Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (195)

Search Parameters:
Keywords = MMR/MSI

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 677 KB  
Review
Can TP53, TMB and TME Expand the Immunotherapy Benefit in Metastatic Colorectal Cancer?
by Monia Specchia, Denise Drittone, Eva Mazzotti and Federica Mazzuca
Cancers 2025, 17(24), 3984; https://doi.org/10.3390/cancers17243984 - 13 Dec 2025
Viewed by 187
Abstract
Background: Metastatic colorectal cancer (mCRC) with TP53 gene mutations, which are commonly found in tumors that are microsatellite stable (MSS) and not prone to genetic errors seen in some cancers, is associated with aggressive cancer behavior and poor outcomes. While MSI-high (MSI-H, referring [...] Read more.
Background: Metastatic colorectal cancer (mCRC) with TP53 gene mutations, which are commonly found in tumors that are microsatellite stable (MSS) and not prone to genetic errors seen in some cancers, is associated with aggressive cancer behavior and poor outcomes. While MSI-high (MSI-H, referring to high levels of gene instability) disease benefits markedly from PD-1-based immunotherapy (drugs that inhibit the PD-1 protein on immune cells), TP53-mutated MSS tumors rarely receive immune checkpoint inhibitors (ICIs, drugs that help immune cells attack cancer) outside of trials and often only in later lines of therapy. Objective: We aimed to synthesize translational and clinical evidence regarding the effects of early rationale-driven immunotherapy combinations on survival outcomes, in TP53-mutated metastatic colorectal cancer, with a focus on practical clinical implications. Methods: This narrative review was conducted in accordance with SANRA criteria. Literature searches were performed in PubMed/MEDLINE, Scopus, and Web of Science (2010–2025). Relevant ESMO and NCCN guidelines and key references were also reviewed. Results: In KEYNOTE-177 study (MSI-H/dMMR), pembrolizumab improved PFS (HR 0.60) and showed durable OS with >5-year follow-up. CheckMate-142 reported sustained activity with nivolumab ± ipilimumab. Preclinical/early clinical data in MSS/TP53 suggest that ICIs may become effective when combined with priming (chemo/DDR) and vascular normalization (anti-VEGF), particularly in subsets with elevated TMB. The randomized ROME trial supports the clinical utility of genomically matched, NGS-guided strategies. Conclusions: A precision approach integrating TP53 status, TMB, and TME modulation could extend the immunotherapy benefit beyond MSI-H to TP53-mutated MSS mCRC; prospective first-line trials are warranted. Full article
(This article belongs to the Section Cancer Metastasis)
Show Figures

Figure 1

46 pages, 3834 KB  
Review
Lynch Syndrome in Focus: A Multidisciplinary Review of Cancer Risk, Clinical Management, and Special Populations
by Seyma Eroglu, Ilhan Birsenogul, Alexandra P. Bowen, Joseph F. Doyle, Stephen E. Pupkin, Joaquin Villar, Christopher M. Tarney, Edwin Gandia, Danielle M. Pastor and Juvianee I. Estrada-Veras
Cancers 2025, 17(24), 3981; https://doi.org/10.3390/cancers17243981 - 13 Dec 2025
Viewed by 153
Abstract
Lynch syndrome (LS), also known as hereditary nonpolyposis colorectal cancer (HNPCC), is a hereditary cancer syndrome significantly increasing the risk of colorectal cancer (CRC) and various extracolonic cancers, including endometrial, ovarian, and gastric cancers. LS results from germline pathogenic variants (GPVs) in DNA [...] Read more.
Lynch syndrome (LS), also known as hereditary nonpolyposis colorectal cancer (HNPCC), is a hereditary cancer syndrome significantly increasing the risk of colorectal cancer (CRC) and various extracolonic cancers, including endometrial, ovarian, and gastric cancers. LS results from germline pathogenic variants (GPVs) in DNA mismatch repair (MMR) genes, such as MLH1, MSH2, MSH6, and PMS2, leading to microsatellite instability (MSI). This review explores the multifaceted aspects of LS, covering clinical presentation, genetic underpinnings, and emerging therapeutic strategies. The discussion explores the importance of identifying at-risk individuals, facilitating personalized cancer surveillance and prevention strategies. Molecular insights into distinguishing between sporadic and LS-associated cancers are also examined, with a focus on somatic testing methods, including MSI and immunohistochemistry (IHC). The gynecological cancer risks, particularly those related to endometrial and ovarian malignancies, are addressed, underscoring the need for early detection and risk-reducing interventions. Recent advancements in the management of colorectal and other LS-related cancers are highlighted, with particular attention to the growing role of immunotherapy, including immune checkpoint inhibitors, and immunoprevention strategies. With ongoing advances in our understanding of LS, opportunities for earlier detection, more effective prevention, and innovative treatments continue to expand. This narrative review adopts a multidisciplinary approach to provide a comprehensive understanding of LS, from its genetic basis to current clinical and therapeutic practices, with the ultimate goal of improving patient outcomes and enhancing the quality of care. Full article
(This article belongs to the Special Issue Lynch Syndrome (HNPCC): Symptoms, Causes, and Outlooks)
Show Figures

Figure 1

24 pages, 460 KB  
Review
Precision Care for Hereditary Urologic Cancers: Genetic Testing, Counseling, Surveillance, and Therapeutic Implications
by Takatoshi Somoto, Takanobu Utsumi, Rino Ikeda, Naoki Ishitsuka, Takahide Noro, Yuta Suzuki, Shota Iijima, Yuka Sugizaki, Ryo Oka, Takumi Endo, Naoto Kamiya and Hiroyoshi Suzuki
Curr. Oncol. 2025, 32(12), 698; https://doi.org/10.3390/curroncol32120698 - 11 Dec 2025
Viewed by 173
Abstract
Hereditary predisposition substantially shapes prevention and management across urologic oncology. This narrative review synthesizes contemporary, practice-oriented guidance on whom to test, what to test, how to act on results, and how to implement care equitably for hereditary forms of prostate cancer, renal cell [...] Read more.
Hereditary predisposition substantially shapes prevention and management across urologic oncology. This narrative review synthesizes contemporary, practice-oriented guidance on whom to test, what to test, how to act on results, and how to implement care equitably for hereditary forms of prostate cancer, renal cell carcinoma (RCC), urothelial carcinoma, pheochromocytoma/paraganglioma (PPGL), and adrenocortical carcinoma (ACC). We delineate between forms of indication-driven germline testing (e.g., universal testing in metastatic prostate cancer; early-onset, bilateral/multifocal, or syndromic RCC; reflex tumor mismatch repair (MMR)/microsatellite instability (MSI) screening in upper-tract urothelial carcinoma (UTUC); universal testing in PPGL; universal TP53 testing in ACC) and pair these strategies with minimum actionable gene sets and syndrome-specific surveillance frameworks. Key points include targeted prostate-specific antigen screening in BRCA2 carriers and the impact of BRCA/ATM variants on reclassification during active surveillance; major hereditary RCC syndromes with genotype-tailored surveillance and pathway-directed therapy (e.g., HIF-2α inhibition for von Hippel–Lindau disease); UTUC/bladder cancer in Lynch syndrome with tumor MMR/MSI screening, annual urinalysis (selective cytology), and immunotherapy opportunities in deficient MMR disease/MSI-H; PPGL management emphasizing universal germline testing, intensified surveillance for SDHB, cortical-sparing adrenalectomy, and emerging HIF-2α inhibition; and ACC care modified by Li–Fraumeni syndrome (minimization of radiation/genotoxic therapy with whole-body imaging surveillance). Testicular germ cell tumor remains largely polygenic, with no routine germline testing in typical presentations. Finally, we provide pre-/post-test genetic-counseling checklists and mainstreamed workflows with equity metrics to operationalize precision care and close real-world access gaps. Full article
(This article belongs to the Section Genitourinary Oncology)
Show Figures

Figure 1

15 pages, 1467 KB  
Article
Real-World Impact of Metformin on Outcomes in Patients with Deficient DNA Mismatch Repair and Microsatellite Instability (dMMR/MSI) Colorectal Cancer Treated with Immune Checkpoint Inhibitors
by Garima Gupta, Negar Sadeghipour, Fuat Bicer, Andrew Elliott, Andrew Hinton, Emil Lou, Ari M. Vanderwalde, Ahmet Anil Ozluk, Moh’d M. Khushman, Midhun Malla, Darryl Outlaw, Syed Qasim Hussaini, Bassel F. El-Rayes and Mehmet Akce
Cancers 2025, 17(24), 3944; https://doi.org/10.3390/cancers17243944 - 10 Dec 2025
Viewed by 300
Abstract
Background/Objectives: Colorectal cancer (CRC) is the second leading cause of cancer-related death in the US. The presence of deficient DNA mismatch repair and microsatellite instability (dMMR/MSI) in CRC is linked to responses to immune checkpoint inhibitors (ICIs). This study investigates the impact [...] Read more.
Background/Objectives: Colorectal cancer (CRC) is the second leading cause of cancer-related death in the US. The presence of deficient DNA mismatch repair and microsatellite instability (dMMR/MSI) in CRC is linked to responses to immune checkpoint inhibitors (ICIs). This study investigates the impact of metformin on the tumor microenvironment (TME) and clinical outcomes of patients with dMMR/MSI CRC treated with ICIs, aiming to better understand its potential role in enhancing ICI efficacy. Methods: Of 25,011 CRC patients in Caris database, 47 received both metformin and ICI therapy (Met-ICI group), and 475 patients received ICI therapy alone (ICI group). Samples underwent genomic or transcriptome sequencing at Caris Life Sciences. Immune cell fractions were estimated using quanTIseq. Univariate and multivariate survival analyses were conducted using the Cox proportional model. Results: The TME analysis of CRC patient samples revealed that TMB-High (≥10 mut/Mb) was more prevalent in the “ICI” group compared to the “Met-ICI” group (99.1% vs. 95.6%, p = 0.036). Mutation rates for most genes between the two groups were similar, but CIC gene mutations were more common in the “ICI” group than in the “Met-ICI” group (23.2% vs. 4.8%, p = 0.006). No significant differences were observed in the PD-L1 positivity rate or immune checkpoint gene expression (including IDO1, IFNG, TIM3, and CTLA4). M1 macrophages and neutrophils showed the highest infiltration among immune cells. However, the fractions of infiltrated immune cells were similar between the two cohorts. Univariate and multivariate analyses showed that there was no significant difference in patient survival between “ICI” and “Met-ICI” cohorts. Conclusions: In this retrospective analysis of real-world clinical outcomes, the concurrent use of metformin with ICIs in patients with dMMR/MSI CRC did not reveal an impact on clinical outcomes. Full article
Show Figures

Figure 1

17 pages, 1004 KB  
Article
Incorporation of Microsatellite Instability and Tumor-Infiltrating Lymphocytes in Opisthorchis viverrini-Associated Cholangiocarcinoma: Predicting Patient Outcomes
by Natcha Khuntikeo, Apiwat Jareanrat, Vasin Thanasukarn, Tharatip Srisuk, Vor Luvira, Watcharin Loilome, Poramate Klanrit, Anchalee Techasen, Jarin Chindaprasirt, Prakasit Sa-Ngiamwibool, Chaiwat Aphivatanasiri, Sureerat Padthaisong, Piya Prajumwongs and Attapol Titapun
J. Mol. Pathol. 2025, 6(4), 30; https://doi.org/10.3390/jmp6040030 - 1 Dec 2025
Viewed by 197
Abstract
Background: Cholangiocarcinoma (CCA) has the highest incidence in Northeastern Thailand, where patients generally present with late diagnosis and poor prognosis. Opisthorchis viverrini (OV) infection is the major cause of CCA, with oxidative stress driving DNA mutations and genetic instability. Microsatellite instability (MSI) is [...] Read more.
Background: Cholangiocarcinoma (CCA) has the highest incidence in Northeastern Thailand, where patients generally present with late diagnosis and poor prognosis. Opisthorchis viverrini (OV) infection is the major cause of CCA, with oxidative stress driving DNA mutations and genetic instability. Microsatellite instability (MSI) is a predictive biomarker in several cancers. This study aimed to investigate MSI status and its association with clinicopathological features and survival of CCA patients. Methods: Tissue and serum samples were collected from 25 surgical CCA patients. MSI status and mismatch repair (MMR) proteins were evaluated using an MSI scanner and immunohistochemistry (IHC). Serum OV IgG was assessed by ELISA, while tumor-infiltrating lymphocytes (TILs) were evaluated by two pathologists. Associations of MSI with clinicopathological features, OV status, MMR, and survival were analyzed. Results: Among CCA patients, 66.7% were MSI-high and 33.3% were MSI-low. MSI-high significantly correlated with age < 57 years, intraductal growth pattern, OV positivity, and early-stage disease. Patients with MSI-high and high TILs showed markedly improved median survival compared to MSI-low with low TILs (94.0 vs. 16.8 and 3.0 months; HR = 6.82 and 14.10; p = 0.004 and 0.001). Incorporation of MSI and TILs remained an independent prognostic factor in multivariate analysis (p < 0.05). Conclusions: MSI-high is highly prevalent in OV-associated CCA and is associated with intraductal growth, OV infection, and early-stage disease. Combined MSI and TIL status may serve as an independent prognostic factor, warranting validation in larger cohorts. Full article
Show Figures

Figure 1

8 pages, 771 KB  
Brief Report
The Role of Microsatellite Instability in Endometrial Hyperplasia and Risk of Carcinoma Development
by Angelina Mollova-Kyosebekirova, Ekaterina Uchikova, Anna Mihaylova, Mariya Koleva-Ivanova, Mariana Parahuleva and Nikoleta Parahuleva
Biomedicines 2025, 13(12), 2953; https://doi.org/10.3390/biomedicines13122953 - 30 Nov 2025
Viewed by 255
Abstract
Background: Endometrial hyperplasia (EH) represents a precursor lesion in the development of endometrial carcinoma, particularly the endometrioid subtype. Among the molecular pathways involved, microsatellite instability (MSI) resulting from DNA mismatch repair (MMR) deficiency has gained increasing attention as an early event in endometrial [...] Read more.
Background: Endometrial hyperplasia (EH) represents a precursor lesion in the development of endometrial carcinoma, particularly the endometrioid subtype. Among the molecular pathways involved, microsatellite instability (MSI) resulting from DNA mismatch repair (MMR) deficiency has gained increasing attention as an early event in endometrial carcinogenesis. Objective: This study aimed to evaluate the expression of key MMR proteins (MLH1, PMS2, MSH2, and MSH6) in endometrial hyperplasia without atypia and endometrial atypical hyperplasia/endometrioid intraepithelial neoplasia (EAH/EIN) to determine the prevalence and potential implications of MMR deficiency at the precancerous stage. Methods: Fifty-six cases of EH were analyzed, including 28 endometrial hyperplasia without atypia and 28 EAH/EIN. Immunohistochemical (IHC) analysis was performed to assess the nuclear expression of MMR proteins. Loss of expression was defined as complete absence of epithelial nuclear staining with retained stromal positivity. Results: MMR protein expression was retained in all cases of endometrial hyperplasia without atypia, while total loss of one or more MMR proteins was observed in 3 of 28 (10.7%) EAH/EIN. The most frequent pattern involved concurrent MLH1/PMS2 loss, consistent with sporadic MLH1 promoter hypermethylation. One case exhibited isolated MSH6 loss, suggesting a potential Lynch syndrome, and another showed combined MSH6/PMS2 loss. Conclusions: MMR deficiency appears confined to atypical EH, supporting its role as an early molecular alteration in the neoplastic sequence leading to endometrioid carcinoma. Identification of abnormal MMR expression in EH may facilitate risk stratification, guide reflex testing for MLH1 methylation, and prompt genetic counseling for hereditary cancer predisposition. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

32 pages, 1317 KB  
Review
Therapeutic Frontiers in Gastroesophageal Cancer: Contemporary Concepts in Management and Therapy
by Supriya Peshin, Ehab Takrori, Naga Anvesh Kodali, Faizan Bashir, Michael Gibson and Sakshi Singal
Int. J. Mol. Sci. 2025, 26(23), 11424; https://doi.org/10.3390/ijms262311424 - 26 Nov 2025
Viewed by 485
Abstract
Gastroesophageal cancer (GEC) represents a global health burden, with rising incidence and high mortality. Despite advancements in early detection and systemic therapies, outcomes remain poor, especially in advanced stages. Management requires a multidisciplinary, multimodal approach that integrates surgery, chemotherapy, radiotherapy, targeted agents, and [...] Read more.
Gastroesophageal cancer (GEC) represents a global health burden, with rising incidence and high mortality. Despite advancements in early detection and systemic therapies, outcomes remain poor, especially in advanced stages. Management requires a multidisciplinary, multimodal approach that integrates surgery, chemotherapy, radiotherapy, targeted agents, and immunotherapy, tailored by tumor histology, location, and molecular profile. For localized disease, perioperative chemotherapy or chemoradiotherapy is standard, with adjuvant immunotherapy now emerging in selected high-risk cases. In metastatic or unresectable settings, systemic therapy forms the backbone of treatment, with biomarker-driven regimens targeting HER2, PD-L1, MSI-H/dMMR, and CLDN18.2, offering improved outcomes. Novel agents and combinations, including bispecific antibodies, FGFR2 inhibitors, and immunotherapy-based strategies, are actively being explored in clinical trials. This review provides a comprehensive overview of the evolving therapeutic landscape of GEC. It emphasizes the growing role of precision medicine and the integration of emerging clinical data into practice. Full article
(This article belongs to the Special Issue Translational Oncology: From Molecular Basis to Therapy)
Show Figures

Figure 1

13 pages, 960 KB  
Systematic Review
The New Horizon for Non-Metastatic dMMR Colorectal Cancer: A Systematic Review of the Adjuvant Chemoimmunotherapy and Neoadjuvant Immunotherapy Revolution
by Nabil Ismaili
Gastroenterol. Insights 2025, 16(4), 43; https://doi.org/10.3390/gastroent16040043 - 16 Nov 2025
Viewed by 1190
Abstract
Background: The management of resected stage III colorectal cancer (CRC) has long been reliant on fluoropyrimidine-based adjuvant chemotherapy. However, the 10–15% of patients with mismatch repair-deficient (dMMR) tumors derive limited benefit from this approach. While immunotherapy has revolutionized the treatment of metastatic dMMR [...] Read more.
Background: The management of resected stage III colorectal cancer (CRC) has long been reliant on fluoropyrimidine-based adjuvant chemotherapy. However, the 10–15% of patients with mismatch repair-deficient (dMMR) tumors derive limited benefit from this approach. While immunotherapy has revolutionized the treatment of metastatic dMMR CRC, its role in the early-stage setting is rapidly evolving, creating a paradigm shift. Methods: A systematic literature review was conducted to identify pivotal clinical trials evaluating therapeutic strategies for non-metastatic dMMR CRC. Databases including PubMed/MEDLINE and conference proceedings from the American Society of Clinical Oncology (ASCO) and European Society for Medical Oncology (ESMO) were searched up to June 2025. The review focused on phase II and III trials reporting on disease-free survival (DFS), pathological complete response (pCR), and safety. Study selection followed PRISMA guidelines. Results: The systematic review identified 14 key studies that were included for narrative synthesis. The evidence base encompassed three areas: (1) Foundational adjuvant chemotherapy trials (e.g., MOSAIC, IDEA); (2) Pivotal metastatic trials (e.g., KEYNOTE-177) validating immunotherapy efficacy in dMMR CRC; and (3) Modern trials in non-metastatic disease. The phase III ATOMIC trial demonstrated that adding atezolizumab to mFOLFOX significantly improved 3-year DFS versus chemotherapy alone (86.4% vs. 76.6%; Hazard Ratio [HR] 0.50, 95% Confidence Interval [CI] 0.34–0.72; p < 0.001). Concurrently, phase II neoadjuvant immunotherapy trials (e.g., NICHE-2) reported remarkable pCR rates of 68% and a 3-year DFS of 100%, with a more favorable safety profile compared to chemoimmunotherapy. Conclusions: The landscape for non-metastatic dMMR CRC is shifting from a chemotherapy-based model to an immunotherapy paradigm. The ATOMIC trial establishes adjuvant chemoimmunotherapy as a new standard, while robust neoadjuvant data suggest a potential future where short-course, chemotherapy-free immunotherapy could become a preferred strategy. Ongoing trials directly comparing these approaches are awaited. Full article
(This article belongs to the Collection Advances in Gastrointestinal Cancer)
Show Figures

Figure 1

14 pages, 531 KB  
Article
Efficacy of Anti-VEGF and Anti-EGFRs in Microsatellite Instable (MSI-H) Metastatic Colorectal Cancer in a Turkish Oncology Group (TOG) Cohort Study
by İlknur Deliktaş Onur, Mutlu Doğan, Mehmet Akif Öztürk, Taha Koray Sahin, Murat Kiracı, Ahmet Melih Arslan, Eda Karapelit, Bahar Beliz Karaoğlan, Nargiz Majidova, Elif Şahin, Sabin Göktaş, Abdullah Sakin, Ali Oğul, Emine Türkmen, Kadriye Başkurt, Zeynep Yüksel Yaşar, Yakup Ergün, Esma Türkmen Bekmez, Şafak Yıldırım Dişli, Sinem Akbaş, Sema Türker, Ömer Dizdar, Öznur Bal, Tuğba Yavuzşen, Melek Karakurt, Arzu Hatime Yaşar, Tuğba Başoğlu, Faysal Dane, Şuayip Yalçın and Öztürk Ateşadd Show full author list remove Hide full author list
Curr. Oncol. 2025, 32(11), 639; https://doi.org/10.3390/curroncol32110639 - 14 Nov 2025
Viewed by 402
Abstract
Background: Mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H) colorectal tumors constitute 5% of metastatic colorectal cancer(mCRC). Immunotherapy is a new standard, but it is difficult to provide for all patients. 5-Flurouracil-based treatment with anti-EGFRs (cetuximab and panitumumab) in RAS/BRAF-wild or anti-VEGF (bevacizumab) is used in [...] Read more.
Background: Mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H) colorectal tumors constitute 5% of metastatic colorectal cancer(mCRC). Immunotherapy is a new standard, but it is difficult to provide for all patients. 5-Flurouracil-based treatment with anti-EGFRs (cetuximab and panitumumab) in RAS/BRAF-wild or anti-VEGF (bevacizumab) is used in mCRC. Data is limited for the efficacy of anti-VEGF or anti-EGFRs in dMMR/MSI-H mCRC due to the small number of cases in the colorectal cancer population in trials. Aims: To evaluate prognostic factors in dMMR/MSI-H mCRC and compare progression-free survival time of patients receiving anti-VEGF and anti-EGFR combined with first-line 5FU-based therapy. Methods: Patients with metastatic dMMR/MSI-H colorectal cancer diagnosed between January 2015 and January 2023 were included in this cohort study. Progression-free survival times of patients treated with first-line therapy were compared. Prognostic factors associated with overall survival were investigated. Results: A total of 132 patients were included. Mutation rates were 35.6% (n:47) for RAS and 12.1% (n: 16) for BRAF (. Median progression-free survival (PFS) was 10.9 (95% CI: 9.2–12.6) months. Median overall survival (OS) was 44 months (95% CI: 26.23–63.03). 82 (62.1%) patients had primary tumor resection (PTR), 26 (19.7%) had PTR and metastasectomy. A total of 17 (12.8%) de novo mCRC patients had maximal cytoreductive surgery (MCS). A total of 14 (10.6%) patients had subsequent immunotherapy (IO). In multivariate analysis, RAS/BRAF mutation status, MCS, and subsequent IO are defined as prognostic factors for OS (p < 0.01, p: 0.022, and p: 0.005, respectively). No statistically significant difference (PFS, OS) was found in patients receiving first-line anti-VEGF or anti-EGFR therapy. Conclusions: dMMR/MSI-H mCRC is an entity with different tumor biology. We consider that dMMR/MSI-H mCRC patients with BRAF wild, MCS and subsequent IO have better outcomes with 1st line 5FU-based treatment with anti-VEGF/anti-EGFRs. Full article
(This article belongs to the Section Gastrointestinal Oncology)
Show Figures

Figure 1

25 pages, 335 KB  
Review
Current Management of Locally Advanced Esophageal and Esophagogastric Junction Cancers: Clinical Evidence and Evolving Strategies
by Andrea Di Donato and Marc Van den Eynde
Cancers 2025, 17(22), 3603; https://doi.org/10.3390/cancers17223603 - 8 Nov 2025
Viewed by 1560
Abstract
The curative management of localized esophageal and esophagogastric junction (EGJ) cancers has undergone major changes over the past decade, shaped by multimodal strategies integrating chemotherapy, chemoradiotherapy, surgery, and more recently, immunotherapy. For esophageal squamous cell carcinoma (SCC), neoadjuvant or definitive chemoradiotherapy remains the [...] Read more.
The curative management of localized esophageal and esophagogastric junction (EGJ) cancers has undergone major changes over the past decade, shaped by multimodal strategies integrating chemotherapy, chemoradiotherapy, surgery, and more recently, immunotherapy. For esophageal squamous cell carcinoma (SCC), neoadjuvant or definitive chemoradiotherapy remains the standard of care in Western countries. In contrast, for adenocarcinoma (AC) of the esophagus and EGJ, perioperative chemotherapy has emerged as the preferred strategy. Despite these advances, long-term outcomes remain suboptimal, and recurrence continues to pose a major challenge, highlighting the need to optimize patient selection and treatment sequencing. The integration of immunotherapy in the perioperative or adjuvant setting has recently led to improvements in surrogate endpoints yet overall survival benefit remains under investigation. For patients with tumors harboring microsatellite instability-high (MSI-H) or mismatch repair deficiency (dMMR), checkpoint inhibitors show exceptional activity, and non-operative management may be feasible in select cases. Conversely, human epidermal growth receptor 2 (HER2)-targeted strategies, although effective in metastatic disease, have not yet translated into practice-changing benefit in the curative setting. The role of circulating tumor deoxyribo nucleic acid (DNA) and functional imaging as real-time tools to assess response and guide treatment adaptation is also being actively explored. This review provides a comprehensive overview of current standards, ongoing developments, and future directions for the treatment of localized esophageal and EGJ cancers, with a focus on emerging personalization strategies and biomarker-driven approaches aimed at improving cure rates and minimizing treatment-related morbidity. Full article
(This article belongs to the Special Issue Current Treatments of Esophageal and Esophagogastric Junction Cancers)
26 pages, 3160 KB  
Review
Gut Microbiota and Ferroptosis in Colorectal Cancer: A Comprehensive Review of Mechanisms and Therapeutic Strategies to Overcome Immune Checkpoint Resistance
by Yingchang Cai, Feng Zhao and Xiaofei Cheng
Biomolecules 2025, 15(11), 1546; https://doi.org/10.3390/biom15111546 - 3 Nov 2025
Cited by 1 | Viewed by 1319
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality worldwide. Although immune checkpoint inhibitors (ICIs) have achieved striking clinical efficacy in the subset of CRCs with mismatch repair deficiency/high microsatellite instability (dMMR/MSI-H), the vast majority of patients—those with proficient mismatch repair/microsatellite-stable (pMMR/MSS) [...] Read more.
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality worldwide. Although immune checkpoint inhibitors (ICIs) have achieved striking clinical efficacy in the subset of CRCs with mismatch repair deficiency/high microsatellite instability (dMMR/MSI-H), the vast majority of patients—those with proficient mismatch repair/microsatellite-stable (pMMR/MSS) tumors—derive little benefit from current immunotherapies. Ferroptosis, an iron-dependent form of regulated cell death driven by lethal accumulation of lipid peroxides, has emerged as a promising antitumor mechanism that can interact with and modulate antitumor immunity. Concurrently, the gut microbiota exerts powerful control over host metabolism and immune tone through microbial community structure and metabolite production; accumulating evidence indicates that microbiota-derived factors can either sensitize tumors to ferroptosis (for example, via short-chain fatty acids) or confer resistance (for example, indole-3-acrylic acid produced by Peptostreptococcus anaerobius acting through the AHR→ALDH1A3→FSP1/CoQ axis). In this review we synthesize mechanistic data linking microbial ecology, iron and lipid metabolism, and immune regulation to ferroptotic vulnerability in CRC. We discuss translational strategies to exploit this “microbiota–ferroptosis” axis—including precision microbiome modulation, dietary interventions, pharmacologic ferroptosis inducers, and tumor-targeted delivery systems—and we outline biomarker frameworks and trial designs to evaluate combinations with ICIs. We also highlight major challenges, such as interindividual microbiome variability, potential collateral harm to ferroptosis-sensitive immune cells, adaptive antioxidant compensation (e.g., NRF2/FSP1 activation), and safety/regulatory issues for live biotherapeutics. In summary, this review highlights that targeting the microbiota-ferroptosis axis may represent a rational and potentially transformative approach to reprogramming the tumor microenvironment and overcoming immune checkpoint resistance in pMMR/MSS colorectal cancer; however, further research is essential to validate this concept and address existing challenges. Full article
Show Figures

Figure 1

29 pages, 893 KB  
Review
Advances in Hereditary Colorectal Cancer: How Precision Medicine Is Changing the Game
by Shenghao Lin, Chenxi Zhou, Hanlin Chen, Xinlei Zhou, Hujia Yang, Leitao Sun, Leyin Zhang and Yuxin Zhang
Cancers 2025, 17(21), 3461; https://doi.org/10.3390/cancers17213461 - 28 Oct 2025
Viewed by 1424
Abstract
Only about 5% of colorectal cancers are hereditary, which is due to the low carrier rate of pathogenic gene mutations. The typical pattern of these cases is intergenerational aggregation within families and early onset. But public awareness of early diagnosis and intervention of [...] Read more.
Only about 5% of colorectal cancers are hereditary, which is due to the low carrier rate of pathogenic gene mutations. The typical pattern of these cases is intergenerational aggregation within families and early onset. But public awareness of early diagnosis and intervention of HCRC is insufficient, resulting in most patients being diagnosed only after developing cancer, thereby missing the optimal window for treatment. This article reviews the latest developments in precision screening, treatment, evaluation and prevention strategies for HCRC, including innovative uses of artificial intelligence (AI) in molecular diagnostics, imaging technology advances, and potential application prospects. Regarding precision screening, tests of genomics, transcriptomics, microbiome, etc., combined with personalised risk stratification, can, respectively, effectively detect pathogenic mutations and “cancer-promoting” intestinal environments in the preclinical stage. AI combined with endoscopic and imaging tools has improved the accuracy of polyp detection and tumor profiling. Liquid biopsy and molecular marker detection provide new non-invasive monitoring solutions. In precision treatment, beyond traditional approaches like surgery and chemotherapy, immunotherapy with checkpoint inhibitors may be considered for HCRC patients with mismatch repair deficiency (dMMR). For patients harboring somatic mutations such as KRAS or BRAF V600E, targeted therapy can be guided by these specific mutations. Regarding precision assessment, AI incorporates microsatellite instability (MSI) detection and imaging diagnostic techniques, crucial for integrating genetic, environmental, and lifestyle data in follow-up. This helps assess the risk of recurrence and adjust the long-term medication regimens, as well as provide effective nutritional support and psychological counselling. In summary, the rapid development of precision medicine is driving the clinical management of HCRC into the era of tailored care, aiming to optimise patient outcomes. Full article
(This article belongs to the Special Issue Hereditary and Familial Colorectal Cancer)
Show Figures

Figure 1

13 pages, 257 KB  
Article
Impact of COVID-19 on Universal Tumor Screening, Referral Rates and Attendance at Cancer Genetic Counseling at a Safety-Net University Hospital
by Dimitrios N. Varvoglis, Kelsey R. Landrum, Lydia H. Comer, Julianne M. O’Daniel, Chris B. Agala, Lacey M. Lee and José G. Guillem
Curr. Oncol. 2025, 32(10), 549; https://doi.org/10.3390/curroncol32100549 - 30 Sep 2025
Viewed by 469
Abstract
Universal tumor screening (UTS) of all newly diagnosed colorectal cancers (CRCs) for the identification of Lynch syndrome (LS) is recommended. We explored the impact of the COVID-19 pandemic on the UTS process in a safety-net university hospital to identify areas of vulnerability and [...] Read more.
Universal tumor screening (UTS) of all newly diagnosed colorectal cancers (CRCs) for the identification of Lynch syndrome (LS) is recommended. We explored the impact of the COVID-19 pandemic on the UTS process in a safety-net university hospital to identify areas of vulnerability and opportunities for improvement. Patients undergoing resection of a primary CRC were categorized into three cohorts based on surgery date relative to the pandemic (pre-[2018,2019], early-[2020,2021] and late-[2022]). Data regarding (1) UTS performance of immunohistochemistry (IHC) for LS genes and microsatellite instability (MSI) testing; (2) referrals to cancer genetic counseling (CGC) based on mismatch repair deficient (dMMR) status and/or age < 50 years at diagnosis; (3) attendance at CGC; and (4) reasons for not attending CGC were extracted. Between 2018 and 2022, 342 patients underwent resection of a CRC. During the three time periods (pre-, early- and late-pandemic), 93%, 94% and 96% of cases were screened with at least MMR IHC, respectively. Of the patients eligible for referral to CGC in each time period, 60%, 71% and 63% had a referral submitted. Of these, 23%, 36% and 20% in each time period did not attend CGC, with the most common reason for not attending being the inability of schedulers to reach the patient. Although the COVID-19 pandemic did not cause significant variation in the different steps of the UTS process, CGC utilization remained suboptimal throughout the three time periods. Further research on barriers preventing physicians from referring patients to CGC as well as schedulers inability to reach eligible patients should be pursued. Full article
(This article belongs to the Section Surgical Oncology)
Show Figures

Graphical abstract

28 pages, 1312 KB  
Review
Traditional and New Views on MSI-H/dMMR Endometrial Cancer
by Chuqi Liu, Huiyu Ping, Mengmeng Yao, Xinru Li, Qingxin Li, Ruotong Hu, Yawen Xu, Kaidi Meng, Fei Gao and Kai Meng
Biomolecules 2025, 15(10), 1370; https://doi.org/10.3390/biom15101370 - 26 Sep 2025
Viewed by 2064
Abstract
MSI-H/dMMR endometrial cancer (EC) is closely linked to the mismatch repair (MMR) pathway, and its pathogenesis is associated with microsatellite instability (MSI) caused by abnormalities in the core genes of the conventional MMR system. This cancer exhibits a distinct immune microenvironment, which makes [...] Read more.
MSI-H/dMMR endometrial cancer (EC) is closely linked to the mismatch repair (MMR) pathway, and its pathogenesis is associated with microsatellite instability (MSI) caused by abnormalities in the core genes of the conventional MMR system. This cancer exhibits a distinct immune microenvironment, which makes it suitable for treatment with immune checkpoint inhibitors (ICIs). This cancer type demonstrates heterogeneity, encompassing Lynch syndrome (LS)-associated EC (characterized by germline mutations), sporadic EC (attributed to MLH1 promoter hypermethylation), and Lynch-like EC (driven by somatic mutations). Research indicates that these three dMMR EC subtypes possess different immune microenvironments, which may influence the therapeutic efficacy of ICIs. However, the impact of somatic mutations in traditional MMR genes on EC has often been overlooked. Furthermore, over 50% of patients with MSI exhibit no response to ICIs, potentially due to abnormalities in nontraditional MMR genes. This review discusses the role of traditional and nontraditional MMR genes in dMMR EC and related treatment strategies, highlights key issues in the current diagnosis and treatment of dMMR EC, and aims to enhance understanding of its heterogeneity and advance precision diagnosis and treatment. Full article
(This article belongs to the Special Issue Human Reproductive Biology: Uncertainties and Controversies)
Show Figures

Figure 1

20 pages, 1388 KB  
Review
Mismatch Repair Deficiency and the Role of Non-Canonical Functions in Cancer: Diagnosis and Therapeutic Implications
by Alicja Dąbrowska, Jakub Mastalerz, Zofia Łapińska, Iwona Deszcz, Agnieszka Chwiłkowska and Nina Rembiałkowska
Int. J. Mol. Sci. 2025, 26(19), 9312; https://doi.org/10.3390/ijms26199312 - 24 Sep 2025
Cited by 1 | Viewed by 1494
Abstract
The mismatch repair system is critical for correcting base–base mismatches and insertion-deletion loops during DNA replication. Deficiencies in MMR (due to mutations in MLH1, MSH2, MSH6, or PMS2) lead to microsatellite instability and contribute to the development of various cancers, such as Lynch [...] Read more.
The mismatch repair system is critical for correcting base–base mismatches and insertion-deletion loops during DNA replication. Deficiencies in MMR (due to mutations in MLH1, MSH2, MSH6, or PMS2) lead to microsatellite instability and contribute to the development of various cancers, such as Lynch syndrome-related colorectal cancer and sporadic tumors. This review will delve into the molecular basis of MMR deficiency. Additionally, the review will cover diagnostic approaches for detecting MSI and MMR deficiency, including next-generation sequencing and PCR-based methods. The implications for treatment will be discussed, emphasizing immune checkpoint inhibitors (e.g., pembrolizumab) that target tumors with high mutational burdens due to MMR deficiency, as well as novel therapeutic approaches like synthetic lethality exploiting DNA repair vulnerabilities. Full article
(This article belongs to the Special Issue Targeted Therapies and Molecular Methods in Cancer, 3rd Edition)
Show Figures

Figure 1

Back to TopTop