2TÜBİTAK Marmara Research Center, Climate Change and Life Sciences, Biotechnology Research Group, Kocaeli-Türkiye
3Department of Emergency Service, Dr. Vefa Tanır Ilgın City Hospital, Konya-Türkiye
4Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos-Switzerland
5Department of Medical Biology and Genetics, İstanbul Health and Technology University Faculty of Medicine, İstanbul-Türkiye DOI : 10.5505/tjo.2024.4477
Summary
Colorectal cancer (CRC) is a multifactorial disease arising from the complex interplay between genetic predisposition and environmental influences. Despite its widespread incidence globally, CRC demonstrates favorable prognostic outcomes when detected at an early stage. Microsatellite instability (MSI) is a common molecular abnormality associated with colorectal tumorigenesis. This review provides a comprehensive analysis of the molecular mechanisms, clinical implications, therapeutic approaches, and immune system interactions in MSI-associated CRC (MSI-CRC). Molecular changes include DNA slippage, dysfunction in the DNA mismatch repair (dMMR) system, and genetic/epigenetic factors that contribute to MSI. Clinically, MSI-CRC is characterized by distinct phenotypic features, including associations with Lynch syndrome, specific diagnostic methodologies, and prognostic relevance. The therapeutic landscape highlights the promising efficacy of immunotherapies and targeted treatments, particularly in dMMR?MSI-H-CRC. Immune dynamics within the tumor microenvironment (TME) reveal patterns of immune infiltration, immune evasion strategies, and opportunities to enhance the effectiveness of immunotherapy. Understanding these interrelated aspects is critical for developing tailored therapeutic strategies and improving patient outcomes in MSI-CRC.Introduction
Colorectal cancer (CRC) presents a significant global public health burden, ranking as the third most common cancer and the second leading cause of cancer-related mortality.[1] The complexity of CRC stems from its diverse molecular pathways and clinical heterogeneity. Among the key factors driving CRC pathogenesis, microsatellite instability (MSI) has emerged as a crucial biomarker, offering profound insights into tumorigenesis and therapeutic strategies. MSI results from defects in the DNA mismatch repair (MMR) system and disrupts the fidelity of DNA replication, leading to widespread genomic instability and the accumulation of mutations across the genome, resulting in a mutator phenotype. These mutations can target critical cancerrelated genes, leading to the activation of oncogenic pathways and promoting tumorigenesis.[2] Moreover, MSI is a hallmark of approximately 80% of Lynch syndrome cases and is also observed in 20% of sporadicx CRC cases, highlighting its importance in both hereditary and sporadic forms of the disease. MSI is further prevalent in around 20% of early-stage sporadic CRC cases (stages I and II) and 12% in stage III disease. Conversely, its incidence is lower in metastatic CRC settings, where it is found in only 4-5% of cases.[3,4]In 1993, Aaltonen et al.[5] first described MSI in Lynch syndrome, utilizing an arbitrarily primed PCR (AP-PCR) approach. Subsequent studies have highlighted MSI's prevalence in CRC, particularly in endometrial, gastric, breast, prostate, and bladder cancers. With the FDA's approval of immune checkpoint inhibitors for dMMR-MSI-H advanced cancers, MSI has gained attention as a key molecular signature, driving interest in its diagnostic and therapeutic potential. Clinically, MSI is associated with a favorable prognosis in CRC, as patients with MSI-high tumors generally experience better outcomes compared to those with microsatellite-stable (MSS) tumors.[6] MSI tumors tend to be less responsive to conventional chemotherapy but demonstrate heightened sensitivity to immune checkpoint inhibitors targeting PD-1, offering a promising therapeutic avenue.[7]
Looking ahead, the incorporation of MSI testing into clinical practice requires rigorous validation, including assessments of analytical validity, clinical validity, and clinical utility.[8] Current methods for detecting MSI, such as immunohistochemistry (IHC) and polymerase chain reaction (PCR), are well-established, while emerging techniques like next-generation sequencing (NGS) and circulating tumor DNA (ctDNA) analysis offer non-invasive alternatives with potential clinical relevance.[9,10]
Further research is necessary to fully elucidate the clinical implications of MSI in CRC, particularly regarding its role in Lynch syndrome screening, prognostic stratification, and therapeutic decision-making. Understanding the complex interplay between MSI and CRC biology may pave the way for more personalized treatment approaches, particularly in leveraging immune interactions within the tumor microenvironment (TME) for therapeutic benefit.
Molecular Mechanisms of MSI in CRC
DNA Slippage and Mismatch Repair (MMR) System
Dysfunction
The integrity of genomic stability hinges upon the intricate
machinery of DNA mismatch repair (MMR),
a system designed to rectify errors that arise during
DNA replication.[11] MMR operates through a meticulous
process, represented in Figure 1, involving the detection and correction of base mismatches and
small loops, thereby ensuring replication fidelity and
preventing the accumulation of mutations.[12] Crucial
to this process are key components of the MMR pathway,
notably MutS and MutL complexes. In human
cells, MutSα and MutSβ complexes, comprising various
combinations of MSH proteins, detect mismatches and
short loops, while MutLα, MutLβ, and MutLγ, formed
by different pairs of MLH proteins, orchestrate the subsequent
repair process.[13] However, when the MMR
system malfunctions, the consequences can be dire,
leading to genomic instability and predisposition to
cancer. Deficiencies in essential MMR proteins, particularly
MSH2 and MLH1, result in severe phenotypes,
underscoring the indispensability of these factors in
maintaining genomic integrity.[14]
Fig. 1. Diagram of Mismatch Repair (MMR) pathway.
Genetic and Epigenetic Factors Contributing to
MSI
The three major molecular pathways of colorectal cancer
(CRC) are the conventional chromosomal instability
(CIN) pathway, the serrated pathway, and the
microsatellite instability (MSI) pathway (Table 1). The
CIN pathway is initiated by APC mutation, followed by
mutations in KRAS, PIK3CA, and SMAD4, and loss of
heterozygosity of TP53, commonly associated with low
CpG island methylation, high CIN, and microsatellite
stability.[15] The serrated pathway can be subdivided
into CIMP-low-MSS tumors with KRAS mutations,
BRAF mutant CIMP-high-MSS tumors, and BRAF
mutant CIMP-high-MSI tumors, often involving the silencing
of MGMT, CDKN2A, or MLH1. Lastly, the MSI
pathway results from the dysfunction of DNA mismatch
repair genes encoding MLH or MSH proteins,
leading to high levels of microsatellite instability.[16]
Table 1 The three major molecular pathways of colorectal cancer
Genetic factors play a significant role in the development of MSI in colorectal cancer. Lynch syndrome (OMIM#120435), which is caused by mono-allelic germline MMR pathogenic variants, stemming from mutations in MMR genes such as MLH1, MSH2, MSH6, and PMS2, stands as a prominent genetic cause of MSI. [17] Specifically, inactivation of the MLH1 or PMS2 alleles is the most frequent cause of Lynch syndrome and is associated with approximately 80% of cases. [17] Mutations in MMR genes associated with Lynch syndrome frequently precipitate the onset of cancer at earlier ages, typically between 40 and 60 years. These mutations are implicated in a diverse spectrum of cancers, predominantly found in the gastrointestinal and genitourinary tracts, most frequently colorectal cancer and endometrial cancer.[3]
On the other hand, constitutional mismatch repair deficiency (CMMRD), a rare condition compared to Lynch syndrome, is caused by bi-allelic germline pathogenic variants in MMR genes.[18] It has an estimated birth incidence of one in a million. Typically, CMMRD manifests with hematological, brain, and intestinal cancers during childhood or adolescence, with a median age of onset under 10 years.[19] MSI-H tumors frequently harbor mutations in key CRC-related genes like APC, KRAS, and TP53, signifying a convergence of genetic alterations in these malignancies.[20] Additionally, RNF43 mutations, a negative feedback regulator of the Wnt/β-catenin pathway, have been observed in CRCs with a high prevalence of MSI-H, suggesting their potential involvement in MSI development.[21]
On the other hand, epigenetic factors also play a significant role in MSI, where the CpG island methylator phenotype (CIMP) is characterized by hypermethylation in tumor suppressor gene promoters, leading to functional loss.[22] A recently identified subgroup of patients with MSI tumors is referred to as having "Lynchlike syndrome" (LLS). These patients were diagnosed with cancers related to Lynch syndrome, with their tumors showing an MSI phenotype and/or loss of MMR gene expression. Cases with loss of MLH1 expression were confirmed to lack MLH1 promoter hypermethylation. However, no germline mutations in MMR genes were detected. Compared to LS patients, individuals with LLS exhibit a lower standardized incidence ratio of LS-related tumors. The mechanisms behind LLS remain unclear, but it is known that acquired MMR deficiency accounts for a significant portion of these cases.[23,24]
CIMP-positive CRCs, often arising via a serrated pathway, exhibit frequent association with MSI-H tumors, emphasizing the interplay between genetic and epigenetic alterations in CRC development.[25]
Detecting MMR Dysfunction: Microsatellite Instability
Analysis
In clinical settings, the diagnosis of dysfunctional
MMR is often achieved by assessing microsatellite instability
(MSI), a hallmark of MMR failure.[26] MSI is
characterized by alterations in the lengths of microsatellite
loci, serving as an indirect indicator of MMR deficiency.
Typically, this is detected through PCR-based
assays targeting specific microsatellite repeats.[27] The
Bethesda panel, comprising mononucleotide and dinucleotide
repeats, is commonly employed for MSI assessment,
with instability at multiple loci indicative of
MSI-high status.[12] Currently, the standard diagnostic
methods for MSI and MMR deficiency include pentaplex
PCR-based methods, incorporating five mononucleotide
and quasi-monomorphic microsatellite regions
(such as BAT-25 and BAT-26), and MMR immunohistochemistry
on tumor tissue samples.[27] Advances in
alternative systems, such as next-generation sequencing
(NGS) and real-time PCR-based methods, offer improved
accuracy and efficiency for MSI detection.[28]
Notably, the development of liquid biopsy techniques allows for the determination of MSI from cellfree DNA (cfDNA) in plasma, facilitating non-invasive testing and real-time monitoring of disease progression. These assays are often developed based on whole-genome sequencing (WGS) or whole-exome sequencing (WES) data from tumor tissue samples, which can be refined to large and customized gene panels.[27] A wellvalidated example is the FDA-authorized Integrated Mutation Profiling of Actionable Cancer Targets (MSKIMPACT) gene panel, developed at the Memorial Sloan Kettering Cancer Center. Initially designed as a hybrid capture-based NGS assay for targeted deep sequencing of key cancer genes in formalin-fixed paraffin-embedded (FFPE) tumor specimens, this assay was used to study DNA from over 10,000 tumor specimens and patient-matched germline DNA from peripheral blood, identifying clinically relevant mutations and mutation signatures.[29] The panel has since expanded to interrogate 468 cancer-related genes, analyzing tumor-derived and matched germline DNA samples in a CLIA-certified laboratory. It is FDA-approved as a tumor profiling test to provide information on somatic alterations and MSI for use by qualified healthcare professionals.[30]
One significant advantage of these newer techniques is their ability to analyze multiple microsatellite loci simultaneously, enhancing the sensitivity and specificity of MSI detection. For instance, the Promega MSI analysis system employs a panel of five mononucleotide markers and two pentanucleotide markers to detect MSI with high precision. Additionally, the Idylla MSI assay, a fully automated PCR-based system, delivers rapid and reliable results with minimal hands-on time, providing automated interpretation of MSI status.[31] These advancements in MSI analysis not only facilitate the diagnosis of MMR dysfunction but also offer insights into the mutational landscape of tumors, guiding therapeutic strategies and prognostic assessments. The importance of MSI testing has increased in personalized medicine, helping identify patients who might benefit from specific treatments like immune checkpoint inhibitors and informing decisions about adjuvant chemotherapy.
Clinical Implications of MSI in CRC
Clinical Features and Prognosis of MSI-CRC
MSI-CRCs exhibit distinct clinical features and prognosis
compared to microsatellite stable (MSS) tumors. Patients
with MSI-CRCs tend to belong to specific demographic
groups, such as females and the elderly, and are commonly
located in the proximal colon.[32] Previous studies indicated
that colorectal cancer (CRC) with microsatellite
instability (MSI) in elderly patients (aged 60?70 years and
over 87 years) is often linked to MLH1 inactivation and
MLH1 promoter methylation.[33] In contrast, tumors in
younger patients (under 45 years) are typically associated
with MSH2 inactivation.[34] This study revealed that patients
aged 50 years and older showed a lower propensity
for having MSH2 and/or MSH6-MSI.[35] Notably,
CRC associated with MSH2-MSI commonly occurs at a
younger age.[36] MSI-CRCs are also associated with mucinous
histology and poor differentiation (Table 2).[37]
Table 2 Clinical features and prognosis of MSI-CRC
Although MSI-CRCs typically exhibit aggressive histopathological features, they often show a lower response to 5-fluorouracil (5-FU) chemotherapy compared to MSS tumors,[6] which has led to the exploration of other therapeutic avenues for these patients. Later it was discovered that MSI-H CRCs are notably more responsive to immune checkpoint inhibitors, such as pembrolizumab or other PD-1 inhibitors, compared to their MSS counterparts. Despite their aggressive histopathological features, MSI-CRCs have been linked to better overall prognosis, particularly in early-stage disease.[38] This improved prognosis is attributed to the enhanced immune response against the high number of neoantigens present in MSI-H tumors, leading to an immunogenic tumor microenvironment.[39] Importantly, MSI status serves as a predictive biomarker for response to immunotherapy, which has resulted in FDA approval of pembrolizumab and nivolumab for treatments targeting MSI-H and MMR-deficient CRCs.[40]
Immune Interactions in the Tumor Microenvironment
Role of Immune Infiltration in MSI-CRC
In colorectal cancer, the tumor microenvironment
(TME) orchestrates disease progression and therapeutic
responses. Particularly in MSI-H CRCs, the TME is
characterized by dense immune infiltration owing to the
high mutational burden resulting from somatic hypermethylation
of MMR genes.[41] This leads to the generation
of numerous immunogenic neopeptide antigens,
attracting various immune cell populations, including
CD8+ T cells, B cells, and macrophages. This influx creates
an immunologically active milieu that influences
disease behavior and therapeutic outcomes.[42] Recent
studies have uncovered significant heterogeneity within
MSI-H CRCs. While MSI-H tumors generally exhibit
better overall survival compared to MSS CRCs, subgroups
within MSI-H CRCs demonstrate distinct immune
profiles and clinical behaviors.[43]
The molecular classifications primarily designed to predict colorectal cancer prognosis and recurrence risk have been proposed. The most well-known of these is the "consensus molecular subtype (CMS) classification," which has enhanced the understanding of the genomic and epigenomic landscapes of colorectal cancer, aiding in better patient management. This classification divides colorectal cancer into four CMS categories (CMS1-4), each with different prognoses. This manuscript places the CMS classification in various contexts, exploring its relationships with precursor lesions, tumor immunophenotype, and gut microbiota. It also examines the CMS classification's role in predicting prognosis and/or response to pharmacological treatments, marking a crucial step toward precision medicine.[44] Some MSI-H tumors display heightened T-cell activity, associated with improved prognosis (classified as CMS1 subtype), while others exhibit diminished T-cell activity and poorer prognosis (classified as MSI-H in CMS3 and CMS4 subtypes). The differential presence of specific immune cell subpopulations, such as memory CD4+/CD8+ cells, γδ T cells, and regulatory T cells, contributes to this heterogeneity, influencing overall immune dynamics and disease outcomes.[45,46]
Mechanisms of Immune Evasion and Checkpoint
Inhibitor Resistance
Despite robust immune infiltration in MSI-H CRCs,
certain tumors develop sophisticated mechanisms to
evade immune surveillance and resist checkpoint inhibitor
therapy. Common strategies involve alterations
in the human leukocyte antigen (HLA) complex and
antigen-processing machinery (APM), critical for effective
antigen presentation and T-cell recognition.[47]
Mutations in genes encoding components of the HLA
complex and APM machinery, such as beta-2-microglobulin
(B2M) and HSPA5, are prevalent in MSI-H
CRCs, impairing tumor antigen presentation, hampering
T-cell recognition, and enabling tumors to evade immune
detection and elimination.[48] Moreover, emerging
evidence implicates the gut microbiota, particularly
Fusobacterium nucleatum, in CRC pathogenesis and
treatment response. F. nucleatum, frequently associated with MSI-H CRCs, exerts profound effects on the TME
and immune response. It can induce pro-inflammatory
environments, foster immunosuppression, and inhibit
T-cell activity, thereby promoting tumor progression
and conferring resistance to immunotherapy.[49,50]
Therapeutic Landscape of MSI-CRC
Immunotherapy and Targeted Therapies for MSI-H
CRC
Immunotherapy has emerged as a promising approach
for the treatment of mismatch repair-deficient MSI-H
colorectal cancer (CRC). Initial studies exploring immune
checkpoint inhibitors (ICIs) in CRC demonstrated
limited clinical activity in unselected patients. However,
subsequent investigations focusing on dMMR?MSI-H
CRC revealed remarkable responses to PD-1 inhibitors.
The CheckMate 142 trial investigated nivolumab, a PD-1
inhibitor, in patients with dMMR?MSI-H metastatic
CRC. The study reported an objective response rate of
31% and a disease control rate of 69%, with promising
progression-free survival (PFS) and overall survival
outcomes.[51] In the phase II CheckMate 142 study,
nivolumab plus low-dose ipilimumab as first-line therapy
for patients with dMMR?MSI-H metastatic colorectal
cancer (mCRC) demonstrated a 69% objective response
rate and an 84% disease control rate, with a 13%
complete response rate. The treatment showed robust
and durable clinical benefit, with a median progressionfree
survival and median overall survival not reached
at the median follow-up of 29.0 months. Additionally,
74% of responders had ongoing responses at data cutoff,
and the treatment was well tolerated, with grade 3-4
treatment-related adverse events occurring in 22% of
patients.[52] Based on these promising results, further
randomized studies are warranted.
Similarly, in the phase III KEYNOTE 177 trial, pembrolizumab, an anti-PD-1 antibody, demonstrated superiority over chemotherapy in first-line treatment of dMMR?MSI-H mCRC, with significantly improved PFS and OS. The trial highlighted a 45.1% objective response rate in the pembrolizumab arm compared to 33.1% in the chemotherapy arm, leading to its approval as the new standard of care.[53]
Ongoing studies are evaluating PD-1 or PD-L1 inhibition in various settings, including first-line therapy, with the aim of further improving outcomes for patients with dMMR?MSI-H CRC. In another study, which aimed to evaluate the safety and efficacy of neoadjuvant PD-1 blockade immunotherapy with sintilimab, another PD-1 inhibitor, for locally advanced dMMR- MSI-H CRC, researchers focused on patients treated at the Sixth Affiliated Hospital of Sun Yat-sen University from June 2020 to June 2022, aiming to provide insights into the clinical and pathological responses to this treatment. By retrospectively analyzing the clinical data of 11 patients who received six injections of sintilimab before radical laparoscopic resection, the study sought to determine the pathological complete response (pCR) rates and the occurrence of immunotherapy-related adverse events. Their findings suggest that single-agent neoadjuvant PD-1 antibody immunotherapy could be a promising treatment approach for locally advanced dMMR?MSI-H CRC, warranting further validation in phase II and III clinical trials.[54]
In contrast, patients with proficient mismatch repair/ microsatellite stable (pMMR-MSI-L) CRC have not benefited significantly from immunotherapy alone. Limited responses were observed in patients with pMMR-MSI-L CRC treated with immune checkpoint inhibitors.[55]
The study investigated in 2023 showed the efficacy of combining PD-1, BRAF, and MEK inhibitors in treating BRAFV600E-CRC, given the limited success of BRAF inhibitor combinations alone. Conducted as a single-arm phase II trial, 37 patients received the PD-1 inhibitor spartalizumab and kinase inhibitors designed for BRAF V600E mutations, dabrafenib and trametinib. The primary endpoint, overall response rate (ORR), was met with a 24.3% response rate overall and 25% in MSS patients. Notably, MSI patients exhibited better and more durable responses, with approximately one-third showing responses lasting over a year. These results suggest that BRAF pathway inhibition may enhance the immune response in BRAFV600E CRC, particularly in MSI patients,[56] although further research is needed to optimize treatment strategies for this subtype.
FDA-Approved Treatments for MSI-H/MMR CRC
Patients
Recognizing the immunological landscape within
dMMR?MSI-H colorectal cancer tumors is crucial for
therapeutic success. These tumors are characterized by
elevated immune cell infiltration and upregulated immune
checkpoints such as PD-1, PD-L1, and CTLA4.
[55] This understanding has paved the way for the development
and FDA approval of pioneering immunotherapy
agents like pembrolizumab and nivolumab, significantly
reshaping treatment paradigms and providing
new hope for patients who were previously difficult to
treat. Key trials such as KEYNOTE-177 and Check-
Mate-142 have demonstrated remarkable improvements
in PFS and ORR with these agents, both as monotherapy
and in combination regimens.[51-53] Further more, neoadjuvant immunotherapy approaches show
promise for curative interventions in select cases, while
ongoing research into tumor-specific antigens, DNA
polymerase mutations (POLD1/POLE), and immune
evasion mechanisms seeks to refine treatment selection
and overcome resistance.[40,57-59] Biomarkers like
tumor mutational burden (TMB), immunoscore, and
PD-L1 expression are under scrutiny to tailor treatment
decisions and predict response to immunotherapy. The
identification of predictive markers and elucidation of
underlying mechanisms are vital for optimizing patient
outcomes in dMMR?MSI-H CRC.
Challenges and Future Directions in MSI-Targeted
Therapy
While the advent of FDA-approved treatments has
propelled MSI-targeted therapy into the spotlight, significant
challenges persist, necessitating a multifaceted
approach to optimize patient outcomes. One pressing
concern is the heterogeneity within dMMR-MSI-H
CRC, underscored by variations in treatment response
and resistance mechanisms.[60] Despite promising
clinical trial results, identifying robust predictive
biomarkers remains elusive. BRAF status and PD-L1
expression, once thought to hold predictive value, have
shown inconsistent correlations, highlighting the complex
interplay of molecular factors influencing treatment
response.[61,62] Additionally, the clinical history
of Lynch syndrome has emerged as a potential modifier
of treatment outcomes, emphasizing the need for
comprehensive patient stratification strategies.[63]
The management of immune-related adverse events (IRAEs) poses another challenge, particularly in patients with pre-existing autoimmune disorders. While immunotherapy offers unprecedented therapeutic potential, the risk of exacerbating autoimmune conditions necessitates vigilant monitoring and a multidisciplinary approach to IRAE management.[64] Looking ahead, elucidating alternative actionable genes and exploring combination therapies represent promising avenues for enhancing treatment efficacy and overcoming resistance. Molecular profiling, coupled with advances in gut microbiome modulation, holds potential for refining patient stratification and optimizing treatment selection in dMMR-MSI-H CRC.[65] Moreover, neoadjuvant immunotherapy strategies offer the possibility of curative interventions in select cases, highlighting the evolving landscape of MSI-targeted therapy beyond conventional approaches.[40] Harnessing the synergy between immunotherapy and other treatment modalities, such as targeted therapies and chemotherapy, presents an opportunity for personalized treatment regimens tailored to individual patient profiles. As research continues to unravel the intricacies of MSI-targeted therapy, collaboration across disciplines and innovative trial designs will be crucial for translating scientific insights into clinical practice.
Potential Strategies to Enhance Immunotherapy
Efficacy in MSI-CRC
To augment the efficacy of immunotherapy in MSI-H
CRCs, several innovative strategies have been proposed:
• Targeting Immune Evasion Mechanisms: Therapeutic interventions aimed at restoring HLA expression or enhancing antigen presentation hold promise for improving T-cell recognition and response to immunotherapy.[66]
• Modulating the Gut Microbiota: Interventions targeting specific microbial populations, such as F. nucleatum, have the potential to enhance treatment response by reshaping the TME and fostering antitumor immune responses.[67]
• Combination Therapies: Combinations of checkpoint inhibitors with agents targeting alternate immune checkpoints or pathways involved in immune evasion could synergistically overcome resistance mechanisms and bolster anti-tumor immunity.[68]
• Patient Stratification: Precision medicine approaches, including subtyping CRCs based on their genetic characteristics and immune landscape, may enable the identification of patients most likely to benefit from immunotherapy, guiding personalized treatment strategies for optimal outcomes.[69]
Recent Insights into Genetic Factors
Werner Syndrome Helicase as a Synthetic Lethal
Target in MSI-CRC
Recent studies have broadened our knowledge of the
genetic factors involved in the development of microsatellite
instability (MSI) in CRC, in addition to well-established
mechanisms such as DNA slippage and dysfunction
in the MMR system. Among these factors, the
WRN gene, encoding the Werner syndrome ATP-dependent
helicase, has emerged as a significant player.
[70] WRN, a multifunctional enzyme with helicase
and exonuclease activities, plays essential roles in multiple
cellular processes vital for genome stability, such
as DNA replication, transcription, DNA repair, and
telomere maintenance. Depletion of WRN leads to cell
cycle arrest, DNA damage, mitotic anomalies, chromosome
fragmentation, and apoptosis. The frequent loss
of heterozygosity at the WRN loci on chromosome 8p11.2-p12 in various cancers underscores its function
as a tumor suppressor gene.[71,72]
This factor is a synthetic lethal target in microsatellite unstable cancers. Synthetic lethality is a phenomenon where the simultaneous occurrence of two genetic events results in cell death, whereas each event alone does not have this effect.[73] WRN deficiency has been associated with the promotion of MSI-H phenotypes, exacerbating replication stress and DNA damage, particularly in cells already harboring MMR defects.[74] The concept of synthetic lethality in MSI-H CRC has gained traction, with emerging experimental data highlighting a synthetic lethal phenomenon driven by expansion mutations in numerous (TA)n dinucleotide repeats. The elongation of (TA)n repeats increases the likelihood of secondary DNA structure formation, requiring WRN intervention for resolution. However, in the absence of functional WRN helicase activity, these unresolved DNA structures impede the progression of DNA replication forks, resulting in substantial DNA damage (Fig. 2).[75]
In another investigation, it was discovered that WRN loss in MMR-deficient cells triggers DNA double- strand breaks (DSBs), leading to the activation of ATM and CHK2 signaling kinases. This activation induces the tumor suppressor p53 and the pro-apoptotic protein PUMA, thereby promoting mitochondria- mediated apoptosis. The study identifies PUMA as a crucial mediator of apoptosis following WRN loss. Specifically, inhibition of WRN results in the activation of PUMA, which is essential for inducing cell death in MMR-deficient CRC cells.[76] Moreover, in MMR-deficient cells, the genome relies more on alternative DNA repair mechanisms to avoid exceeding the tolerable mutation threshold, which would otherwise lead to cell death. Consequently, any loss of function in the WRN gene disrupts these compensatory repair mechanisms, such as homologous recombination (HR) and non-homologous end joining (NHEJ), ultimately causing programmed cell death in the cancerous cell.[77]
The discovery of WRN's role in MSI-H cancers has opened exciting avenues for targeted therapy. Researchers are developing specific WRN inhibitors and employing gene-editing techniques such as CRISPR-Cas9 to selectively kill MSI-H cancer cells while leaving healthy cells unharmed.[78-80] Understanding the interplay between WRN and MSI-H in CRC provides valuable insights into potential therapeutic targets and strategies for personalized treatment approaches.
Conclusion
The multifaceted landscape of microsatellite instability (MSI) in colorectal cancer (CRC) presents a rich tapestry of clinical implications, therapeutic opportunities, and challenges. Through this comprehensive review, we have explored the clinical features and prognostic significance of MSI CRCs, delved into the intricate mechanisms underlying immune interactions within the tumor microenvironment, and discussed the evolving therapeutic landscape of MSI-targeted therapy.MSI CRCs exhibit distinct clinical behaviors and responses to treatment, driven by their unique molecular characteristics and immune microenvironment. MSI-H tumors demonstrate heightened immune infiltration and favorable responses to immunotherapy. However, challenges such as tumor heterogeneity and immune evasion mechanisms underscore the need for precision medicine approaches and innovative therapeutic strategies.
The advent of immunotherapy, particularly immune checkpoint inhibitors, has revolutionized the treatment paradigm for MSI-H CRCs, offering newfound hope and improved outcomes for patients previously considered therapeutically challenging. FDAapproved agents like pembrolizumab and nivolumab have reshaped clinical practice, with ongoing research exploring novel combinations and neoadjuvant approaches to further enhance treatment efficacy.
Significant challenges remain, including the heterogeneity within MSI-H CRCs, the complexity of immune evasion mechanisms, and the management of immunerelated adverse events. Addressing these challenges will require collaborative efforts across disciplines, innovative trial designs, and the integration of emerging technologies and biomarkers into clinical practice.
As we navigate the complexities of MSI CRCs, fueled by advancements in understanding tumor biology and immunology, the future holds promise for personalized and precise therapeutic interventions tailored to individual patient profiles. By unraveling the intricacies of immune interactions in the TME and leveraging the power of targeted therapy, we can strive toward improved outcomes and a paradigm shift in CRC management.
In essence, the journey towards optimizing MSI-targeted therapy is multifaceted. However, with collaboration and innovation, we can continue to push the boundaries of possibility, reshaping the standard of care and improving the lives of patients affected by colorectal cancer.
Conflict of Interest: All authors declared no conflict of interest.
Use of AI for Writing Assistance: No AI technologies utilized.
Financial Support: Not applicable.
Peer-review: Externally peer-reviewed.
References
1) Siegel RL, Wagle NS, Cercek A, Smith RA, Jemal A.
Colorectal cancer statistics, 2023. CA Cancer J Clin
2023;73(3):233-54.
2) Gian LdA, Lorena B, Cinzia A, Gioacchino L, Federica
G, Francesca N. Microsatellite instability in colorectal
cancer. Acta Bio Med 2018;89(Suppl 9):97.
3) Roudko V, Cimen Bozkus C, Greenbaum B, Lucas A,
Samstein R, Bhardwaj N. Lynch syndrome and MSI-H
cancers: From mechanisms to "off-the-shelf " cancer
vaccines. Front Immunol 2021;12:757804.
4) Battaglin F, Naseem M, Lenz HJ, Salem ME. Microsatellite
instability in colorectal cancer: Overview
of its clinical significance and novel perspectives. Clin
Adv Hematol Oncol 2018;16(11):735.
5) Peltomaki P, Aaltonen LA, Sistonen P, et al. Genetic
mapping of a locus predisposing to human colorectal
cancer. Science. 1993;260(5109):810-812.
6) Kawakami H, Zaanan A, Sinicrope FA. Microsatellite
instability testing and its role in the management of colorectal
cancer. Curr Treat Options Oncol 2015;16:1-15.
7) Borelli B, Antoniotti C, Carullo M, Germani MM, Conca
V, Masi G. Immune-checkpoint inhibitors (ICIs) in
metastatic colorectal cancer (mCRC) patients beyond
microsatellite instability. Cancers 2022;14(20):4974.
8) Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van
Krieken J, Aderka D, et al. ESMO consensus guidelines
for the management of patients with metastatic
colorectal cancer. Ann Oncol 2016;27(8):1386-422.
9) Baudrin LG, Deleuze JF, How-Kit A. Molecular and
computational methods for the detection of microsatellite
instability in cancer. Front Oncol 2018;8:621.
10) Willis J, Lefterova MI, Artyomenko A, Kasi PM,
Nakamura Y, Mody K, et al. Validation of microsatellite
instability detection using a comprehensive plasma-based genotyping panel. Clin Cancer Res
2019;25(23):7035-45.
11) Li Z, Pearlman AH, Hsieh P. DNA mismatch repair and
the DNA damage response. DNA Repair 2016;38:94?101.
12) Baretti M, Le DT. DNA mismatch repair in cancer.
Pharmacol Ther 2018;189:45-62.
13) Zhang X, Li J. Era of universal testing of microsatellite
instability in colorectal cancer. World J Gastrointest
Oncol 2013;5(2):12.
14) Richman S. Deficient mismatch repair: Read all about
it. Int J Oncol 2015;47(4):1189-202.
15) Nguyen LH, Goel A, Chung DC. Pathways of colorectal
carcinogenesis. Gastroenterology 2020;158(2):291?302.
16) Huang Z, Yang M. Molecular network of colorectal
cancer and current therapeutic options. Front Oncol
2022;12:852927.
17) Peltomäki P, Nyström M, Mecklin JP, Seppälä TT.
Lynch syndrome genetics and clinical implications.
Gastroenterology 2023;164(5):783-99.
18) Tamura K, Kaneda M, Futagawa M, Takeshita M, Kim
S, Nakama M, et al. Genetic and genomic basis of the
mismatch repair system involved in Lynch syndrome.
Int J Clin Oncol 2019;24:999-1011.
19) Gallon R, Brekelmans C, Martin M, Bours V, Schamschula
E, Amberger A, et al. Constitutional mismatch
repair deficiency mimicking Lynch syndrome is associated
with hypomorphic mismatch repair gene variants.
NPJ Precis Oncol 2024;8(1):119.
20) Lin EI, Tseng LH, Gocke CD, Reil S, Le DT, Azad NS, et
al. Mutational profiling of colorectal cancers with microsatellite
instability. Oncotarget 2015;6(39):42334.
21) Elez E, Ros J, Fernández J, Villacampa G, Moreno-Cárdenas
AB, Arenillas C, et al. RNF43 mutations predict
response to anti-BRAF/EGFR combinatory therapies
in BRAF V600E metastatic colorectal cancer. Nat Med
2022;28(10):2162-70.
22) Mojarad EN, Kuppen PJ, Aghdaei HA, Zali MR. The
CpG island methylator phenotype (CIMP) in colorectal
cancer. Gastroenterol Hepatol Bed Bench
2013;6(3):120.
23) Thibodeau SN, French AJ, Cunningham JM, Tester D,
Burgart LJ, Roche PC, et al. Microsatellite instability
in colorectal cancer: Different mutator phenotypes
and the principal involvement of hMLH1. Cancer Res
1998;58(8):1713-8.
24) Porkka N, Lahtinen L, Ahtiainen M, Böhm JP, Kuopio
T, Eldfors S, et al. Epidemiological, clinical and molecular
characterization of Lynch-like syndrome: A population-
based study. Int J Cancer 2019;145(1):87-98.
25) Jasmine F, Haq Z, Kamal M, Raza M, da Silva G,
Gorospe K, et al. Interaction between microsatellite
instability (MSI) and tumor DNA methylation
in the pathogenesis of colorectal carcinoma. Cancers
2021;13(19):4956.
26) Suraweera N, Duval A, Reperant M, Vaury C, Furlan
D, Leroy K, et al. Evaluation of tumor microsatellite
instability using five quasimonomorphic mononucleotide
repeats and pentaplex PCR. Gastroenterology
2002;123(6):1804-11.
27) Gilson P, Merlin JL, Harlé A. Detection of microsatellite
instability: State of the art and future applications
in circulating tumour DNA (ctDNA). Cancers
2021;13(7):1491.
28) Tieng FYF, Abu N, Lee LH, Ab Mutalib NS. Microsatellite
instability in colorectal cancer liquid biopsy-current
updates on its potential in non-invasive detection,
prognosis and as a predictive marker. Diagnostics
2021;11(3):544.
29) Ignatiadis M, Sledge GW, Jeffrey SS. Liquid biopsy
enters the clinic-implementation issues and future
challenges. Nat Rev Clin Oncol 2021;18(5):297?312.
30) Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim
HR, et al. Mutational landscape of metastatic cancer
revealed from prospective clinical sequencing of
10,000 patients. Nat Med 2017;23(6):703-13.
31) Velasco A, Tokat F, Bonde J, Trim N, Bauer E, Meeney
A, et al. Multi-center real-world comparison of the
fully automated Idylla microsatellite instability assay
with routine molecular methods and immunohistochemistry
on formalin-fixed paraffin-embedded tissue
of colorectal cancer. Virchows Arch 2021;478:851-63.
32) Cesana N. A Comparative study regarding the morphologic
implications of microsatellite instability in
malignant colorectal neoplasia. Lietuvos Sveikatos
Mokslu Universitetas 2022.
33) Kim HG, Lee S, Kim DY, Ryu SY, Joo JK, Kim JC, et al.
Aberrant methylation of DNA mismatch repair genes
in elderly patients with sporadic gastric carcinoma:
A comparison with younger patients. J Surg Oncol
2010;101(1):28-35.
34) Salem ME, Bodor JN, Puccini A, Xiu J, Goldberg RM,
Grothey A, et al. Relationship between MLH1, PMS2,
MSH2 and MSH6 gene-specific alterations and tumor
mutational burden in 1057 microsatellite instabilityhigh
solid tumors. Int J Cancer 2020;147(10):2948-56.
35) Yiu R, Qiu H, Lee SH, García-Aguilar J. Mechanisms
of microsatellite instability in colorectal cancer patients
in different age groups. Dis Colon Rectum
2005;48:2061-9.
36) Coggins R, Cawkwell L, Bell S, Crockford G, Quirke
P, Finan P, et al. Association between family history
and mismatch repair in colorectal cancer. Gut
2005;54(5):636-42.
37) Khan M, Loree JM, Advani SM, Ning J, Li W, Pereira AA,
et al. Prognostic implications of mucinous differentiation
in metastatic colorectal carcinoma can be explained
by distinct molecular and clinicopathologic characteristics.
Clin Colorectal Cancer 2018;17(4):e699-709.
38) Lizardo DY, Kuang C, Hao S, Yu J, Huang Y, Zhang L.
Immunotherapy efficacy on mismatch repair-deficient
colorectal cancer: From bench to bedside. Biochim
Biophys Acta Rev Cancer 2020;1874(2):188447.
39) Mulet-Margalef N, Linares J, Badia-Ramentol J, Jimeno
M, Sanz Monte C, Manzano Mozo JL, et al. Challenges
and therapeutic opportunities in the dMMR/MSI-H
colorectal cancer landscape. Cancers 2023;15(4):1022.
40) Zhang X, Wu T, Cai X, Dong J, Xia C, Zhou Y, et al.
Neoadjuvant immunotherapy for MSI-H/dMMR locally
advanced colorectal cancer: New strategies and unveiled
opportunities. Front Immunol 2022;13:795972.
41) Cohen R, Rousseau B, Vidal J, Colle R, Diaz LA, André
T. Immune checkpoint inhibition in colorectal cancer:
Microsatellite instability and beyond. Target Oncol
2020;15:11-24.
42) Lin A, Zhang J, Luo P. Crosstalk between the MSI status
and tumor microenvironment in colorectal cancer.
Front Immunol 2020;11:552467.
43) Gupta R, Sinha S, Paul RN. The impact of microsatellite
stability status in colorectal cancer. Curr Probl
Cancer 2018;42(6):548-59.
44) Guinney J, Dienstmann R, Wang X, De Reynies A, Schlicker
A, Soneson C, et al. The consensus molecular subtypes
of colorectal cancer. Nat Med 2015;21(11):1350-6.
45) Toor SM, Sasidharan Nair V, Murshed K, Abu Nada
M, Elkord E. Tumor-infiltrating lymphoid cells in colorectal
cancer patients with varying disease stages and
microsatellite instability-high/stable tumors. Vaccines
2021;9(1):64.
46) Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia
J, Segal NH, et al. Immunotherapy in colorectal cancer:
Rationale, challenges and potential. Nat Rev Gastroenterol
Hepatol 2019;16(6):361-75.
47) Heregger R, Huemer F, Steiner M, Gonzalez-Martinez A,
Greil R, Weiss L. Unraveling resistance to immunotherapy
in MSI-high colorectal cancer. Cancers 2023;15(20):5090.
48) Koi M, Carethers JM. The colorectal cancer immune
microenvironment and approach to immunotherapies.
Future Oncol 2017;13(18):1633-47.
49) Sun CH, Li BB, Wang B, Zhao J, Zhang XY, Li TT, et
al. The role of fusobacterium nucleatum in colorectal
cancer: From carcinogenesis to clinical management.
Chronic Dis Transl Med 2019;5(3):178-87.
50) Wu J, Li Q, Fu X. Fusobacterium nucleatum contributes
to the carcinogenesis of colorectal cancer by inducing
inflammation and suppressing host immunity.
Transl Oncol 2019;12(6):846-51.
51) Overman MJ, McDermott R, Leach JL, Lonardi S,
Lenz HJ, Morse MA, et al. Nivolumab in patients with
metastatic DNA mismatch repair-deficient or microsatellite
instability-high colorectal cancer (Check-
Mate 142): An open-label, multicentre, phase 2 study.
Lancet Oncol 2017;18(9):1182-91.
52) Lenz HJ, Van Cutsem E, Luisa Limon M, Wong KYM,
Hendlisz A, Aglietta M, et al. First-line nivolumab plus
low-dose ipilimumab for microsatellite instabilityhigh/
mismatch repair-deficient metastatic colorectal
cancer: The phase II CheckMate 142 study. J Clin Oncol
2022;40(2):161-70.
53) Andre T, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt
CJ, et al. Pembrolizumab versus chemotherapy for microsatellite
instability-high/mismatch repair-deficient
metastatic colorectal cancer: The phase 3 KEYNOTE-177
Study. J Clin Oncol 2020;38(18):2163-72.
54) Pei F, Wu J, Zhao Y, He W, Yao Q, Huang M, et al. Single-
agent neoadjuvant immunotherapy with a PD-1
antibody in locally advanced mismatch repair-deficient
or microsatellite instability-high colorectal cancer.
Clin Colorectal Cancer 2023;22(1):85-91.
55) He R, Lao Y, Yu W, Zhang X, Jiang M, Zhu C. Progress
in the application of immune checkpoint inhibitorbased
immunotherapy for targeting different types of
colorectal cancer. Front Oncol 2021;11:764618.
56) Tian J, Chen JH, Chao SX, Pelka K, Giannakis M, Hess
J, et al. Combined PD-1, BRAF and MEK inhibition
in BRAFV600E colorectal cancer: A phase 2 trial. Nat
Med 2023;29(2):458-66.
57) Rus Bakarurraini NAA, Ab Mutalib NS, Jamal R, Abu
N. The landscape of tumor-specific antigens in colorectal
cancer. Vaccines 2020;8(3):371.
58) Ma X, Dong L, Liu X, Ou K, Yang L. POLE/POLD1
mutation and tumor immunotherapy. J Exp Clin
Cancer Res 2022;41(1):216.
59) Ozcan M, Janikovits J, von Knebel Doeberitz M, Kloor
M. Complex pattern of immune evasion in MSI colorectal
cancer. Oncoimmunology 2018;7(7):e1445453.
60) Huang Q, Yu T, Li L, Zhang Q, Zhang S, Li B, et al. Intraindividual
tumor heterogeneity of mismatch repair
status in metastatic colorectal cancer. Appl Immunohistochem
Mol Morphol 2023;31(2):84-93.
61) Chung BS, Liao IC, Lin PC, Wu SY, Kang JW, Lin BW, et
al. PD-L1 expression in high-risk early-stage colorectal
cancer?its clinical and biological significance in immune
microenvironment. Int J Mol Sci 2022;23(21):13277.
62) Twomey JD, Zhang B. Cancer immunotherapy update:
FDA-approved checkpoint inhibitors and companion
diagnostics. AAPS J 2021;23:1-11.
63) Scott RJ. Modifier genes and Lynch syndrome: Some
considerations. Hered Cancer Clin Pract 2022;20(1):35.
64) Ramos-Casals M, Brahmer JR, Callahan MK, Flores-
Chávez A, Keegan N, Khamashta MA, et al. Immune-
related adverse events of checkpoint inhibitors.
Nat Rev Dis Primers 2020;6(1):38.
65) Temraz S, Nassar F, Nasr R, Charafeddine M, Mukherji
D, Shamseddine A. Gut microbiome: A promising
biomarker for immunotherapy in colorectal cancer.
Int J Mol Sci 2019;20(17):4155.
66) Jhunjhunwala S, Hammer C, Delamarre L. Antigen
presentation in cancer: Insights into tumour immunogenicity
and immune evasion. Nat Rev Cancer
2021;21(5):298-312.
67) Perillo F, Amoroso C, Strati F, Giuffrè MR, Díaz-Basabe
A, Lattanzi G, et al. Gut microbiota manipulation
as a tool for colorectal cancer management: Recent advances
in its use for therapeutic purposes. Int J Mol Sci
2020;21(15):5389.
68) Sharma P, Allison JP. Immune checkpoint targeting in
cancer therapy: Toward combination strategies with
curative potential. Cell 2015;161(2):205-14.
69) Molinari C, Marisi G, Passardi A, Matteucci L, De
Maio G, Ulivi P. Heterogeneity in colorectal cancer:
A challenge for personalized medicine? Int J Mol Sci
2018;19(12):3733.
70) Chan EM, Shibue T, McFarland JM, Gaeta B, Ghandi
M, Dumont N, et al. WRN helicase is a synthetic
lethal target in microsatellite unstable cancers. Nature
2019;568(7753):551-6.
71) Brosh Jr RM, Opresko PL, Bohr VA. Enzymatic mechanism
of the WRN helicase/nuclease. Methods Enzymol
2006;409:52-85.
72) Jacinto FV, Esteller M. Mutator pathways unleashed
by epigenetic silencing in human cancer. Mutagenesis
2007;22(4):247-53.
73) Huang A, Garraway LA, Ashworth A, Weber B. Synthetic
lethality as an engine for cancer drug target discovery.
Nat Rev Drug Discov 2020;19(1):23-38.
74) Lieb S, Blaha-Ostermann S, Kamper E, Rippka J,
Schwarz C, Ehrenhöfer-Wölfer K, et al. Werner syndrome
helicase is a selective vulnerability of microsatellite
instability-high tumor cells. Elife 2019;8:e43333.
75) van Wietmarschen N, Nathan WJ, Nussenzweig A. The
WRN helicase: Resolving a new target in microsatellite
unstable cancers. Curr Opin Genet Dev 2021;71:34?8.
76) Sobol RW. WRN suppresses p53/PUMA-induced
apoptosis in colorectal cancer with microsatellite instability/
mismatch repair deficiency. Proc Natl Acad
Sci U S A 2023;120(2):e2219963120.
77) Zhou J, Zhou XA, Zhang N, Wang J. Evolving insights:
How DNA repair pathways impact cancer evolution.
Cancer Biol Med 2020;17(4):805.
78) Zhou F, Yang G, Liu Y, Xue L, Guo Y, Li Z, et al. Discovery
of a novel WRN inhibitor, ZM-3329 that efficiently
inhibits MSI-H tumor growth. Cancer Res
2024;84(6_Suppl):7278.