1
|
Sriramadasu K, Ravichandran S, Li YH, Lai MT, Chiang AJ, Li CJ, Tsui KH, Chen CM, Chuang HH, Hwang T, Ding WY, Chung C, Chang CYY, Sheu JJC. Molecular evolution of driver mutations in cancer with microsatellite instability and their impact on tumor progression: Implications for precision medicine in patients with UCEC. Comput Biol Med 2025; 192:110275. [PMID: 40311467 DOI: 10.1016/j.compbiomed.2025.110275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 04/07/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
Cancer development is driven by genetic alterations, particularly cancer driver mutations (CDMs), which are associated with aggressive phenotypes and shorter survival. In contrast, higher mutation loads caused by microsatellite instability (MSI) or mismatch repair deficiency (MMRd) can induce anti-cancer immunity, leading to tumor shrinkage and improved responses to immune checkpoint inhibitor (ICI) therapies. However, understanding how CDMs and MSI/MMRd influence cancer evolution remains limited. We opted uterine corpus endometrial carcinoma (UCEC) as a model in this study due to its MSI-high/MMRd characteristics. Somatic mutation screening revealed that UCEC has a significantly higher mutation rate in cancer driver genes compared to ovarian cancer (OVCA) and cervical squamous cell carcinoma (CSCC), despite these cancers arising from histologically connected organs in the reproductive tract. Interestingly, these CDMs did not necessarily drive tumor progression. Using a cutoff of 7.0 (mutations/Mb) for tumor mutation burden (TMB), we classified UCEC patients into two groups with distinct clinical features, genetic profiles, and drug sensitivities. Among the known CDMs, TP53 mutations and their functional networks emerged as key drivers in UCEC progression, while mutations in CTNNB1, PTEN, and ARID1A may enhance anti-tumor immunity, correlating with longer overall survivals. Drug screening using GDSC and CTRPv2 databases suggested that GSK-3 inhibitor IX may be effective for treating aggressive UCEC patients with a non-MSI phenotype. Curcumin showed efficacy for UCEC patients with MSI, especially with ICI therapy. Our study highlights the importance of immune regulation and tolerance over CDMs in cancer development, particularly in those with an MSI-high/MMRd phenotype. We propose that TMB could serve as a valuable screening method alongside molecular and histopathological classifications to guide treatment strategies for UCEC patients.
Collapse
Affiliation(s)
- Kalpana Sriramadasu
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan
| | - Senthilkumar Ravichandran
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan; Department of Dermatology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Yau-Hong Li
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan; Department of Obstetrics and Gynecology, Pingtung Veterans General Hospital, Pingtung, 900053, Taiwan
| | - Ming-Tsung Lai
- Department of Pathology, Taichung Hospital, Ministry of Health and Welfare, Taichung, 403301, Taiwan
| | - An-Jen Chiang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan; Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan
| | - Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan; Institute of Biopharmaceutical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813414, Taiwan; Institute of Biopharmaceutical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan
| | - Chih-Mei Chen
- Genetics Center, China Medical University Hospital, Taichung, 404332, Taiwan
| | - Hsiang-Hao Chuang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan
| | - Tritium Hwang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan
| | - Wendy Yarou Ding
- Genetics Center, China Medical University Hospital, Taichung, 404332, Taiwan
| | - Ching Chung
- Genetics Center, China Medical University Hospital, Taichung, 404332, Taiwan
| | - Cherry Yin-Yi Chang
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, 404332, Taiwan; Department of Medicine, School of Medicine, China Medical University Hospital, Taichung, 404333, Taiwan.
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan; Institute of Biopharmaceutical Sciences, National Sun Yat-Sen University, Kaohsiung, 804201, Taiwan; School of Chinese Medicine, China Medical University, Taichung, 404333, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan.
| |
Collapse
|
2
|
Jain S. Does Schistosoma mansoni trigger colorectal cancer? Mol Biochem Parasitol 2025; 262:111672. [PMID: 39894059 DOI: 10.1016/j.molbiopara.2025.111672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
In this work the relationship between Schistosoma mansoni (Sm) and the induction and progression of colorectal cancer (CRC) is examined. Various clinical studies reviewed here yield inconsistent results, with some reporting no association between Sm infection and CRC and others suggesting a probable to strong association. Here we propose a number of plausible mechanisms whereby Sm infection might contribute to CRC induction and/or progression. These factors are (1) chronic inflammation, (2) exposure to parasite linked antigens and genotoxic products, especially soluble egg antigens (SEAs) and (3) alteration of the intestinal microbiota. These factors probably predispose humans towards CRC and can help in CRC progression however only widespread epidemiological, clinical and pathological studies can firmly establish their role or a complete lack of it.
Collapse
Affiliation(s)
- Sidhant Jain
- Institute for Globally Distributed Open Research and Education (IGDORE), India.
| |
Collapse
|
3
|
Soldati G, Saccardo C, Raniero D, De Leo D, Turrina S. Unveiling STRs instability in a colorectal cancer FFPE sample: a case report. Int J Legal Med 2025; 139:61-65. [PMID: 39377931 PMCID: PMC11732918 DOI: 10.1007/s00414-024-03341-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/20/2024] [Indexed: 10/09/2024]
Abstract
In forensic genetics, sometimes formalin-fixed paraffin-embedded (FFPE) biopsy material taken during life is the only biological sample available for individual identification or paternity testing. In most cases, this biological tissue is characterized by the presence of tumor cells characterized by instability and loss of heterozygosity of microsatellites (MSI/LOH) compared to the DNA present in cells of normal tissue.In this case report, two FFPE samples from the same male subject were available for genetic investigation: one sample with colorectal cancer tissue and the other with normal tissue (no cancerous histopathological features). The comparison of the genetic profiles obtained from DNA extracted from the two tissues showed in the tumor tissue the presence of three genomic instability phenomena affecting FGA, CSF1P0, D21S2055 loci, located on three distinct autosomal chromosomes, and one duplication phenomenon affecting the DYS438. Therefore, due to the MSI/LOH phenomena, the genetic profile acquired from the tumor tissue was distorted and thus generated a fictitious genetic profile, not corresponding to the subject's real one (normal tissue free of tumor cells).
Collapse
Affiliation(s)
- Giulia Soldati
- Department of Diagnostics and Public Health, Section of Forensic Medicine, Forensic Genetics Lab, University of Verona, Verona, Italy.
| | - Chiara Saccardo
- Department of Diagnostics and Public Health, Section of Forensic Medicine, Forensic Genetics Lab, University of Verona, Verona, Italy
| | - Dario Raniero
- Department of Diagnostics and Public Health, Section of Forensic Medicine, Forensic Genetics Lab, University of Verona, Verona, Italy
| | - Domenico De Leo
- Department of Diagnostics and Public Health, Section of Forensic Medicine, Forensic Genetics Lab, University of Verona, Verona, Italy
| | - Stefania Turrina
- Department of Diagnostics and Public Health, Section of Forensic Medicine, Forensic Genetics Lab, University of Verona, Verona, Italy
| |
Collapse
|
4
|
Duta-Ion SG, Juganaru IR, Hotinceanu IA, Dan A, Burtavel LM, Coman MC, Focsa IO, Zaruha AG, Codreanu PC, Bohiltea LC, Radoi VE. Redefining Therapeutic Approaches in Colorectal Cancer: Targeting Molecular Pathways and Overcoming Resistance. Int J Mol Sci 2024; 25:12507. [PMID: 39684219 DOI: 10.3390/ijms252312507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Colorectal cancer (CRC) arises through a combination of genetic and epigenetic alterations that affect key pathways involved in tumor growth and progression. This review examines the major molecular pathways driving CRC, including Chromosomal Instability (CIN), Microsatellite Instability (MSI), and the CpG Island Methylator Phenotype (CIMP). Key mutations in genes such as APC, KRAS, NRAS, BRAF, and TP53 activate signaling pathways like Wnt, EGFR, and PI3K/AKT, contributing to tumorigenesis and influencing responses to targeted therapies. Resistance mechanisms, including mutations that bypass drug action, remain challenging in CRC treatment. This review highlights the role of molecular profiling in guiding the use of targeted therapies such as tyrosine kinase inhibitors and immune checkpoint inhibitors. Novel combination treatments are also discussed as strategies to improve outcomes and overcome resistance. Understanding these molecular mechanisms is critical to advancing personalized treatment approaches in CRC and improving patient prognosis.
Collapse
Affiliation(s)
- Simona Gabriela Duta-Ion
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Ioana Ruxandra Juganaru
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Iulian Andrei Hotinceanu
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Andra Dan
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Livia Malina Burtavel
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Madalin Codrut Coman
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Ina Ofelia Focsa
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Andra Giorgiana Zaruha
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Patricia Christina Codreanu
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Laurentiu Camil Bohiltea
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- "Alessandrescu-Rusescu" National Institute for Maternal and Child Health, 20382 Bucharest, Romania
| | - Viorica Elena Radoi
- Department of Medical Genetics, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- "Alessandrescu-Rusescu" National Institute for Maternal and Child Health, 20382 Bucharest, Romania
| |
Collapse
|
5
|
Macagno M, Pessei V, Congiusta N, Lazzari L, Bellomo SE, Idrees F, Cavaliere A, Pietrantonio F, Raimondi A, Gusmaroli E, Zampino MG, Gervaso L, Ciardiello D, Mondello G, Santoro A, Personeni N, Bonoldi E, Aquilano MC, Valtorta E, Siena S, Sartore-Bianchi A, Amatu A, Bonazzina EF, Bencardino KB, Serini G, Marsoni S, Barault L, Di Nicolantonio F, Maione F. A Comparative Study of Methyl-BEAMing and Droplet Digital PCR for MGMT Gene Promoter Hypermethylation Detection. Diagnostics (Basel) 2024; 14:2467. [PMID: 39594133 PMCID: PMC11592929 DOI: 10.3390/diagnostics14222467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Background: O-6-methylguanine-DNA methyltransferase is responsible for the direct repair of O6-methylguanine lesions induced by alkylating agents, including temozolomide. O-6-methylguanine-DNA methyltransferase promoter hypermethylation is a well-established biomarker for temozolomide response in glioblastoma patients, also correlated with therapeutic response in colorectal cancer. Objectives: The ARETHUSA clinical trial aims to stratify colorectal cancer patients based on their mismatch repair status. Mismatch repair-deficient patients are eligible for treatment with immune checkpoint inhibitors (anti-PDL-1), whereas mismatch repair-proficient samples are screened for O-6-methylguanine-DNA methyltransferase promoter methylation to identify those suitable for temozolomide treatment. Methods: In this context, a subset of ARETHUSA metastatic colorectal cancer samples was used to compare two different techniques for assessing O-6-methylguanine-DNA methyltransferase hypermethylation: Methyl-BEAMing, a highly sensitive digital PCR approach that combines emulsion PCR and flow cytometry, and droplet digital PCR, a more automated procedure that enables the rapid, operator-independent analysis of a large number of samples. Results: Our study clearly demonstrates that the results obtained using Methyl-BEAMing and droplet digital PCR are comparable, with both techniques showing similar accuracy, sensitivity, and reproducibility. Conclusions: Digital droplet PCR proved to be an efficient method for detecting gene promoter methylation. However, the Methyl-BEAMing method has proved more sensitive for detecting low quantities of DNA.
Collapse
Affiliation(s)
- Marco Macagno
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy; (M.M.); (V.P.); (N.C.); (S.E.B.); (F.I.); (A.C.); (G.S.); (F.D.N.)
| | - Valeria Pessei
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy; (M.M.); (V.P.); (N.C.); (S.E.B.); (F.I.); (A.C.); (G.S.); (F.D.N.)
| | - Noemi Congiusta
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy; (M.M.); (V.P.); (N.C.); (S.E.B.); (F.I.); (A.C.); (G.S.); (F.D.N.)
- Department of Oncology, University of Torino, 10043 Orbassano, TO, Italy;
| | - Luca Lazzari
- IFOM ETS—The AIRC Institute of Molecolar Oncology, 20139 Milano, MI, Italy; (L.L.); (S.M.)
| | - Sara Erika Bellomo
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy; (M.M.); (V.P.); (N.C.); (S.E.B.); (F.I.); (A.C.); (G.S.); (F.D.N.)
| | - Fariha Idrees
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy; (M.M.); (V.P.); (N.C.); (S.E.B.); (F.I.); (A.C.); (G.S.); (F.D.N.)
- Department of Oncology, University of Torino, 10043 Orbassano, TO, Italy;
| | - Alessandro Cavaliere
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy; (M.M.); (V.P.); (N.C.); (S.E.B.); (F.I.); (A.C.); (G.S.); (F.D.N.)
- Department of Oncology, University of Torino, 10043 Orbassano, TO, Italy;
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS, Istituto Nazionale dei Tumori, 20133 Milan, MI, Italy; (F.P.); (A.R.); (E.G.)
| | - Alessandra Raimondi
- Department of Medical Oncology, Fondazione IRCCS, Istituto Nazionale dei Tumori, 20133 Milan, MI, Italy; (F.P.); (A.R.); (E.G.)
| | - Eleonora Gusmaroli
- Department of Medical Oncology, Fondazione IRCCS, Istituto Nazionale dei Tumori, 20133 Milan, MI, Italy; (F.P.); (A.R.); (E.G.)
| | - Maria Giulia Zampino
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IRCCS, 20139 Milano, MI, Italy; (M.G.Z.); (L.G.); (D.C.)
| | - Lorenzo Gervaso
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IRCCS, 20139 Milano, MI, Italy; (M.G.Z.); (L.G.); (D.C.)
| | - Davide Ciardiello
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IRCCS, 20139 Milano, MI, Italy; (M.G.Z.); (L.G.); (D.C.)
| | - Giuseppe Mondello
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy; (G.M.); (A.S.)
| | - Armando Santoro
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, 20089 Rozzano, MI, Italy; (G.M.); (A.S.)
| | | | - Emanuela Bonoldi
- Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162 Milano, MI, Italy; (E.B.); (M.C.A.); (E.V.)
| | - Maria Costanza Aquilano
- Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162 Milano, MI, Italy; (E.B.); (M.C.A.); (E.V.)
| | - Emanuele Valtorta
- Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162 Milano, MI, Italy; (E.B.); (M.C.A.); (E.V.)
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milano, MI, Italy; (S.S.); (A.S.-B.); (A.A.); (E.F.B.); (K.B.B.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milano, MI, Italy
| | - Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milano, MI, Italy; (S.S.); (A.S.-B.); (A.A.); (E.F.B.); (K.B.B.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milano, MI, Italy
| | - Alessio Amatu
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milano, MI, Italy; (S.S.); (A.S.-B.); (A.A.); (E.F.B.); (K.B.B.)
| | - Erica Francesca Bonazzina
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milano, MI, Italy; (S.S.); (A.S.-B.); (A.A.); (E.F.B.); (K.B.B.)
| | - Katia Bruna Bencardino
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milano, MI, Italy; (S.S.); (A.S.-B.); (A.A.); (E.F.B.); (K.B.B.)
| | - Guido Serini
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy; (M.M.); (V.P.); (N.C.); (S.E.B.); (F.I.); (A.C.); (G.S.); (F.D.N.)
- Department of Oncology, University of Torino, 10043 Orbassano, TO, Italy;
| | - Silvia Marsoni
- IFOM ETS—The AIRC Institute of Molecolar Oncology, 20139 Milano, MI, Italy; (L.L.); (S.M.)
| | - Ludovic Barault
- Department of Oncology, University of Torino, 10043 Orbassano, TO, Italy;
| | - Federica Di Nicolantonio
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy; (M.M.); (V.P.); (N.C.); (S.E.B.); (F.I.); (A.C.); (G.S.); (F.D.N.)
- Department of Oncology, University of Torino, 10043 Orbassano, TO, Italy;
| | - Federica Maione
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy; (M.M.); (V.P.); (N.C.); (S.E.B.); (F.I.); (A.C.); (G.S.); (F.D.N.)
- Department of Oncology, University of Torino, 10043 Orbassano, TO, Italy;
| |
Collapse
|
6
|
Peter BC, Mansour M, Prasad K, Jha T, Venkatesan S, Jha M. Compliance of a Tertiary Centre With Molecular Testing Strategies for Lynch Syndrome in Colorectal Cancer. Cureus 2024; 16:e73475. [PMID: 39529921 PMCID: PMC11554376 DOI: 10.7759/cureus.73475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Lynch syndrome is one of the most common hereditary cancers associated with germline alterations of DNA mismatch repair genes. Recent advances have shed light on its molecular pathogenesis, leading to the development of various testing strategies. The aim of this study is to evaluate whether all colorectal adenocarcinomas undergo immunohistochemistry (IHC) and microsatellite instability (MSI) testing to identify potential Lynch syndrome patients. Methods This study evaluated the compliance of MSI and IHC testing for Lynch syndrome in a tertiary cancer research centre. All patients undergoing colorectal cancer resection between April 2022 and December 2022 were identified from a prospectively maintained register. The histology, MSI, and IHC testing reports for these patients were recorded to check if they were done and reported. Only patients with colorectal adenocarcinoma were included in this study. Results A total of 314 patients had colorectal cancer resections. A total of 301 were included in the study, with a median age of 65 years (IQR = 35-95). Thirteen (4.14%) patients did not have MSI and IHC testing reported on the system. Of these, eight (61.53%) did not have an MSI/IHC testing request, four (30.76%) did not have sufficient specimens to be sent for further testing, one (7.69%) had MSI and IHC testing but the final report was yet to be authorized. Out of the eight who did not have the testing done, three had early polyp cancer and one had a goblet cell adenocarcinoma. Conclusion According to the data, our hospital has a 97.45% compliance with current guidelines, not considering the insufficient sample for testing and the authorization of the final report. A more comprehensive proforma may be needed to provide a feedback loop for further MSI and IHC testing when entering data into the system. Further auditing is required to check the effectiveness of the proforma in achieving complete compliance.
Collapse
Affiliation(s)
- Bennett C Peter
- General Surgery, James Cook University Hospital, Middlesbrough, GBR
| | | | - Kolanu Prasad
- Pathology, James Cook University Hospital, Middlesbrough, GBR
| | - Trisha Jha
- Medicine and Surgery, Keele University School of Medicine, Newcastle-under-Lyme, GBR
| | | | - Madan Jha
- Colorectal Surgery, James Cook University Hospital, Middlesbrough, GBR
| |
Collapse
|
7
|
Wu J, Wang H, Yang R, Liu D, Li W. Prognostic significance of the modified Glasgow Prognostic Score in NSCLC patients undergoing immune checkpoint inhibitor therapy: a meta-analysis. Front Oncol 2024; 14:1449853. [PMID: 39464717 PMCID: PMC11502296 DOI: 10.3389/fonc.2024.1449853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/25/2024] [Indexed: 10/29/2024] Open
Abstract
Background The modified Glasgow Prognosis Score (mGPS), which considers both inflammatory response and nutritional status, has been linked to the prognosis of various tumors. The relationship between mGPS and non-small cell lung cancer (NSCLC) patients receiving immune checkpoint inhibitors (ICIs) is still a subject of debate. This meta-analysis aims to comprehensively assess the association between mGPS and survival in NSCLC treated with ICIs. Methods A thorough review of studies from PubMed, Web of Science, Scopus, and Embase was conducted up to June 4, 2024. Fixed-effect or random-effect models were employed, combining hazard ratios (HRs) and 95% confidence intervals (CI), to assess the prognostic value of mGPS for OS and PFS in patients with NSCLC receiving immunotherapy. Results A total of 1,022 patients from 11 studies were recruited. Combined results showed that mGPS elevation was significantly associated with poor OS (HR = 1.63, 95%CI: 1.42-1.87, P < 0.01) and PFS (HR = 1.71, 95%CI: 1.31-2.24, P < 0.01). Subgroup analysis and sensitivity analysis further determined the predictive effect of elevated mGPS on OS and PFS deterioration in NSCLC patients receiving immunotherapy. Conclusion mGPS can be used as a good noninvasive biomarker to demonstrate prognostic and clinical significance in patients with NSCLC undergoing immunotherapy. Systematic review registration http://www.crd.york.ac.uk/prospero/ PROSPERO, identifier CRD42023432661.
Collapse
Affiliation(s)
- Jiaxuan Wu
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
- Institute of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Haoyu Wang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
- Institute of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruiyuan Yang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
- Institute of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan Liu
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
- Institute of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
- Institute of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Jong KXJ, Mohamed EHM, Syafruddin SE, Faruqu FN, Vellasamy KM, Ibrahim K, Ibrahim ZA. IL-8 and PI3K pathway influence the susceptibility of TRAIL-sensitive colorectal cancer cells to TRAIL-induced cell death. Mol Biol Rep 2024; 51:978. [PMID: 39269555 DOI: 10.1007/s11033-024-09895-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is an apoptosis inducer that exhibits an ideal therapeutic safety profile with less adverse effects than conventional chemotherapy. However, the occurrence of TRAIL resistance has been reported in various cancers including colorectal cancer (CRC). Substantial efforts have been channelled towards managing TRAIL resistance including identifying molecular targets. Interleukins (ILs) have been recently shown to play critical roles in modulating TRAIL sensitivity in cancer cells. METHODS AND RESULTS This study investigated the roles of two ILs, IL-8 and IL⍺, in TRAIL resistance in CRC. TRAIL-resistant HT-29 and TRAIL-sensitive HCT 116 cells, were treated with human recombinant IL-8 and IL-1⍺. The results indicated that treatment with IL-8 (2.5 ng/mL) significantly protected TRAIL-sensitive HCT 116 cells from TRAIL-induced cell death (p < 0.05). However, IL-1⍺ did not play a role in modulating CRC cells' responses to TRAIL. Data from RT-qPCR and Western blotting revealed the molecular regulations of IL-8 on TRAIL decoy receptor genes (OPG) and autophagy-related genes (BECN1 and LC3B) expression. The activation of the phosphoinositide 3-kinase (PI3K) pathway was shown to counteract TRAIL-induced cell death. By inhibiting its activation with wortmannin, the protective role of IL-8 against TRAIL treatment was reversed, suggesting the involvement of the PI3K pathway. CONCLUSION Collectively, findings from this study identified the role of IL-8 and PI3K in modulating CRC cells' sensitivity to TRAIL. Further validation of these two potential molecular targets is warranted to overcome TRAIL resistance in CRC.
Collapse
Affiliation(s)
- Kelly Xue Jing Jong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | | | - Saiful Effendi Syafruddin
- UKM Medical Molecular Biology Institute (UMBI), UKM Medical Centre, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Farid Nazer Faruqu
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Kumutha Malar Vellasamy
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Kamariah Ibrahim
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Zaridatul Aini Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia.
| |
Collapse
|
9
|
Bodalal Z, Hong EK, Trebeschi S, Kurilova I, Landolfi F, Bogveradze N, Castagnoli F, Randon G, Snaebjornsson P, Pietrantonio F, Lee JM, Beets G, Beets-Tan R. Non-invasive CT radiomic biomarkers predict microsatellite stability status in colorectal cancer: a multicenter validation study. Eur Radiol Exp 2024; 8:98. [PMID: 39186200 PMCID: PMC11347521 DOI: 10.1186/s41747-024-00484-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 05/30/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Microsatellite instability (MSI) status is a strong predictor of response to immunotherapy of colorectal cancer. Radiogenomic approaches promise the ability to gain insight into the underlying tumor biology using non-invasive routine clinical images. This study investigates the association between tumor morphology and the status of MSI versus microsatellite stability (MSS), validating a novel radiomic signature on an external multicenter cohort. METHODS Preoperative computed tomography scans with matched MSI status were retrospectively collected for 243 colorectal cancer patients from three hospitals: Seoul National University Hospital (SNUH); Netherlands Cancer Institute (NKI); and Fondazione IRCCS Istituto Nazionale dei Tumori, Milan Italy (INT). Radiologists delineated primary tumors in each scan, from which radiomic features were extracted. Machine learning models trained on SNUH data to identify MSI tumors underwent external validation using NKI and INT images. Performances were compared in terms of area under the receiving operating curve (AUROC). RESULTS We identified a radiomic signature comprising seven radiomic features that were predictive of tumors with MSS or MSI (AUROC 0.69, 95% confidence interval [CI] 0.54-0.84, p = 0.018). Integrating radiomic and clinical data into an algorithm improved predictive performance to an AUROC of 0.78 (95% CI 0.60-0.91, p = 0.002) and enhanced the reliability of the predictions. CONCLUSION Differences in the radiomic morphological phenotype between tumors MSS or MSI could be detected using radiogenomic approaches. Future research involving large-scale multicenter prospective studies that combine various diagnostic data is necessary to refine and validate more robust, potentially tumor-agnostic MSI radiogenomic models. RELEVANCE STATEMENT Noninvasive radiomic signatures derived from computed tomography scans can predict MSI in colorectal cancer, potentially augmenting traditional biopsy-based methods and enhancing personalized treatment strategies. KEY POINTS Noninvasive CT-based radiomics predicted MSI in colorectal cancer, enhancing stratification. A seven-feature radiomic signature differentiated tumors with MSI from those with MSS in multicenter cohorts. Integrating radiomic and clinical data improved the algorithm's predictive performance.
Collapse
Affiliation(s)
- Zuhir Bodalal
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Eun Kyoung Hong
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
- Seoul National University Hospital, Seoul, South Korea
| | - Stefano Trebeschi
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Ieva Kurilova
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Federica Landolfi
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Radiology Unit, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Nino Bogveradze
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
- Department of Radiology, American Hospital Tbilisi, Tbilisi, Georgia
| | - Francesca Castagnoli
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Radiology, Royal Marsden Hospital, London, UK
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Giovanni Randon
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Petur Snaebjornsson
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
- Oncology and Hemato-oncology Department, University of Milan, Milan, Italy
| | - Jeong Min Lee
- Seoul National University Hospital, Seoul, South Korea
| | - Geerard Beets
- GROW Research Institute for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Regina Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- GROW Research Institute for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
10
|
Milan N, Navarria F, Cecchin E, De Mattia E. Somatic pharmacogenomics in the treatment prognosis of locally advanced rectal cancer patients: a narrative review of the literature. Expert Rev Clin Pharmacol 2024; 17:683-719. [PMID: 39046146 DOI: 10.1080/17512433.2024.2375449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Standard treatment for patients with locally advanced rectal cancer (LARC) includes neoadjuvant chemoradiotherapy (nCRT) with fluoropyrimidines, followed by surgical excision. The newly introduced therapeutic strategies propose intensified regimens or more conservative approaches based on risk stratification algorithms that currently include clinicoradiological criteria but not molecular variables. How to better stratify patients is a burning clinical question, and pharmacogenomics may prove useful in identifying new genetic markers that could be incorporated into clinical algorithms to personalize nCRT. An emerging area could be the evaluation of somatic mutations as potential genetic markers that correlate with patient prognosis. Tumor mutations in the RAS/BRAF genes, as well as microsatellite instability (MSI) status, are currently used in treatment selection for colorectal cancer (CRC); however, their clinical value in LARC is still unclear. AREA COVERED This literature review discusses the relevant findings on the prognostic role of mutations in the key oncogenes RAS, KRAS, BRAF, PIK3CA, SMAD4 and TP53, including MSI status in LARC patients treated with nCRT. EXPERT OPINION KRAS proved to be the most promising marker, consistently associated with poorer disease-free survival and overall survival. Therefore, KRAS could be a good candidate for integration into the risk stratification algorithm to develop a personalized treatment.
Collapse
Affiliation(s)
- Noemi Milan
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Federico Navarria
- Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Erika Cecchin
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Elena De Mattia
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| |
Collapse
|
11
|
Guo T, Zhao S, Zhu W, Zhou H, Cheng H. Research progress on the biological basis of Traditional Chinese Medicine syndromes of gastrointestinal cancers. Heliyon 2023; 9:e20653. [PMID: 38027682 PMCID: PMC10643116 DOI: 10.1016/j.heliyon.2023.e20653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 09/23/2023] [Accepted: 10/03/2023] [Indexed: 12/01/2023] Open
Abstract
Gastrointestinal cancers account for 11.6 % of all cancers, and are the second most frequently diagnosed type of cancer worldwide. Traditional Chinese medicine (TCM), together with Western medicine or alone, has unique advantages for the prevention and treatment of cancers, including gastrointestinal cancers. Syndrome differentiation and treatment are basic characteristics of the theoretical system of TCM. TCM syndromes are the result of the differentiation of the syndrome and the basis of treatment. Genomics, transcriptomics, proteomics, metabolomics, intestinal microbiota, and serology, generated around the central law, are used to study the biological basis of TCM syndromes in gastrointestinal cancers. This review summarizes current research on the biological basis of TCM syndrome in gastrointestinal cancers and provides useful references for future research on TCM syndrome in gastrointestinal cancers.
Collapse
Affiliation(s)
- Tianhao Guo
- Institute of Health and Regimen, Jiangsu Open University, Nanjing, Jiangsu 210036, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu 210023, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Shuoqi Zhao
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu 210023, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Wenjian Zhu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu 210023, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Hongguang Zhou
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu 210023, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
- Departments of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, Jiangsu 210023, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
- Departments of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| |
Collapse
|
12
|
Sharma A, Raphael V, Lyngdoh BS, Harris C, Jagtap VK. Role of Mismatch Repair Deficiency Status and Microsatellite Instability in Relation to the Expression of Immune Checkpoint Proteins in Colorectal Cancer. Cureus 2023; 15:e43571. [PMID: 37719521 PMCID: PMC10503400 DOI: 10.7759/cureus.43571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
Introduction Colorectal cancer (CRC) is the third most common cancer in the world among men and second among women worldwide. One of the major molecular pathways responsible for the development of colorectal cancer (CRC) is the microsatellite instability (MSI) pathway. During carcinogenesis, the tumor cells express programmed death ligand-1 (PD-L1), which reduces the immunogenicity leading to the escape of immune attack. Anti-PD-L1 interaction is an upcoming line of research for the treatment of colorectal carcinoma patients. Materials and methods The present study was an ambispective study where the mismatch repair deficiency status (MMR) and programmed death ligand-1 (PD-L1) expression were studied using immunohistochemistry and then later analyzed and compared with the clinicopathological parameters and MSI status in relation to the expression of programmed death ligand-1 (PD-L1) in neoplastic and immune cells in a total of 55 biopsy specimen. MMR expression was reported as retained or loss of nuclear staining, and PD-L1 expression was taken as positive with a cut-off of more than or equal to 5% membranous positivity in both tumor cells and immune cells. Results The analysis showed a significant correlation of microsatellite instability (MSI) status with two of the clinicopathological parameters, which were the site of the tumor (p-value<0.001) and M stage (p-value<0.001). PD-L1 expression in neoplastic cells showed no significant correlation with the clinicopathological parameters, whereas PD-L1 expression in immune cells showed a significant association with gender (p-value=0.043). Also, MSI status showed a significant association with PD-L1 expression in tumor cells (p-value <0.001).
Collapse
Affiliation(s)
- Antariksha Sharma
- Pathology, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences, Shillong, IND
| | - Vandana Raphael
- Pathology, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences, Shillong, IND
| | - Bifica S Lyngdoh
- Pathology, All India Institute of Medical Sciences, Guwahati, IND
| | - Caleb Harris
- Surgical Oncology, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences, Shillong, IND
| | - Vikas K Jagtap
- Radiation Oncology, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences, Shillong, IND
| |
Collapse
|
13
|
Jeanjean SI, Renault V, Daunay A, Shen Y, Hardy LM, Deleuze JF, How-Kit A. LT-RPA: An Isothermal DNA Amplification Approach for Improved Microsatellite Genotyping and Microsatellite Instability Detection. Methods Mol Biol 2023; 2621:91-109. [PMID: 37041442 DOI: 10.1007/978-1-0716-2950-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Microsatellites are short tandem repeats of one to six nucleotides that are highly polymorphic and extensively used as genetic markers in numerous biomedical applications, including the detection of microsatellite instability (MSI) in cancer. The standard analytical method for microsatellite analysis relies on PCR amplification followed by capillary electrophoresis or, more recently, next-generation sequencing (NGS). However, their amplification during PCR generates undesirable frameshift products known as stutter peaks caused by polymerase slippage, complicating data analysis and interpretation, while very few alternative methods for microsatellite amplification have been developed to reduce the formation of these artifacts. In this context, the recently developed low-temperature recombinase polymerase amplification (LT-RPA) is an isothermal DNA amplification method at low temperature (32 °C) that drastically reduces and sometimes completely abolishes the formation of stutter peaks. LT-RPA greatly simplifies the genotyping of microsatellites and improves the detection of MSI in cancer. In this chapter, we describe in detail all the experimental steps necessary for the development of LT-RPA simplex and multiplex assays for microsatellite genotyping and MSI detection, including the design, optimization, and validation of the assays combined with capillary electrophoresis or NGS.
Collapse
Affiliation(s)
- Sophie I Jeanjean
- School of Biology, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Victor Renault
- Laboratoire de Bio-informatique Clinique, Institut Curie, Paris, France
| | - Antoine Daunay
- School of Biology, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Yimin Shen
- School of Biology, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Lise M Hardy
- School of Biology, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
- Laboratory of Excellence GenMed, Paris, France
| | - Jean-François Deleuze
- School of Biology, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
- Laboratory of Excellence GenMed, Paris, France
- Centre National de Recherche en Génomique Humaine, CEA- Institut François Jacob, Evry, France
| | - Alexandre How-Kit
- School of Biology, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand.
| |
Collapse
|
14
|
Tian Y, Wang J, Wen Q, Gao A, Huang A, Li R, Zhang Y, Su G, Sun Y. The Significance of Tumor Microenvironment Score for Breast Cancer Patients. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5673810. [PMID: 35528180 PMCID: PMC9071896 DOI: 10.1155/2022/5673810] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/16/2022] [Indexed: 12/30/2022]
Abstract
Purpose This study was designed to clarify the prognostic value of tumor microenvironment score and abnormal genomic alterations in TME for breast cancer patients. Method The TCGA-BRCA data were downloaded from TCGA and analyzed with R software. The results from analyses were further validated using the dataset from GSE96058, GSE124647, and GSE25066. Results After analyzing the TCGA data and verifying it with the GEO data, we developed a TMEscore model based on the TME infiltration pattern and validated it in 3273 breast cancer patients. The results suggested that our TMEscore model has high prognostic value. TME features with the TMEscore model can help to predict breast cancer patients' response to immunotherapy and provide new strategies for breast cancer treatment. Signature 24 was first found in breast cancer. In focal SCNAs, a total of 95 amplified genes and 169 deletion genes in the TMEscore high group were found to be significantly related to the prognosis of breast cancer patients, while 61 amplified genes and 174 deletion genes in the TMEscore low group were identified. LRRC48, CFAP69, and cg25726128 were first discovered and reported to be related to the survival of breast cancer patients. We identified specific mutation signatures that correlate with TMEscore and prognosis. Conclusion TMEscore model has high predictive value regarding prognosis and patients' response to immunotherapy. Signature 24 was first found in breast cancer. Specific mutation signatures that correlate with TMEscore and prognosis might be used for providing additional indicators for disease evaluation.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013 Shandong, China
- Somatic Radiotherapy Department, Shandong Second Provincial General Hospital, Shandong Provincial ENT Hospital, Jinan, Shandong 250023, China
| | - Jingnan Wang
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013 Shandong, China
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| | - Qing Wen
- Jinan Clinical Research Center of Shandong First Medical University, Jinan, China
| | - Aiqin Gao
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013 Shandong, China
| | - Alan Huang
- Department of Oncology, Jinan Central Hospital, The Hospital Affiliated with Shandong First Medical University, Jinan, Shandong 250013, China
| | - Ran Li
- Department of Oncology, Jinan Central Hospital, Weifang Medical University, Weifang, 261053 Shandong, China
| | - Ye Zhang
- Department of Oncology, Jinan Central Hospital, Weifang Medical University, Weifang, 261053 Shandong, China
| | - Guohai Su
- Department of Cardiovascular Diseases, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013 Shandong, China
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013 Shandong, China
- Phase I Clinical Trial Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250012, China
| |
Collapse
|
15
|
Qureshi S, Chan N, George M, Ganesan S, Toppmeyer D, Omene C. Immune Checkpoint Inhibitors in Triple Negative Breast Cancer: The Search for the Optimal Biomarker. Biomark Insights 2022; 17:11772719221078774. [PMID: 35221668 PMCID: PMC8874164 DOI: 10.1177/11772719221078774] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/04/2022] [Indexed: 12/14/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a high-risk and aggressive malignancy characterized by the absence of estrogen receptors (ER) and progesterone receptors (PR) on the surface of malignant cells, and by the lack of overexpression of human epidermal growth factor 2 (HER2). It has limited therapeutic options compared to other subtypes of breast cancer. There is now a growing body of evidence on the role of immunotherapy in TNBC, however much of the data from clinical trials is conflicting and thus, challenging for clinicians to integrate the data into clinical practice. Landmark phase III trials using immunotherapy in the early-stage neoadjuvant setting concluded that the addition of immunotherapy to chemotherapy improved the pathologic complete response (pCR) rate compared to chemotherapy with placebo while others found no significant improvement in pCR. Phase III trials have investigated the utility of immunotherapy in previously untreated metastatic TNBC, and these studies have similarly arrived at inconsistent conclusions. Some studies showed no benefit while others demonstrated a clinically significant improvement in overall survival in the PD-L1 positive population. It is not yet clear which biomarkers are most useful, and assays for these biomarkers have not been standardized. Given the often serious and severe side effects of immunotherapy, it is important and necessary to identify predictive biomarkers of response and resistance in order to enhance patient selection. In this review, we will discuss both the challenges of traditional biomarkers and the opportunities of emerging biomarkers for patient selection.
Collapse
Affiliation(s)
- Sadaf Qureshi
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Nancy Chan
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Mridula George
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Shridar Ganesan
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Deborah Toppmeyer
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Coral Omene
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
16
|
Manzoor S, Saber-Ayad M, Maghazachi AA, Hamid Q, Muhammad JS. MLH1 mediates cytoprotective nucleophagy to resist 5-Fluorouracil-induced cell death in colorectal carcinoma. Neoplasia 2022; 24:76-85. [PMID: 34952246 PMCID: PMC8695220 DOI: 10.1016/j.neo.2021.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 01/01/2023]
Abstract
Colorectal Cancer (CRC) with Microsatellite instability (MSI) and mutLhomolog-1 (MLH1) gene deficiency are less aggressive than MLH1 proficient cancers. MLH1 is involved in several cellular processes, but its connection with the autophagy-dependent cellular response towards anticancer drugs remains unclear. In this study, we aimed to investigate the interaction between MLH1 and the autophagy marker LC3, which facilitated nucleophagy induction, and its potential role in determining sensitivity to 5-Fluorouracil (5-FU) induced cell death. To examine the role of MLH1 in DNA-damage-induced nucleophagy in CRC cells, we utilized a panel of MLH1 deficient and MLH1 proficient CRC cell lines. We included a parental HCT116 cell line (MLH1-/-) and its isogenic cell line HCT116 MLH1+/- in which a single allele of the MLH1 gene was introduced using CRISPR-Cas9 gene editing. We observed that MLH1 proficient cells were less sensitive to the 5-FU-induced cytotoxic effect. The 5-FU induced DNA damage led to LC3 up-regulation, which was dependent on MLH1 overexpression. Moreover, immunofluorescence and immunoprecipitation data showed LC3 and MLH1 were colocalized in CRC cells. Consequently, MLH1 dependent 5-FU-induced DNA damage contributed to the formation of micronuclei. These micronuclei colocalize with autolysosome, indicating a cytoprotective role of MLH1 dependent nucleophagy. Interestingly, siRNA knockdown of MLH1 in HCT116 MLH1+/- prevented LC3 upregulation and micronuclei formation. These novel data are the first to show an essential role of MLH1 in mediating the chemoresistance and survival of cancer cells by increasing the LC3 expression and inducing nucleophagy in 5-FU treated CRC cells.
Collapse
Affiliation(s)
- Shaista Manzoor
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Maha Saber-Ayad
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Azzam A Maghazachi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Qutayba Hamid
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Meakins-Christie Laboratories, Research Institute of the McGill University Health Center, Montreal, QC H4A 3J1, Canada
| | - Jibran Sualeh Muhammad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
| |
Collapse
|
17
|
Frias-Gomes C, Sousa AC, Rolim I, Henriques AR, Branco F, Janeiro A, Malveiro S, Dário AR, Oliveira MH, Borralho P, Teixeira JA, Faria A, Maio R, Fonseca I, Cravo M. Phenotype-Genotype Correlation in Colorectal Cancer: A Real-Life Study. GE PORTUGUESE JOURNAL OF GASTROENTEROLOGY 2022; 29:13-21. [PMID: 35111960 PMCID: PMC8787526 DOI: 10.1159/000516009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/06/2021] [Indexed: 09/26/2023]
Abstract
BACKGROUND AND AIMS Colorectal cancer (CRC) is a heterogeneous disease with distinctive genetic pathways, such as chromosomal instability, microsatellite instability and methylator pathway. Our aim was to correlate clinical and genetic characteristics of CRC patients in order to understand clinical implications of tumour genotype. METHODS Single-institution retrospective cohort of patients who underwent curative surgery for CRC, from 2012 to 2014. RAS and BRAF mutations were evaluated with the real-time PCR technique Idylla®. Mismatch repair deficiency (dMMR) was characterized by absence of MLH1, MSH6, MSH2 and/or PMS2 expression, evaluated by tissue microarrays. Overall survival (OS) and disease-free survival (DFS) were assessed using survival analysis. RESULTS Overall, 242 patients were included (males 57.4%, age 69.3 ± 12.9 years; median follow-up 49 months). RAS-mutated tumours were associated with reduced DFS (p = 0.02) and OS (p = 0.045) in stage I-III CRC. BRAF-mutated tumours were more predominant in females and in the right colon, similarly to dMMR tumours. BRAF status did not influence OS (4 years)/DFS (3.5 years) in stage I-III disease. However, after relapse, length of survival was 3.5 months in BRAF-mutated tumours in contrast to 18.6 months in BRAF wild-type tumours (p = NS). No germline mutations in mismatch repair genes were so far identified in the patients with dMMR tumours. Molecular phenotype (RAS, BRAF and MMR) did not influence OS in metastatic patients. Our small sample size may be a limitation of the study. CONCLUSION In our cohort, RAS-mutated tumours were associated with worse DFS and OS in early-stage CRC, whereas the remaining molecular variables had no prognostic influence.
Collapse
Affiliation(s)
- Catarina Frias-Gomes
- Serviço de Gastrenterologia, Departamento de Cirurgia, Hospital Beatriz Ângelo, Loures, Portugal
| | | | - Inês Rolim
- Serviço de Anatomia Patológica, Instituto Português de Oncologia Francisco Gentil, Lisboa, Portugal
- Instituto de Anatomia Patológica, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Raquel Henriques
- Instituto de Anatomia Patológica, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Francisco Branco
- Serviço de Oncologia Médica, Departamento de Oncologia, Hospital Beatriz Ângelo, Loures, Portugal
| | - André Janeiro
- Genomed, Instituto de Medicina Molecular, Lisboa, Portugal
| | - Sara Malveiro
- Genomed, Instituto de Medicina Molecular, Lisboa, Portugal
| | - Ana Rita Dário
- Genomed, Instituto de Medicina Molecular, Lisboa, Portugal
| | | | - Paula Borralho
- Instituto de Anatomia Patológica, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Serviço de Anatomia Patológica, Hospital CUF Descobertas, Lisboa, Portugal
| | - José Alberto Teixeira
- Serviço de Oncologia Médica, Departamento de Oncologia, Hospital Beatriz Ângelo, Loures, Portugal
| | - Ana Faria
- Serviço de Oncologia Médica, Departamento de Oncologia, Hospital Beatriz Ângelo, Loures, Portugal
| | - Rui Maio
- Serviço de Cirurgia Geral, Departamento de Cirurgia, Hospital Beatriz Ângelo, Loures, Portugal
| | - Isabel Fonseca
- Serviço de Anatomia Patológica, Instituto Português de Oncologia Francisco Gentil, Lisboa, Portugal
- Instituto de Anatomia Patológica, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Marília Cravo
- Serviço de Gastrenterologia, Departamento de Cirurgia, Hospital Beatriz Ângelo, Loures, Portugal
| |
Collapse
|
18
|
Assessment of Microsatellite Instability from Next-Generation Sequencing Data. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1361:75-100. [DOI: 10.1007/978-3-030-91836-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
19
|
Venetis K, Fusco N, Sajjadi E. Commentary: Mismatch Repair Deficiency and Microsatellite Instability in Triple-Negative Breast Cancer: A Retrospective Study of 440 Patients. Front Oncol 2021; 11:735476. [PMID: 34660298 PMCID: PMC8511768 DOI: 10.3389/fonc.2021.735476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Affiliation(s)
- Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
20
|
Zhang C, Ding J, Xu X, Liu Y, Huang W, Da L, Ma Q, Chen S. Tumor Microenvironment Characteristics of Pancreatic Cancer to Determine Prognosis and Immune-Related Gene Signatures. Front Mol Biosci 2021; 8:645024. [PMID: 34169093 PMCID: PMC8217872 DOI: 10.3389/fmolb.2021.645024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/30/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Pancreatic cancer (PC) is one of the most lethal types of cancer with extremely poor diagnosis and prognosis, and the tumor microenvironment plays a pivotal role during PC progression. Poor prognosis is closely associated with the unsatisfactory results of currently available treatments, which are largely due to the unique pancreatic tumor microenvironment (TME). Methods: In this study, a total of 177 patients with PC from The Cancer Genome Atlas (TCGA) cohort and 65 patients with PC from the GSE62452 cohort in Gene Expression Omnibus (GEO) were included. Based on the proportions of 22 types of infiltrated immune cell subpopulations calculated by cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT), the TME was classified by K-means clustering and differentially expressed genes (DEGs) were determined. A combination of the elbow method and the gap statistic was used to explore the likely number of distinct clusters in the data. The ConsensusClusterPlus package was utilized to identify radiomics clusters, and the samples were divided into two subtypes. Result: Survival analysis showed that the patients with TMEscore-high phenotype had better prognosis. In addition, the TMEscore-high had better inhibitory effect on the immune checkpoint. A total of 10 miRNAs, 311 DEGs, and 68 methylation sites related to survival were obtained, which could be biomarkers to evaluate the prognosis of patients with pancreatic cancer. Conclusions: Therefore, a comprehensive description of TME characteristics of pancreatic cancer can help explain the response of pancreatic cancer to immunotherapy and provide a new strategy for cancer treatment.
Collapse
Affiliation(s)
- Congjun Zhang
- Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jun Ding
- Department of Hepatopancreatobiliary Surgery, Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao Xu
- Department of Oncology, Xintai People's Hospital, Xintai, China
| | - Yangyang Liu
- Department of Oncology, Xintai People's Hospital, Xintai, China
| | - Wei Huang
- Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liangshan Da
- Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qiang Ma
- Department of Oncology, Xintai People's Hospital, Xintai, China
| | - Shengyang Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
Sajjadi E, Venetis K, Piciotti R, Invernizzi M, Guerini-Rocco E, Haricharan S, Fusco N. Mismatch repair-deficient hormone receptor-positive breast cancers: Biology and pathological characterization. Cancer Cell Int 2021; 21:266. [PMID: 34001143 PMCID: PMC8130151 DOI: 10.1186/s12935-021-01976-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/07/2021] [Indexed: 12/15/2022] Open
Abstract
The clinical outcome of patients with a diagnosis of hormone receptor (HR)+ breast cancer has improved remarkably since the arrival of endocrine therapy. Yet, resistance to standard treatments is a major clinical challenge for breast cancer specialists and a life-threatening condition for the patients. In breast cancer, mismatch repair (MMR) status assessment has been demonstrated to be clinically relevant not only in terms of screening for inherited conditions such as Lynch syndrome, but also for prognostication, selection for immunotherapy, and early identification of therapy resistance. Peculiar traits characterize the MMR biology in HR+ breast cancers compared to other cancer types. In these tumors, MMR genetic alterations are relatively rare, occurring in ~3 % of cases. On the other hand, modifications at the protein level can be observed also in the absence of gene alterations and vice versa. In HR+ breast cancers, the prognostic role of MMR deficiency has been confirmed by several studies, but its predictive value remains a matter of controversy. The characterization of MMR status in these patients is troubled by the lack of tumor-specific guidelines and/or companion diagnostic tests. For this reason, precise identification of MMR-deficient breast cancers can be problematic. A deeper understanding of the MMR biology and clinical actionability in HR+ breast cancer may light the path to effective tumor-specific diagnostic tools. For a precise MMR status profiling, the specific strengths and limitations of the available technologies should be taken into consideration. This article aims at providing a comprehensive overview of the current state of knowledge of MMR alterations in HR+ breast cancer. The available armamentarium for MMR testing in these tumors is also examined along with possible strategies for a tailored pathological characterization.
Collapse
Affiliation(s)
- Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Roberto Piciotti
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Marco Invernizzi
- Physical and Rehabilitative Medicine, Department of Health Sciences, University of Eastern Piedmont, Viale Piazza D'Armi, 1, 28100, Novara, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Svasti Haricharan
- Department of Tumor Microenvironment and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, 92037, La Jolla, CA, USA
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141, Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy.
| |
Collapse
|
22
|
Sun SY, Hu XT, Yu XF, Zhang YY, Liu XH, Liu YH, Wu SH, Li YY, Cui SX, Qu XJ. Nuclear translocation of ATG5 induces DNA mismatch repair deficiency (MMR-D)/microsatellite instability (MSI) via interacting with Mis18α in colorectal cancer. Br J Pharmacol 2021; 178:2351-2369. [PMID: 33645631 DOI: 10.1111/bph.15422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 01/30/2021] [Accepted: 02/15/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE It is well known that microsatellite instability-high (MSI-H) is associated with 5-fluorouracil (5-FU) resistance in colorectal cancer. MSI-H is the phenotype of DNA mismatch repair deficiency (MMR-D), mainly occurring due to hypermethylation of MLH1 promoter CpG island. However, the mechanisms of MMR-D/MSI-H are unclear. We aim to investigate the pathway of MMR-D/MSI-H involved in 5-FU resistance. EXPERIMENTAL APPROACH Human colorectal cancer specimens were diagnosed for MSI-H by immunohistochemistry and western blotting. Proteome microarray interactome assay was performed to screen nuclear proteins interacting with ATG5. Nuclear ATG5 and ATG5-Mis18α overexpression were analysed in ATG5high colorectal cancer bearing mice. The methylation assay determined the hypermethylation of hMLH1 promoter CpG island in freshly isolated human colorectal cancer tissue samples and HT29atg5 and SW480atg5 cancer cells. KEY RESULTS In ATG5high colorectal cancer patients, 5-FU-based therapy resulted in nuclear translocation of ATG5, leading to MSI-H. Colorectal cancer in Atg5 Tg mice demonstrated 5-FU resistance, compared to Atg5+/- and WT mice. Proteome microarray assay identified Mis18α, a protein localized on the centromere and a source for methylation of the underlying chromatin, which responded to the translocated nuclear ATG5 leading to ATG5-Mis18α conjugate overexpression. This resulted in MLH1 deficiency due to hypermethylation of hMLH1 promoter CpG island, while the deletion of nuclear Mis18α failed to induce ATG5-Mis18α complex and MMR-D/MSI-H. CONCLUSIONS AND IMPLICATIONS Nuclear ATG5 resulted in MMR-D/MSI-H through its interaction with Mis18α in ATG5high colorectal cancer cells. We suggest that ATG5-Mis18α or Mis18α may be a therapeutic target for treating colorectal cancer.
Collapse
Affiliation(s)
- Shi-Yue Sun
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Institute of Acu-moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xue-Tao Hu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xin-Feng Yu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yue-Ying Zhang
- Department of Experimental Pathology, College of Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiao-Hui Liu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuan-Hang Liu
- Department of Pathology, Hospital of Bin Zhou Medical College, Binzhou, China
| | - Shu-Hua Wu
- Department of Pathology, Hospital of Bin Zhou Medical College, Binzhou, China
| | - Yang-Yang Li
- Department of Pathology, Hospital of Bin Zhou Medical College, Binzhou, China
| | - Shu-Xiang Cui
- Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Xian-Jun Qu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Mismatch Repair Status Characterization in Oncologic Pathology: Taking Stock of the Real-World Possibilities. JOURNAL OF MOLECULAR PATHOLOGY 2021. [DOI: 10.3390/jmp2020009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The mismatch repair (MMR) system has a key role in supporting the DNA polymerase proofreading function and in maintaining genome stability. Alterations in the MMR genes are driving events of tumorigenesis, tumor progression, and resistance to therapy. These genetic scars may occur in either hereditary or sporadic settings, with different frequencies across tumor types. Appropriate characterization of the MMR status is a crucial task in oncologic pathology because it allows for both the tailored clinical management of cancer patients and surveillance of individuals at risk. The currently available MMR testing methods have specific strengths and weaknesses, and their application across different tumor types would require a tailored approach. This article highlights the indications and challenges in MMR status assessment for molecular pathologists, focusing on the possible strategies to overcome analytical and pre-analytical issues.
Collapse
|
24
|
Liu F, Long Q, He H, Dong S, Zhao L, Zou C, Wu W. Combining the Fecal Immunochemical Test with a Logistic Regression Model for Screening Colorectal Neoplasia. Front Pharmacol 2021; 12:635481. [PMID: 33897424 PMCID: PMC8058550 DOI: 10.3389/fphar.2021.635481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/19/2021] [Indexed: 01/05/2023] Open
Abstract
Background: The fecal immunochemical test (FIT) is a widely used strategy for colorectal cancer (CRC) screening with moderate sensitivity. To further increase the sensitivity of FIT in identifying colorectal neoplasia, in this study, we established a classifier model by combining FIT result and other demographic and clinical features. Methods: A total of 4,477 participants were examined with FIT and those who tested positive (over 100 ng/ml) were followed up by a colonoscopy examination. Demographic and clinical information of participants including four domains (basic information, clinical history, diet habits and life styles) that consist of 15 features were retrieved from questionnaire surveys. A mean decrease accuracy (MDA) score was used to select features that are mostly related to CRC. Five different algorithms including logistic regression (LR), classification and regression tree (CART), support vector machine (SVM), artificial neural network (ANN) and random forest (RF) were used to generate a classifier model, through a 10X cross validation process. Area under curve (AUC) and normalized mean squared error (NMSE) were used in the evaluation of the performance of the model. Results: The top six features that are mostly related to CRC include age, gender, history of intestinal adenoma or polyposis, smoking history, gastrointestinal discomfort symptom and fruit eating habit were selected. LR algorithm was used in the generation of the model. An AUC score of 0.92 and an NMSE score of 0.076 were obtained by the final classifier model in separating normal individuals from participants with colorectal neoplasia. Conclusion: Our results provide a new “Funnel” strategy in colorectal neoplasia screening via adding a classifier model filtering step between FIT and colonoscopy examination. This strategy minimizes the need of colonoscopy examination while increases the sensitivity of FIT-based CRC screening.
Collapse
Affiliation(s)
- Feiyuan Liu
- Department of Scientific Research, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Qiaoyun Long
- Department of Clinical Research Center, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China.,Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen, China
| | - Hui He
- Department of Health Management, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Shaowei Dong
- Department of Clinical Research Center, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China.,Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen, China
| | - Li Zhao
- Department of Health Management, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Chang Zou
- Department of Clinical Research Center, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China.,Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen, China
| | - Weiqing Wu
- Department of Health Management, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| |
Collapse
|
25
|
Alsayed A, Salem SE, El Serafi MM, Abdellateif MS, Zekri ARN, Mohanad M, Bahnassy AA. Assessment of the Circulating Tumor Cells and Microsatellite Instability in Colorectal Cancer Patients: Prognostic and Diagnostic Value. Onco Targets Ther 2021; 14:1937-1951. [PMID: 33758513 PMCID: PMC7981167 DOI: 10.2147/ott.s292551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/24/2021] [Indexed: 11/23/2022] Open
Abstract
Background Microsatellite instability (MSI) and circulating tumor cells (CTCs) play important roles in the diagnosis, prognosis and management of colorectal cancer (CRC) patients. Methods CTCs and MSI were assessed in the blood and representative tumor tissues of 100 CRC patients by flow cytometry (FCM) and PCR amplification. The data were correlated to relevant clinicopathological features of the patients, progression-free survival (PFS) and overall survival (OS) rates. Results MSI-high was detected in 44 (44.0%) patients, MSI-low in 37 (37%), and microsatellite stable (MSS) in 19 (19.0%) patients (P=0.007). The baseline CTCs count (<4 cells/7mL blood) was reported in 39% of the patients, and CTCs ≥4 cells/7mL blood in 61% of the patients (P=0.028). Improved PFS and OS rates were associated significantly with MSI-high (P<0.001), decreased CTC levels during the course of treatment (P<0.001) and post-treatment CTCs (P=0.008). There was no significant association between MSI-high and PFS or OS in early-stage patients (P=0.187 and P=0.187; respectively); however, it was associated significantly with better PFS and OS in late-stage patients (P<0.001). Multivariate analysis showed that only a change in serial CTC levels is considered an independent prognostic factor for OS (P<0.012). Post-treatment CTCs level, serial CTCs level changes during the course of treatment, lymph nodes and distant metastasis were independent prognostic factors for PFS (P<0.001, P= 0.047, P=0.001 and P<0.001; respectively). Conclusion MSI and CTCs could be used as accurate, reliable and sensitive diagnostic and prognostic biomarkers for CRC patients’ survival rates and outcomes.
Collapse
Affiliation(s)
- Aya Alsayed
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, 11976, Egypt
| | - Salem E Salem
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, 11976, Egypt
| | - Mostafa M El Serafi
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, 11976, Egypt
| | - Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11976, Egypt
| | - Abdel-Rahman N Zekri
- Molecular Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11976, Egypt
| | - Marwa Mohanad
- Biochemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, 12945, Egypt
| | - Abeer A Bahnassy
- Pathology Department, National Cancer Institute, Cairo University, Cairo, 11976, Egypt
| |
Collapse
|
26
|
Novel Epigenetic Eight-Gene Signature Predictive of Poor Prognosis and MSI-Like Phenotype in Human Metastatic Colorectal Carcinomas. Cancers (Basel) 2021; 13:cancers13010158. [PMID: 33466447 PMCID: PMC7796477 DOI: 10.3390/cancers13010158] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The global methylation profile of two human metastatic colorectal carcinoma subgroups with significantly different outcomes (primary-resistant versus drug-sensitive tumors) was analyzed and compared with the gene expression and methylation data from The Cancer Genome Atlas COlon ADenocarcinoma (TCGA COAD) metastatic colorectal carcinoma dataset with the aim to identify a prognostic signature of functionally methylated genes. A novel epigenetic eight-gene signature, with hypermethylation of the promoter regions, was identified and validated for its capacity to predict poor outcome, which had a CpG-island methylator phenotype (CIMP)-high status and microsatellite instability (MSI)-like phenotype. Abstract Epigenetics is involved in tumor progression and drug resistance in human colorectal carcinoma (CRC). This study addressed the hypothesis that the DNA methylation profiling may predict the clinical behavior of metastatic CRCs (mCRCs). The global methylation profile of two human mCRC subgroups with significantly different outcome was analyzed and compared with gene expression and methylation data from The Cancer Genome Atlas COlon ADenocarcinoma (TCGA COAD) and the NCBI GENE expression Omnibus repository (GEO) GSE48684 mCRCs datasets to identify a prognostic signature of functionally methylated genes. A novel epigenetic signature of eight hypermethylated genes was characterized that was able to identify mCRCs with poor prognosis, which had a CpG-island methylator phenotype (CIMP)-high and microsatellite instability (MSI)-like phenotype. Interestingly, methylation events were enriched in genes located on the q-arm of chromosomes 13 and 20, two chromosomal regions with gain/loss alterations associated with adenoma-to-carcinoma progression. Finally, the expression of the eight-genes signature and MSI-enriching genes was confirmed in oxaliplatin- and irinotecan-resistant CRC cell lines. These data reveal that the hypermethylation of specific genes may provide prognostic information that is able to identify a subgroup of mCRCs with poor prognosis.
Collapse
|
27
|
Wielandt AM, Hurtado C, Moreno C M, Villarroel C, Castro M, Estay M, Simian D, Martinez M, Vial MT, Kronberg U, López-Köstner F. Characterization of Chilean patients with sporadic colorectal cancer according to the three main carcinogenic pathways: Microsatellite instability, CpG island methylator phenotype and Chromosomal instability. Tumour Biol 2020; 42:1010428320938492. [PMID: 32635826 DOI: 10.1177/1010428320938492] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Molecular classification of colorectal cancer is difficult to implement in clinical settings where hundreds of genes are involved, and resources are limited. This study aims to characterize the molecular subtypes of patients with sporadic colorectal cancer based on the three main carcinogenic pathways microsatellite instability (MSI), CpG island methylator phenotype (CIMP), and chromosomal instability (CIN) in a Chilean population. Although several reports have characterized colorectal cancer, most do not represent Latin-American populations. Our study includes 103 colorectal cancer patients who underwent surgery, without neoadjuvant treatment, in a private hospital between 2008 and 2017. MSI, CIN, and CIMP status were assessed. Frequent mutations in KRAS, BRAF, and PIK3CA genes were analyzed by Sanger sequencing, and statistical analysis was performed by Fisher's exact and/or chi-square test. Survival curves were estimated with Kaplan-Meier and log-rank test. Based on our observations, we can classify the tumors in four subgroups, Group 1: MSI-high tumors (15%) are located in the right colon, occur at older age, and 60% show a BRAF mutation; Group 2: CIN-high tumors (38%) are in the left colon, and 26% have KRAS mutations. Group 3: [MSI/CIN/CIMP]-low/negative tumors (30%) are left-sided, and 39% have KRAS mutations; Group 4: CIMP-high tumors (15%) were more frequent in men and left side colon, with 27% KRAS and 7% presented BRAF mutations. Three percent of patients could not be classified. We found that CIMP-high was associated with a worse prognosis, both in MSI-high and MSI stable patients (p = 0.0452). Group 3 (Low/negative tumors) tend to have better overall survival compared with MSI-high, CIMP-high, and CIN-high tumors. This study contributes to understanding the heterogeneity of tumors in the Chilean population being one of the few characterizations performed in Latin-America. Given the limited resources of these countries, these results allow to improve molecular characterization in Latin-American colorectal cancer populations and confirm the possibility of using the three main carcinogenic pathways to define therapeutic strategies.
Collapse
Affiliation(s)
- Ana María Wielandt
- Oncology and Molecular Genetics Laboratory, Coloproctology Unit, Clínica Las Condes, Santiago, Chile.,Coloproctology Unit, Clínica Las Condes, Santiago, Chile
| | - Claudia Hurtado
- Oncology and Molecular Genetics Laboratory, Coloproctology Unit, Clínica Las Condes, Santiago, Chile.,Coloproctology Unit, Clínica Las Condes, Santiago, Chile
| | - Mauricio Moreno C
- Oncology and Molecular Genetics Laboratory, Coloproctology Unit, Clínica Las Condes, Santiago, Chile.,Coloproctology Unit, Clínica Las Condes, Santiago, Chile
| | - Cynthia Villarroel
- Oncology and Molecular Genetics Laboratory, Coloproctology Unit, Clínica Las Condes, Santiago, Chile
| | - Magdalena Castro
- Academic Department Research Unit, Clínica Las Condes, Santiago, Chile
| | - Marlene Estay
- Coloproctology Unit, Clínica Las Condes, Santiago, Chile
| | - Daniela Simian
- Academic Department Research Unit, Clínica Las Condes, Santiago, Chile
| | - Maripaz Martinez
- Academic Department Research Unit, Clínica Las Condes, Santiago, Chile
| | | | - Udo Kronberg
- Coloproctology Unit, Clínica Las Condes, Santiago, Chile
| | | |
Collapse
|
28
|
Predicting liver metastases growth patterns: Current status and future possibilities. Semin Cancer Biol 2020; 71:42-51. [PMID: 32679190 DOI: 10.1016/j.semcancer.2020.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022]
Abstract
Colorectal cancer is highly incident worldwide and presents a health burden with elevated mortality rate despite prevention, detection, and treatment, mainly due to metastatic liver disease. Histological growth patterns of colorectal cancer liver metastases have emerged as a reproducible prognostic factor, with biological implications and therapeutic windows. Nonetheless, the histological growth patterns of colorectal cancer liver metastases are only known after pathological examination of a liver resection specimen, thus limiting the possibilities of pre-surgical decision. Predicting the histological growth pattern of colorectal cancer liver metastases would provide valuable information for patient-tailored medicine. In this article, we perform a review of the histological growth patterns and their implications, with a focus on the possibilities for their prediction.
Collapse
|
29
|
Ranjbar R, Esfahani AT, Nazemalhosseini-Mojarad E, Olfatifar M, Aghdaei HA, Mohammadpour S. EMAST frequency in colorectal cancer: a meta-analysis and literature review. Biomark Med 2020; 14:1021-1030. [PMID: 32940074 DOI: 10.2217/bmm-2020-0068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/21/2020] [Indexed: 01/07/2023] Open
Abstract
Aim: The prognostic and predictive value of Elevated Microsatellite Alterations at Selected Tetranucleotide (EMAST) has been reported in colorectal cancer (CRC). The prevalence of EMAST in CRC varied across the literature. We conducted a meta-analysis to determine the prevalence of EMAST in CRC. Materials & methods: Three international databases including PubMed, ISI and Scopus were searched to identify related articles that described the frequency of EMAST. Results: Analysis was performed on 16 eligible studies including 4922 patients. The overall EMAST prevalence among CRCs patients was 33% (95% CI: 23-43%, I2 = 98%). Conclusion: This study indicated that approximately a third of the CRC patients are diagnosed with EMAST, hereupon EMAST as a prognostic and predictive biomarker should be more studied clinically.
Collapse
Affiliation(s)
- Reza Ranjbar
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amir T Esfahani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Nazemalhosseini-Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Olfatifar
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid A Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Mohammadpour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Zhao L, Shan G, Li L, Yu Y, Cheng G, Zheng X. A robust method for the rapid detection of microsatellite instability in colorectal cancer. Oncol Lett 2020; 20:1982-1988. [PMID: 32724444 DOI: 10.3892/ol.2020.11702] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 01/23/2020] [Indexed: 12/21/2022] Open
Abstract
Although several computational tools using next-generation sequencing (NGS) data have been proposed to detect microsatellite instability (MSI) status, they still have limitations and need improvement. We developed a NovoPM-MSI method to detect MSI status based on NGS data. This method evaluated target mononucleotide microsatellite loci that were sequenced during targeted gene enrichment analysis and reported sample instability score as the fraction of unstable loci within the target set after assessing locus instability by comparing length distribution in paired tumor-normal samples. We validated this method against the conventional MSI-PCR method in 113 paired colorectal cancer (CRC) specimens and compared the performance of NovoPM-MSI to that of mSINGS and MANTIS in accuracy and runtime efficiency. By using the MSI status from MSI-PCR as the gold standard, the three computational methods showed the same sensitivity of 88.9% but different specificities (NovoPM-MSI 97.1%, MANTIS 86.5% and mSINGS 99.0%). Only NovoPM-MSI could greatly improve both the sensitivity and specificity by setting an ambiguous interval. MANTIS had the shortest average runtime (16.3 sec), followed by NovoPM-MSI (18.3 sec) and mSINGS (109.0 sec). In short, the NovoPM-MSI method provides a fast and reliable MSI detection method with accuracy comparable to MSI-PCR in paired CRC samples.
Collapse
Affiliation(s)
- Lin Zhao
- Beijing Novogene Bioinformatics Technology Co., Ltd, Beijing 100015, P.R. China
| | - Guangyu Shan
- Beijing Novogene Bioinformatics Technology Co., Ltd, Beijing 100015, P.R. China
| | - Lei Li
- Beijing Novogene Bioinformatics Technology Co., Ltd, Beijing 100015, P.R. China
| | - Yang Yu
- Beijing Novogene Bioinformatics Technology Co., Ltd, Beijing 100015, P.R. China
| | - Gang Cheng
- Beijing Novogene Bioinformatics Technology Co., Ltd, Beijing 100015, P.R. China
| | - Xu Zheng
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
31
|
Adenocarcinoma with Neuroendocrine Differentiation of the Colon Accompanying Osteoclast-Like Giant Cells. Case Rep Pathol 2020. [DOI: 10.1155/2020/1976319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction. Neuroendocrine differentiation in colorectal cancer is reportedly associated with poorer grade of tumor differentiation, nodal and distant metastasis, and other unfavorable features, contributing to a worse clinical outcome. Colorectal cancer with osteoclast-like giant cells (OGCs) is extremely rare. Case Presentation. An 86-year-old woman was diagnosed as double cancer of the transverse and sigmoid colon. Both tumors were simultaneously removed. The transverse colon cancer directly invaded the area of the right gastroepiploic vessels and spread to the nodes and histologically consisted of both the tubuloglandular and solid components. CD8/granzyme B-positive tumor-infiltrating lymphocytes and CD163/CD68-positive macrophages, frequently forming OGCs, were observed particularly at the invasion front. The carcinoma cells were labeled focally for synaptophysin and diffusely for the DR locus of the human leukocyte antigen and programmed death-ligand 1 (PD-L1). Deficient expression of DNA mismatch repair (dMMR) proteins was immunohistochemically confirmed. The patient died 16 months after surgery. Conclusion. This is the first report of colonic adenocarcinoma with neuroendocrine differentiation accompanying OGCs. Histopathologic factors of the poor prognosis in the present case included (a) the presence of more than 2% cells with neuroendocrine differentiation, (b) infiltration of CD163/CD68-positive OGCs at the invasion front, (c) deficiency of dMMR proteins, and (d) PD-L1 expression.
Collapse
|
32
|
Ju JY, Dibbern ME, Mahadevan MS, Fan J, Kunk PR, Stelow EB. Mismatch Repair Protein Deficiency/Microsatellite Instability Is Rare in Cholangiocarcinomas and Associated With Distinctive Morphologies. Am J Clin Pathol 2020; 153:598-604. [PMID: 31844887 DOI: 10.1093/ajcp/aqz199] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Although germline mutations of mismatch repair (MMR) genes (Lynch syndrome) are not typically associated with cholangiocarcinomas, the US Food and Drug Administration recently approved the use of pembrolizumab in patients with advanced solid tumors at all sites that show MMR deficiency or associated high microsatellite instability. METHODS We analyzed 96 cases of intra- and extrahepatic cholangiocarcinomas for morphology using H&E and for MMR status using immunohistochemical staining. We submitted any results with MMR loss for microsatellite instability testing. RESULTS We found that 6% of samples showed MMR deficiency. The best predictive factor was a nontypical infiltrating pattern of invasion (P < .0001). No patients with MMR deficiency had a history of a cancer typically associated with Lynch syndrome. CONCLUSIONS Solid, mucinous, or signet-ring appearance of a cholangiocarcinoma should prompt MMR testing for immunotherapy options but should not necessarily raise concern about Lynch syndrome.
Collapse
Affiliation(s)
- Jennifer Y Ju
- Departments of Pathology, University of Virginia, Charlottesville
| | - Megan E Dibbern
- Departments of Pathology, University of Virginia, Charlottesville
| | - Mani S Mahadevan
- Departments of Pathology, University of Virginia, Charlottesville
| | - Jinbo Fan
- Departments of Pathology, University of Virginia, Charlottesville
| | - Paul R Kunk
- Hematology/Oncology, University of Virginia, Charlottesville
| | - Edward B Stelow
- Departments of Pathology, University of Virginia, Charlottesville
| |
Collapse
|
33
|
Paulose RR, Ail DA, Biradar S, Vasudevan A, Sundaram KR. Prognostic and predictive significance of microsatellite instability in stage II colorectal carcinoma: An 8-year study from a tertiary center in South India. Indian J Cancer 2020; 56:302-308. [PMID: 31607697 DOI: 10.4103/ijc.ijc_365_18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Microsatellite instability (MSI) accounts for 15-20% of colorectal cancer (CRC) and is considered to have favorable stage-adjusted prognosis compared to Microsatellite stable (MSS) CRCs. Determination of MSI in stage II CRC is important for management decisions regarding adjuvant chemotherapy administration. The aim of this study was to determine the prognostic and predictive significance of MSI in stage 2 CRC in the Indian scenario. MATERIALS AND METHODS A total of 195 patients who underwent curative surgery for stage II CRC from 2010 to 2017 were included. MSI testing by immunohistochemistry (DNA MisMatch Repair proteins) was performed in all. Various clinicopathological factors and disease-free survival and overall survival were assessed between MSI and MSS groups. The effect of treatment in terms of survival benefits with adjuvant therapy in the MSI group was also assessed. RESULTS 27.1% of the CRCs' showed MSI. Younger age (<50 years), family history of cancer, synchronous/metachronous malignancies, proximal (right sided) location, poor morphological tumour differentiation, mucin production, and presence of peritumoral (Crohn's-like) lymphocytic response showed statistically significant association with MSI. Majority (56%) of our patients showed combined loss of MLH1 and PMS2. Overall, survival among the MSI patients was significantly higher (76.6 ± 4.149 months) than the MSS patients (65.05 ± 3.555)P= 0.04. MSI patients did not show any differences in survival with or without treatment. CONCLUSION This study highlights the distinct clinicopathological features of MSI-related CRC and the relevance of MSI testing of stage II CRC for management decisions and prognostication.
Collapse
Affiliation(s)
- Roopa R Paulose
- Department of Pathology, Amrita Institute of Medical Sciences, Kochi, Kerala, India
| | - Divya A Ail
- Department of Pathology, Amrita Institute of Medical Sciences, Kochi, Kerala, India
| | - Shital Biradar
- Department of Pathology, Amrita Institute of Medical Sciences, Kochi, Kerala, India
| | - Anu Vasudevan
- Department of Biostatistics, Amrita Institute of Medical Sciences, Kochi, Kerala, India
| | - K R Sundaram
- Department of Biostatistics, Amrita Institute of Medical Sciences, Kochi, Kerala, India
| |
Collapse
|
34
|
Basade M, Mane A. Optimum patient selection for irinotecan-containing regimens in metastatic colorectal cancer: Literature review and lessons from clinical practice. Indian J Cancer 2020; 58:5-16. [PMID: 33402591 DOI: 10.4103/ijc.ijc_507_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Metastatic colorectal cancer (mCRC) accounts for over 20% of CRC cases and is associated with a poor prognosis. Irinotecan is an important first- and second-line chemotherapy option for mCRC. In this review, we summarize the clinical efficacy and safety of irinotecan-based regimens for the treatment of mCRC and discuss various tumor- and patient-related factors that affect the clinical response, survival, and toxicity associated with these regimens. Uridine diphosphate glucuronosyltransferase (UGT) gene polymorphisms such as UGT1A1*28/*6, age, performance status, serum lactate dehydrogenase levels, and bilirubin levels could be important considerations for predicting outcomes and tolerability with irinotecan-based regimens. The role of tumor location; chemotherapy backbone; and emerging evidence on the presence of microsatellite instability-high status, consensus molecular subtype 4 tumors, and signet-ring morphology in predicting response to irinotecan-based therapy have also been highlighted. Careful consideration of these factors will help guide clinicians in optimizing the selection of mCRC patients for irinotecan-based treatment.
Collapse
Affiliation(s)
- Maheboob Basade
- Consultant Medical Oncologist, Panchsheel Plaza, Off Hughes Road, Gamdevi, Mumbai, Maharashtra, India
| | - Akshata Mane
- Medical Affairs, Pfizer Limited, The Capital 1802/1901, Bandra Kurla Complex, Bandra(E), Mumbai, Maharashtra, India
| |
Collapse
|
35
|
Galon J, Bruni D. The Role of the Immune Infiltrate in Distinct Cancer Types and Its Clinical Implications : Lymphocytic Infiltration in Colorectal Cancer. Cancer Treat Res 2020; 180:197-211. [PMID: 32215871 DOI: 10.1007/978-3-030-38862-1_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) represents a major public health challenges, with one of the highest incidences worldwide. The two affected anatomical sites in CRC, i.e. the colon and the rectum, share important underlying features, but often differ in terms of therapeutic management. Current guidelines for CRC define its clinical stratification according to classical, tumor cell-based and pathological parameters. Novel ground-breaking findings in the recent years revealed the prominent role of the immune system in shaping CRC development. This chapter provides a detailed overview of the main genomic and immune features driving (or hampering) CRC progression, with a focus on the main immune cells and factors shaping its evolution. Furthermore, we discuss how tumor-infiltrating immunity could be leveraged both for therapeutic and stratification purposes.
Collapse
Affiliation(s)
- Jérôme Galon
- INSERM Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, 75006, Paris, France.
| | - Daniela Bruni
- INSERM Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, 75006, Paris, France
| |
Collapse
|
36
|
Fu Y, Qi L, Guo W, Jin L, Song K, You T, Zhang S, Gu Y, Zhao W, Guo Z. A qualitative transcriptional signature for predicting microsatellite instability status of right-sided Colon Cancer. BMC Genomics 2019; 20:769. [PMID: 31646964 PMCID: PMC6813057 DOI: 10.1186/s12864-019-6129-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/23/2019] [Indexed: 12/16/2022] Open
Abstract
Background Microsatellite instability (MSI) accounts for about 15% of colorectal cancer and is associated with prognosis. Today, MSI is usually detected by polymerase chain reaction amplification of specific microsatellite markers. However, the instability is identified by comparing the length of microsatellite repeats in tumor and normal samples. In this work, we developed a qualitative transcriptional signature to individually predict MSI status for right-sided colon cancer (RCC) based on tumor samples. Results Using RCC samples, based on the relative expression orderings (REOs) of gene pairs, we extracted a signature consisting of 10 gene pairs (10-GPS) to predict MSI status for RCC through a feature selection process. A sample is predicted as MSI when the gene expression orderings of at least 7 gene pairs vote for MSI; otherwise the microsatellite stability (MSS). The classification performance reached the largest F-score in the training dataset. This signature was verified in four independent datasets of RCCs with the F-scores of 1, 0.9630, 0.9412 and 0.8798, respectively. Additionally, the hierarchical clustering analyses and molecular features also supported the correctness of the reclassifications of the MSI status by 10-GPS. Conclusions The qualitative transcriptional signature can be used to classify MSI status of RCC samples at the individualized level.
Collapse
Affiliation(s)
- Yelin Fu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China
| | - Lishuang Qi
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China
| | - Wenbing Guo
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China
| | - Liangliang Jin
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China
| | - Kai Song
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China
| | - Tianyi You
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China
| | - Shuobo Zhang
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China
| | - Yunyan Gu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China
| | - Wenyuan Zhao
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China.
| | - Zheng Guo
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China. .,Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China. .,Key Laboratory of Medical Bioinformatics, Fujian Province, Fuzhou, 350122, China.
| |
Collapse
|
37
|
Tiwari V, Wilson DM. DNA Damage and Associated DNA Repair Defects in Disease and Premature Aging. Am J Hum Genet 2019; 105:237-257. [PMID: 31374202 PMCID: PMC6693886 DOI: 10.1016/j.ajhg.2019.06.005] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/05/2019] [Indexed: 12/14/2022] Open
Abstract
Genetic information is constantly being attacked by intrinsic and extrinsic damaging agents, such as reactive oxygen species, atmospheric radiation, environmental chemicals, and chemotherapeutics. If DNA modifications persist, they can adversely affect the polymerization of DNA or RNA, leading to replication fork collapse or transcription arrest, or can serve as mutagenic templates during nucleic acid synthesis reactions. To combat the deleterious consequences of DNA damage, organisms have developed complex repair networks that remove chemical modifications or aberrant base arrangements and restore the genome to its original state. Not surprisingly, inherited or sporadic defects in DNA repair mechanisms can give rise to cellular outcomes that underlie disease and aging, such as transformation, apoptosis, and senescence. In the review here, we discuss several genetic disorders linked to DNA repair defects, attempting to draw correlations between the nature of the accumulating DNA damage and the pathological endpoints, namely cancer, neurological disease, and premature aging.
Collapse
Affiliation(s)
- Vinod Tiwari
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA.
| | - David M Wilson
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, National Institutes of Health, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA.
| |
Collapse
|
38
|
Genome-wide investigation of microsatellite polymorphism in coding region of the giant panda (Ailuropoda melanoleuca) genome: a resource for study of phenotype diversity and abnormal traits. MAMMAL RES 2019. [DOI: 10.1007/s13364-019-00418-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
39
|
Hao S, Ren M, Li D, Sui Y, Wang Q, Chen G, Li Z, Yang Q. Fisher linear discriminant analysis for classification and prediction of genomic susceptibility to stomach and colorectal cancers based on six STR loci in a northern Chinese Han population. PeerJ 2019; 7:e7004. [PMID: 31179189 PMCID: PMC6544021 DOI: 10.7717/peerj.7004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/23/2019] [Indexed: 01/06/2023] Open
Abstract
Objective Gastrointestinal cancer is the leading cause of cancer-related death worldwide. The aim of this study was to verify whether the genotype of six short tandem repeat (STR) loci including AR, Bat-25, D5S346, ER1, ER2, and FGA is associated with the risk of gastric cancer (GC) and colorectal cancer (CRC) and to develop a model that allows early diagnosis and prediction of inherited genomic susceptibility to GC and CRC. Methods Alleles of six STR loci were determined using the peripheral blood of six colon cancer patients, five rectal cancer patients, eight GC patients, and 30 healthy controls. Fisher linear discriminant analysis (FDA) was used to establish the discriminant formula to distinguish GC and CRC patients from healthy controls. Leave-one-out cross validation and receiver operating characteristic (ROC) curves were used to validate the accuracy of the formula. The relationship between the STR status and immunohistochemical (IHC) and tumor markers was analyzed using multiple correspondence analysis. Results D5S346 was confirmed as a GC- and CRC-related STR locus. For the first time, we established a discriminant formula on the basis of the six STR loci, which was used to estimate the risk coefficient of suffering from GC and CRC. The model was statistically significant (Wilks’ lambda = 0.471, χ2 = 30.488, df = 13, and p = 0.004). The results of leave-one-out cross validation showed that the sensitivity of the formula was 73.7% and the specificity was 76.7%. The area under the ROC curve (AUC) was 0.926, with a sensitivity of 73.7% and a specificity of 93.3%. The STR status was shown to have a certain relationship with the expression of some IHC markers and the level of some tumor markers. Conclusions The results of this study complement clinical diagnostic criteria and present markers for early prediction of GC and CRC. This approach will aid in improving risk awareness of susceptible individuals and contribute to reducing the incidence of GC and CRC by prevention and early detection.
Collapse
Affiliation(s)
- Shuhong Hao
- Department of Hematology and Oncology, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Ming Ren
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Dong Li
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yujie Sui
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qingyu Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Gaoyang Chen
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zhaoyan Li
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qiwei Yang
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
40
|
Reilly NM, Novara L, Di Nicolantonio F, Bardelli A. Exploiting DNA repair defects in colorectal cancer. Mol Oncol 2019; 13:681-700. [PMID: 30714316 PMCID: PMC6441925 DOI: 10.1002/1878-0261.12467] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/11/2019] [Accepted: 01/19/2019] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related deaths worldwide. Therapies that take advantage of defects in DNA repair pathways have been explored in the context of breast, ovarian, and other tumor types, but not yet systematically in CRC. At present, only immune checkpoint blockade therapies have been FDA approved for use in mismatch repair-deficient colorectal tumors. Here, we discuss how systematic identification of alterations in DNA repair genes could provide new therapeutic opportunities for CRCs. Analysis of The Cancer Genome Atlas Colon Adenocarcinoma (TCGA-COAD) and Rectal Adenocarcinoma (TCGA-READ) PanCancer Atlas datasets identified 141 (out of 528) cases with putative driver mutations in 29 genes associated with DNA damage response and repair, including the mismatch repair and homologous recombination pathways. Genetic defects in these pathways might confer repair-deficient characteristics, such as genomic instability in the absence of homologous recombination, which can be exploited. For example, inhibitors of poly(ADP)-ribose polymerase are effectively used to treat cancers that carry mutations in BRCA1 and/or BRCA2 and have shown promising results in CRC preclinical studies. HR deficiency can also occur in cells with no detectable BRCA1/BRCA2 mutations but exhibiting BRCA-like phenotypes. DNA repair-targeting therapies, such as ATR and CHK1 inhibitors (which are most effective against cancers carrying ATM mutations), can be used in combination with current genotoxic chemotherapies in CRCs to further improve therapy response. Finally, therapies that target alternative DNA repair mechanisms, such as thiopurines, also have the potential to confer increased sensitivity to current chemotherapy regimens, thus expanding the spectrum of therapy options and potentially improving clinical outcomes for CRC patients.
Collapse
Affiliation(s)
- Nicole M. Reilly
- Fondazione Piemontese per la Ricerca sul Cancro ONLUSCandioloItaly
| | - Luca Novara
- Candiolo Cancer InstituteFPO‐IRCCSCandioloItaly
| | - Federica Di Nicolantonio
- Candiolo Cancer InstituteFPO‐IRCCSCandioloItaly
- Department of OncologyUniversity of TorinoCandioloItaly
| | - Alberto Bardelli
- Candiolo Cancer InstituteFPO‐IRCCSCandioloItaly
- Department of OncologyUniversity of TorinoCandioloItaly
| |
Collapse
|
41
|
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; 40:142-161. [PMID: 30461124 PMCID: PMC6328345 DOI: 10.1002/humu.23688] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/24/2018] [Accepted: 11/14/2018] [Indexed: 12/30/2022]
Abstract
The epithelial cell adhesion molecule gene (EPCAM, previously known as TACSTD1 or TROP1) encodes a membrane-bound protein that is localized to the basolateral membrane of epithelial cells and is overexpressed in some tumors. Biallelic mutations in EPCAM cause congenital tufting enteropathy (CTE), which is a rare chronic diarrheal disorder presenting in infancy. Monoallelic deletions of the 3' end of EPCAM that silence the downstream gene, MSH2, cause a form of Lynch syndrome, which is a cancer predisposition syndrome associated with loss of DNA mismatch repair. Here, we report 13 novel EPCAM mutations from 17 CTE patients from two separate centers, review EPCAM mutations associated with CTE and Lynch syndrome, and structurally model pathogenic missense mutations. Statistical analyses indicate that the c.499dupC (previously reported as c.498insC) frameshift mutation was associated with more severe treatment regimens and greater mortality in CTE, whereas the c.556-14A>G and c.491+1G>A splice site mutations were not correlated with treatments or outcomes significantly different than random simulation. These findings suggest that genotype-phenotype correlations may be useful in contributing to management decisions of CTE patients. Depending on the type and nature of EPCAM mutation, one of two unrelated diseases may occur, CTE or Lynch syndrome.
Collapse
Affiliation(s)
- Sagar J. Pathak
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
- Rady Children's HospitalSan DiegoCalifornia
| | - James L. Mueller
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
| | - Kevin Okamoto
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
| | - Barun Das
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
| | - Jozef Hertecant
- Genetics/Metabolics ServiceTawam HospitalAl AinUnited Arab Emirates
| | | | - Trevor Cole
- West Midlands Regional Genetics Service and Birmingham Health PartnersBirmingham Women's HospitalBirminghamUK
| | - Vered Pinsk
- Division of Pediatrics, Pediatric Gastroenterology UnitSoroka University Medical Center and Faculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Baruch Yerushalmi
- Division of Pediatrics, Pediatric Gastroenterology UnitSoroka University Medical Center and Faculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Odul E. Gurkan
- Department of PediatricsGazi University School of MedicineAnkaraTurkey
| | - Michael Yourshaw
- Department of Human GeneticsUniversity of California, Los AngelesLos AngelesCalifornia
| | - Erick Hernandez
- Pediatric GastroenterologyMiami Children's Health SystemMiamiFlorida
| | | | - Sandhia Naik
- Paediatric GastroenterologyBarts and the London School of MedicineLondonUK
| | - Ian R. Sanderson
- Paediatric GastroenterologyBarts and the London School of MedicineLondonUK
| | - Irene Axelsson
- Department of PediatricsSkane University HospitalMalmoSweden
| | - Daniel Agardh
- Department of Clinical SciencesLund University, Skane University HospitalMalmoSweden
| | - C. Richard Boland
- Department of MedicineUniversity of California, San DiegoLa JollaCalifornia
| | - Martin G. Martin
- Department of PediatricsUniversity of California, Los AngelesLos AngelesCalifornia
| | - Christopher D. Putnam
- Department of MedicineUniversity of California, San DiegoLa JollaCalifornia
- San Diego BranchLudwig Institute for Cancer ResearchLa JollaCalifornia
| | - Mamata Sivagnanam
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
- Rady Children's HospitalSan DiegoCalifornia
| |
Collapse
|
42
|
Yi M, Jiao D, Xu H, Liu Q, Zhao W, Han X, Wu K. Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors. Mol Cancer 2018; 17:129. [PMID: 30139382 PMCID: PMC6107958 DOI: 10.1186/s12943-018-0864-3] [Citation(s) in RCA: 558] [Impact Index Per Article: 79.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 07/24/2018] [Indexed: 12/30/2022] Open
Abstract
Programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) is a negative modulatory signaling pathway for activation of T cell. It is acknowledged that PD-1/PD-L1 axis plays a crucial role in the progression of tumor by altering status of immune surveillance. As one of the most promising immune therapy strategies, PD-1/PD-L1 inhibitor is a breakthrough for the therapy of some refractory tumors. However, response rate of PD-1/PD-L1 inhibitors in overall patients is unsatisfactory, which limits the application in clinical practice. Therefore, biomarkers which could effectively predict the efficacy of PD-1/PD-L1 inhibitors are crucial for patient selection. Biomarkers reflecting tumor immune microenvironment and tumor cell intrinsic features, such as PD-L1 expression, density of tumor infiltrating lymphocyte (TIL), tumor mutational burden, and mismatch-repair (MMR) deficiency, have been noticed to associate with treatment effect of anti-PD-1/anti-PD-L1 therapy. Furthermore, gut microbiota, circulating biomarkers, and patient previous history have been found as valuable predictors as well. Therefore establishing a comprehensive assessment framework involving multiple biomarkers would be meaningful to interrogate tumor immune landscape and select sensitive patients.
Collapse
Affiliation(s)
- Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dechao Jiao
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hanxiao Xu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|