunningham JM, Sargent DJ, Walsh-Vockley C, Petersen GM, Walsh MD, Leggett BA, Young JP, Barker MA, Jass JR, Hopper J, Gallinger S, Bapat B, Redston M, Thibodeau SN. Immunohistochemistry versus microsatellite instability testing in phenotyping colorectal tumors. J Clin Oncology: Official J Am Soc Clin Oncol. 2002;20(4):1043–8.

    Article  CAS  Google Scholar 

  67. Zhao S, Chen L, Zang Y, Liu W, Liu S, Teng F, Xue F, Wang Y. Endometrial cancer in Lynch syndrome. Int J Cancer. 2022;150(1):7–17.

    Article  CAS  PubMed  Google Scholar 

  68. Bartley AN, Mills AM, Konnick E, Overman M, Ventura CB, Souter L, Colasacco C, Stadler ZK, Kerr S, Howitt BE, Hampel H, Adams SF, Johnson W, Magi-Galluzzi C, Sepulveda AR, Broaddus RR. Mismatch repair and microsatellite instability testing for Immune checkpoint inhibitor therapy: Guideline from the College of American Pathologists in Collaboration with the Association for Molecular Pathology and Fight Colorectal Cancer. Arch Pathol Lab Med. 2022;146(10):1194–210.

    Article  CAS  PubMed  Google Scholar 

  69. Shin US, Cho SS, Moon SM, Park SH, Jee SH, Jung EJ, Hwang DY. Is microsatellite instability really a good prognostic factor of colorectal cancer? Annals Coloproctology. 2014;30(1):28–34.

    Article  Google Scholar 

  70. Karahan B, Argon A, Yıldırım M, Vardar E. Relationship between MLH-1, MSH-2, PMS-2,MSH-6 expression and clinicopathological features in colorectal cancer. Int J Clin Exp Pathol. 2015;8(4):4044–53.

    PubMed  PubMed Central  Google Scholar 

  71. Kim YH, Min BH, Choi HK, Kim SJ, Kim KM, Kim JY, Chang DK, Son HJ, Rhee PL, Kim JJ, Rhee JC, Chun HK. Sporadic colorectal carcinomas with low-level microsatellite instability in Korea: do they form a distinct subgroup with distinguished clinicopathological features? J Surg Oncol. 2009;99(6):351–5.

    Article  CAS  PubMed  Google Scholar 

  72. Lin A, Zhang J, Luo P. Crosstalk between the MSI Status and Tumor Microenvironment in Colorectal Cancer. Front Immunol. 2020;11:2039.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Ogino S, Nosho K, Irahara N, Meyerhardt JA, Baba Y, Shima K, Glickman JN, Ferrone CR, Mino-Kenudson M, Tanaka N, Dranoff G, Giovannucci EL, Fuchs CS. Lymphocytic reaction to colorectal cancer is associated with longer survival, independent of lymph node count, microsatellite instability, and CpG island methylator phenotype. Clin cancer Research: Official J Am Association Cancer Res. 2009;15(20):6412–20.

    Article  CAS  Google Scholar 

  74. Jass JR. HNPCC and sporadic MSI-H colorectal cancer: a review of the morphological similarities and differences. Fam Cancer. 2004;3(2):93–100.

    Article  CAS  PubMed  Google Scholar 

  75. Nakayama Y, Iijima T, Inokuchi T, Kojika E, Takao M, Takao A, Koizumi K, Horiguchi SI, Hishima T, Yamaguchi T. Clinicopathological features of sporadic MSI colorectal cancer and Lynch syndrome: a single-center retrospective cohort study. Int J Clin Oncol. 2021;26(10):1881–9.

    Article  PubMed  Google Scholar 

  76. Liu GC, Liu RY, Yan JP, An X, Jiang W, Ling YH, Chen JW, Bei JX, Zuo XY, Cai MY, Liu ZX, Zuo ZX, Liu JH, Pan ZZ, Ding PR. The heterogeneity between Lynch-Associated and sporadic MMR Deficiency in Colorectal cancers. J Natl Cancer Inst. 2018;110(9):975–84.

    Article  PubMed  Google Scholar 

  77. Ramchander NC, Ryan NAJ, Walker TDJ, Harries L, Bolton J, Bosse T, Evans DG, Crosbie EJ. Distinct immunological landscapes characterize inherited and sporadic mismatch repair deficient endometrial Cancer. Front Immunol. 2019;10:3023.

    Article  CAS  PubMed  Google Scholar 

  78. Yoshino T, Andre T, Kim TW, Yong WP, Shiu KK, Jensen BV, Jensen LH, Punt CJA, Smith D, Garcia-Carbonero R, Alcaide-Garcia J, Gibbs P, de la Fouchardiere C, Rivera F, Elez E, Le DT, Adachi N, Fogelman D, Marinello P, Diaz LA. Jr., Pembrolizumab in Asian patients with microsatellite-instability-high/mismatch-repair-deficient colorectal cancer. Cancer Sci. 2023;114(3):1026–36.

    Article  CAS  PubMed  Google Scholar 

  79. Boccaccino A, Borelli B, Intini R, Antista M, Bensi M, Rossini D, Passardi A, Tamberi S, Giampieri R, Antonuzzo L, Noto L, Roviello G, Zichi C, Salati M, Puccini A, Noto C, Parisi A, Rihawi K, Persano M, Crespi V, Libertini M, Giordano M, Moretto R, Lonardi S, Cremolini C. Encorafenib plus Cetuximab with or without binimetinib in patients with BRAF V600E-mutated metastatic colorectal cancer: real-life data from an Italian multicenter experience. ESMO open. 2022;7(3):100506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Chalabi M, Fanchi LF, Dijkstra KK, Van den Berg JG, Aalbers AG, Sikorska K, Lopez-Yurda M, Grootscholten C, Beets GL, Snaebjornsson P, Maas M, Mertz M, Veninga V, Bounova G, Broeks A, Beets-Tan RG, de Wijkerslooth TR, van Lent AU, Marsman HA, Nuijten E, Kok NF, Kuiper M, Verbeek WH, Kok M, Van Leerdam ME, Schumacher TN, Voest EE, Haanen JB. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat Med. 2020;26(4):566–76.

    Article  CAS  PubMed  Google Scholar 

  81. Hu H, Kang L, Zhang J, Wu Z, Wang H, Huang M, Lan P, Wu X, Wang C, Cao W, Hu J, Huang Y, Huang L, Wang H, Shi L, Cai Y, Shen C, Ling J, Xie X, Cai Y, He X, Dou R, Zhou J, Ma T, Zhang X, Luo S, Deng W, Ling L, Liu H, Deng Y. Neoadjuvant PD-1 blockade with toripalimab, with or without celecoxib, in mismatch repair-deficient or microsatellite instability-high, locally advanced, colorectal cancer (PICC): a single-centre, parallel-group, non-comparative, randomised, phase 2 trial. The lancet. Gastroenterology & hepatology 2022, 7 (1), 38–48.

  82. Chen G, Jin Y, Guan WL, Zhang RX, Xiao WW, Cai PQ, Liu M, Lin JZ, Wang FL, Li C, Quan TT, Xi SY, Zhang HZ, Pan ZZ, Wang F, Xu RH. Neoadjuvant PD-1 blockade with sintilimab in mismatch-repair deficient, locally advanced rectal cancer: an open-label, single-centre phase 2 study. Lancet Gastroenterol Hepatol. 2023;8(5):422–31.

    Article  PubMed  Google Scholar 

  83. André T, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt C, Smith D, Garcia-Carbonero R, Benavides M, Gibbs P, de la Fouchardiere C, Rivera F, Elez E, Bendell J, Le DT, Yoshino T, Van Cutsem E, Yang P, Farooqui MZH, Marinello P, Diaz LA. Jr., Pembrolizumab in microsatellite-instability-high Advanced Colorectal Cancer. N Engl J Med. 2020;383(23):2207–18.

    Article  PubMed  Google Scholar 

  84. Chalabi M, Verschoor YL, Tan PB, Balduzzi S, Van Lent AU, Grootscholten C, Dokter S, Büller NV, Grotenhuis BA, Kuhlmann K, Burger JW, Huibregtse IL, Aukema TS, Hendriks ER, Oosterling SJ, Snaebjornsson P, Voest EE, Wessels LF, Beets-Tan RG, Van Leerdam ME, Schumacher TN, van den Berg JG, Beets GL, Haanen JB. Neoadjuvant immunotherapy in locally advanced mismatch repair-deficient Colon cancer. N Engl J Med. 2024;390(21):1949–58.

    Article  CAS  PubMed  Google Scholar 

  85. Verschoor YL, Berg Jvd, Beets G, Sikorska K, Aalbers A, Lent Av, Grootscholten C, Huibregtse I, Marsman H, Oosterling S, Belt M v. d.;, Kok M, Schumacher T, Leerdam MEv, Haanen JBAG, Voest EE, Chalabi M. Neoadjuvant nivolumab, ipilimumab, and celecoxib in MMR-proficient and MMR-deficient colon cancers: Final clinical analysis of the NICHE study. Journal of Clinical Oncology 2022, 40 (16_suppl), 3511–3511.

  86. Verschoor YL, van den Berg J, Balduzzi S, van Blijderveen JC, Oosterling S, Burger P. T. Aukema., LBA31 neoadjuvant nivolumab plus relatlimab (anti-LAG3) in locally advanced MMR-deficient colon cancers: the NICHE-3 study.

  87. Ludford K, Ho WJ, Thomas JV, Raghav KPS, Murphy MB, Fleming ND, Lee MS, Smaglo BG, You YN, Tillman MM, Kamiya-Matsuoka C, Thirumurthi S, Messick C, Johnson B, Vilar E, Dasari A, Shin S, Hernandez A, Yuan X, Yang H, Foo WC, Qiao W, Maru D, Kopetz S, Overman MJ. Neoadjuvant Pembrolizumab in localized microsatellite instability High/Deficient mismatch repair solid tumors. J Clin Oncology: Official J Am Soc Clin Oncol. 2023;41(12):2181–90.

    Article  CAS  Google Scholar 

  88. Cercek A, Lumish M, Sinopoli J, Weiss J, Shia J, Lamendola-Essel M, El Dika IH, Segal N, Shcherba M, Sugarman R, Stadler Z, Yaeger R, Smith JJ, Rousseau B, Argiles G, Patel M, Desai A, Saltz LB, Widmar M, Iyer K, Zhang J, Gianino N, Crane C, Romesser PB, Pappou EP, Paty P, Garcia-Aguilar J, Gonen M, Gollub M, Weiser MR, Schalper KA, Diaz LA Jr. PD-1 blockade in Mismatch Repair-Deficient, locally advanced rectal Cancer. N Engl J Med. 2022;386(25):2363–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. André T, Berton D, Curigliano G, Sabatier R, Tinker AV, Oaknin A, Ellard S, de Braud F, Arkenau HT, Trigo J, Gravina A, Kristeleit R, Moreno V, Abdeddaim C, Vano YA, Samouëlian V, Miller R, Boni V, Torres AA, Gilbert L, Brown J, Dewal N, Dabrowski C, Antony G, Zografos E, Veneris J, Banerjee S. Antitumor Activity and Safety of Dostarlimab Monotherapy in patients with mismatch repair deficient solid tumors: a Nonrandomized Controlled Trial. JAMA Netw open 2023, 6 (11), e2341165.

  90. Xu R-H, Wang F, Chen G, Qiu M, Ma J, Liu H, Mo X, Li Y, Wan X, Luo J, Hu Y, Huang X, Jiang W, Li L, Zhou Y, Lu X, Chen Y, Guo Q, Li L, Zhou H. Neoadjuvant treatment of IBI310 (anti-CTLA-4 antibody) plus sintilimab (anti-PD-1 antibody) in patients with microsatellite instability-high/mismatch repair-deficient colorectal cancer: results from a randomized, open-labeled, phase ib study. J Clin Oncol. 2024;42(16suppl):3505–3505.

    Article  Google Scholar 

  91. Ding K-F, Chen G, Ye Y, Liu Y, Sun Y, Gao Y, Zhang J, Wei S, Li C, Li D, Zhou Y, Liu M, Zhang J, Zhang L, Kong D. Preoperative (neoadjuvant) therapy with tislelizumab for locally advanced colorectal cancer with high microsatellite instability or deficient mismatch repair: an open-label, single-arm, multicenter phase II study. J Clin Oncol. 2024;42(16suppl):3599–3599.

    Article  Google Scholar 

  92. Venderbosch S, Nagtegaal ID, Maughan TS, Smith CG, Cheadle JP, Fisher D, Kaplan R, Quirke P, Seymour MT, Richman SD, Meijer GA, Ylstra B, Heideman DA, de Haan AF, Punt CJ, Koopman M. Mismatch repair status and BRAF mutation status in metastatic colorectal cancer patients: a pooled analysis of the CAIRO, CAIRO2, COIN, and FOCUS studies. Clin cancer Research: Official J Am Association Cancer Res. 2014;20(20):5322–30.

    Article  CAS  Google Scholar 

  93. Casak SJ, Marcus L, Fashoyin-Aje L, Mushti SL, Cheng J, Shen YL, Pierce WF, Her L, Goldberg KB, Theoret MR, Kluetz PG, Pazdur R, Lemery SJ. FDA approval Summary: Pembrolizumab for the first-line treatment of patients with MSI-H/dMMR Advanced Unresectable or metastatic colorectal carcinoma. Clin cancer Research: Official J Am Association Cancer Res. 2021;27(17):4680–4.

    Article  CAS  Google Scholar 

  94. Diaz LA, Shiu J, Kim KK, Jensen TW, Jensen BV, Punt LH, Smith C, Garcia-Carbonero D, Benavides R, Gibbs M, de la Fourchardiere P, Rivera C, Elez F, Le E, Yoshino DT, Zhong T, Fogelman WY, Marinello D, Andre P. Pembrolizumab versus chemotherapy for microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer (KEYNOTE-177): final analysis of a randomised, open-label, phase 3 study. Lancet Oncol. 2022;23(5):659–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Grothey A. Pembrolizumab in MSI-H-dMMR Advanced Colorectal Cancer - A New Standard of Care. N Engl J Med. 2020;383(23):2283–5.

    Article  CAS  PubMed  Google Scholar 

  96. Lenz HJ, Van Cutsem E, Luisa Limon M, Wong KYM, Hendlisz A, Aglietta M, García-Alfonso P, Neyns B, Luppi G, Cardin DB, Dragovich T, Shah U, Abdullaev S, Gricar J, Ledeine JM, Overman MJ, Lonardi S. Colorectal Cancer: the phase II CheckMate 142 study. J Clin Oncology: Official J Am Soc Clin Oncol. 2022;40(2):161–70. First-Line Nivolumab Plus Low-Dose Ipilimumab for Microsatellite Instability-High/Mismatch Repair-Deficient Metastatic.

    Article  CAS  Google Scholar 

  97. André T, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, Morse MA, Van Cutsem E, McDermott R, Hill A, Sawyer MB, Hendlisz A, Neyns B, Abdullaev S, Memaj A, Lei M, Dixon M, Kopetz S, Overman MJ. Nivolumab plus low-dose ipilimumab in previously treated patients with microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: 4-year follow-up from CheckMate 142. Annals Oncology: Official J Eur Soc Med Oncol. 2022;33(10):1052–60.

    Article  Google Scholar 

  98. Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, Morse MA, Van Cutsem E, McDermott R, Hill A, Sawyer MB, Hendlisz A, Neyns B, Svrcek M, Moss RA, Ledeine JM, Cao ZA, Kamble S, Kopetz S, André T. Durable clinical benefit with Nivolumab Plus Ipilimumab in DNA mismatch Repair-Deficient/Microsatellite instability-high metastatic colorectal Cancer. J Clin Oncology: Official J Am Soc Clin Oncol. 2018;36(8):773–9.

    Article  CAS  Google Scholar 

  99. Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, Diaz LA Jr. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat Rev Gastroenterol Hepatol. 2019;16(6):361–75.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, Biedrzycki B, Donehower RC, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Duffy SM, Goldberg RM, de la Chapelle A, Koshiji M, Bhaijee F, Huebner T, Hruban RH, Wood LD, Cuka N, Pardoll DM, Papadopoulos N, Kinzler KW, Zhou S, Cornish TC, Taube JM, Anders RA, Eshleman JR, Vogelstein B, Diaz LA Jr. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. The New England journal of medicine 2015, 372 (26), 2509-20.

  101. Le DT, Kim TW, Van Cutsem E, Geva R, Jäger D, Hara H, Burge M, O’Neil B, Kavan P, Yoshino T, Guimbaud R, Taniguchi H, Elez E, Al-Batran SE, Boland PM, Crocenzi T, Atreya CE, Cui Y, Dai T, Marinello P, Diaz LA Jr., André T. Phase II open-label study of Pembrolizumab in Treatment-Refractory, microsatellite Instability-High/Mismatch repair-deficient metastatic colorectal Cancer: KEYNOTE-164. J Clin Oncology: Official J Am Soc Clin Oncol. 2020;38(1):11–9.

    Article  CAS  Google Scholar 

  102. Le DT, Diaz LA Jr., Kim TW, Van Cutsem E, Geva R, Jäger D, Hara H, Burge M, O’Neil BH, Kavan P, Yoshino T, Guimbaud R, Taniguchi H, Élez E, Al-Batran SE, Boland PM, Cui Y, Leconte P, Marinello P, André T. Pembrolizumab for previously treated, microsatellite instability-high/mismatch repair-deficient advanced colorectal cancer: final analysis of KEYNOTE-164. European journal of cancer (Oxford, England: 1990) 2023, 186, 185–195.

  103. Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, Desai J, Hill A, Axelson M, Moss RA, Goldberg MV, Cao ZA, Ledeine JM, Maglinte GA, Kopetz S, André T. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol. 2017;18(9):1182–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Taïeb J, André T, El Hajbi F, Barbier E, Toullec C, Kim S, Bouche O, Di Fiore F, Chauvenet M, Perrier H, Evesque L, Laurent-Puig P, Emile JF, Bez J, Lepage C, Tougeron D. Avelumab versus standard second line treatment chemotherapy in metastatic colorectal cancer patients with microsatellite instability: the SAMCO-PRODIGE 54 randomised phase II trial. Dig Liver Disease: Official J Italian Soc Gastroenterol Italian Association Study Liver. 2021;53(3):318–23.

    Article  Google Scholar 

  105. Li J, Deng Y, Zhang W, Zhou AP, Guo W, Yang J, Yuan Y, Zhu L, Qin S, Xiang S, Lu H, Gong J, Xu T, Liu D, Shen L. Subcutaneous envafolimab monotherapy in patients with advanced defective mismatch repair/microsatellite instability high solid tumors. J Hematol Oncol. 2021;14(1):95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Qin S, Li J, Zhong H, Jin C, Chen L, Yuan X, Fan Q, Chen K, Cao P, Xiao J, Jiang D, Zhang T, Zhang H, Wang X, Wang W, Han L, Wang Q, Zhu J. Serplulimab, a novel anti-PD-1 antibody, in patients with microsatellite instability-high solid tumours: an open-label, single-arm, multicentre, phase II trial. Br J Cancer. 2022;127(12):2241–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Picco G, Cattaneo CM, van Vliet EJ, Crisafulli G, Rospo G, Consonni S, Vieira SF, Rodríguez IS, Cancelliere C, Banerjee R, Schipper LJ, Oddo D, Dijkstra KK, Cinatl J, Michaelis M, Yang F, Di Nicolantonio F, Sartore-Bianchi A, Siena S, Arena S, Voest EE, Bardelli A, Garnett MJ. Werner Helicase is a Synthetic-Lethal vulnerability in Mismatch Repair-Deficient Colorectal Cancer Refractory to targeted therapies, Chemotherapy, and Immunotherapy. Cancer Discov. 2021;11(8):1923–37.

    Article  CAS  PubMed  Google Scholar 

  108. Zimmer K, Puccini A, Xiu J, Baca Y, Spizzo G, Lenz HJ, Battaglin F, Goldberg RM, Grothey A, Shields AF, Salem ME, Marshall JL, Korn WM, Wolf D, Kocher F, Seeber A. WRN-Mutated Colorectal Cancer is characterized by a distinct genetic phenotype. Cancers 2020, 12 (5).

  109. Baltgalvis KA, Lamb KN, Symons KT, Wu CC, Hoffman MA, Snead AN, Song X, Glaza T, Kikuchi S, Green JC, Rogness DC, Lam B, Rodriguez-Aguirre ME, Woody DR, Eissler CL, Rodiles S, Negron SM, Bernard SM, Tran E, Pollock J, Tabatabaei A, Contreras V, Williams HN, Pastuszka MK, Sigler JJ, Pettazzoni P, Rudolph MG, Classen M, Brugger D, Claiborne C, Plancher JM, Cuartas I, Seoane J, Burgess LE, Abraham RT, Weinstein DS, Simon GM, Patricelli MP, Kinsella TM. Chemoproteomic discovery of a covalent allosteric inhibitor of WRN helicase. Nature. 2024;629(8011):435–42.

    Article  CAS  PubMed  Google Scholar 

  110. Ferretti S, Hamon J, de Kanter R, Scheufler C, Andraos-Rey R, Barbe S, Bechter E, Blank J, Bordas V, Dammassa E, Decker A, Di Nanni N, Dourdoigne M, Gavioli E, Hattenberger M, Heuser A, Hemmerlin C, Hinrichs J, Kerr G, Laborde L, Jaco I, Núñez EJ, Martus HJ, Quadt C, Reschke M, Romanet V, Schaeffer F, Schoepfer J, Schrapp M, Strang R, Voshol H, Wartmann M, Welly S, Zécri F, Hofmann F, Möbitz H, Cortés-Cros M. Discovery of WRN inhibitor HRO761 with synthetic lethality in MSI cancers. Nature. 2024;629(8011):443–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Wang Y, Wiesnoski DH, Helmink BA, Gopalakrishnan V, Choi K, DuPont HL, Jiang ZD, Abu-Sbeih H, Sanchez CA, Chang CC, Parra ER, Francisco-Cruz A, Raju GS, Stroehlein JR, Campbell MT, Gao J, Subudhi SK, Maru DM, Blando JM, Lazar AJ, Allison JP, Sharma P, Tetzlaff MT, Wargo JA, Jenq RR. Fecal microbiota transplantation for refractory immune checkpoint inhibitor-associated colitis. Nat Med. 2018;24(12):1804–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Zhao W, Lei J, Ke S, Chen Y, Xiao J, Tang Z, Wang L, Ren Y, Alnaggar M, Qiu H, Shi W, Yin L, Chen Y. Fecal microbiota transplantation plus tislelizumab and fruquintinib in refractory microsatellite stable metastatic colorectal cancer: an open-label, single-arm, phase II trial (RENMIN-215). EClinicalMedicine 2023, 66, 102315.

  113. Bai X, Duan Z, Deng J, Zhang Z, Fu R, Zhu C, Fan D. Ginsenoside Rh4 inhibits colorectal cancer via the modulation of gut microbiota-mediated bile acid metabolism. J Adv Res 2024.

  114. de Carvalho AC, de Mattos Pereira L, Datorre JG, Dos Santos W, Berardinelli GN, Matsushita MM, Oliveira MA, Durães RO, Guimarães DP, Reis RM. Microbiota Profile and Impact of Fusobacterium nucleatum in Colorectal Cancer patients of Barretos Cancer Hospital. Front Oncol. 2019;9:813.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Zepeda-Rivera M, Minot SS, Bouzek H, Wu H, Blanco-Míguez A, Manghi P, Jones DS, LaCourse KD, Wu Y, McMahon EF, Park SN, Lim YK, Kempchinsky AG, Willis AD, Cotton SL, Yost SC, Sicinska E, Kook JK, Dewhirst FE, Segata N, Bullman S, Johnston CD. A distinct Fusobacterium nucleatum clade dominates the colorectal cancer niche. Nature. 2024;628(8007):424–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Taieb J, Sinicrope FA, Pederson L, Lonardi S, Alberts SR, George TJ, Yothers G, Van Cutsem E, Saltz L, Ogino S, Kerr R, Yoshino T, Goldberg RM, André T, Laurent-Puig P, Shi Q. Different prognostic values of KRAS exon 2 submutations and BRAF V600E mutation in microsatellite stable (MSS) and unstable (MSI) stage III colon cancer: an ACCENT/IDEA pooled analysis of seven trials. Annals Oncology: Official J Eur Soc Med Oncol. 2023;34(11):1025–34.

    Article  CAS  Google Scholar 

  117. Colle R, Lonardi S, Cachanado M, Overman MJ, Elez E, Fakih M, Corti F, Jayachandran P, Svrcek M, Dardenne A, Cervantes B, Duval A, Cohen R, Pietrantonio F, André T. BRAF V600E/RAS mutations and Lynch Syndrome in patients with MSI-H/dMMR metastatic colorectal Cancer treated with Immune Checkpoint inhibitors. Oncologist. 2023;28(9):771–9.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Taieb J, Svrcek M, Cohen R, Basile D, Tougeron D, Phelip JM. Deficient mismatch repair/microsatellite unstable colorectal cancer: Diagnosis, prognosis and treatment. European journal of cancer (Oxford, England: 1990) 2022, 175, 136–157.

Download references

Acknowledgements

We appreciate Dr. Qiao Bin’s critical reading of the article.

Funding

This study was funded by the Shanxi Province “Four Batch” Program (2020XM39) to Zhao Jun.

Author information

Authors and Affiliations

Authors

Contributions

Design: W-XF, JZ. Manuscript writing: W-XF and JZ. Manuscript revision: W-XF, JZ, YZ, FS, X-LZ, Y-YD, W-LL, Y-JG, W-QH. Final approval of the manuscript: JZ. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Jun Zhao.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fan, WX., Su, F., Zhang, Y. et al. Oncological characteristics, treatments and prognostic outcomes in MMR-deficient colorectal cancer. Biomark Res 12, 89 (2024). https://doi.org/10.1186/s40364-024-00640-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40364-024-00640-7

Keywords