51
|
Evaristo G, Katz A, Ramírez-GarcíaLuna JL, Issac MSM, Sangwan V, Thai DV, Bertos N, Guiot MC, Camilleri-Broët S, Marcus V, Mueller C, Cools-Lartigue J, Fiset PO, Ferri LE. Relation between mismatch repair status, chemoresponse, survival and anatomic location in gastroesophageal adenocarcinoma. Can J Surg 2023; 66:E79-E87. [PMID: 36792128 PMCID: PMC9943547 DOI: 10.1503/cjs.017021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2022] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND It has recently been reported that mismatch repair (MMR) status and microsatellite instability (MSI) status in gastroesophageal carcinomas predict surgical, chemotherapeutic and immunotherapeutic outcomes; however, there is extensive variability in the reported incidence and clinical implications of MMR/MSI status in gastroesophaegal adenocarcinomas. We characterized a Canadian surgical patient cohort with respect to MMR status, clinicopathologic correlates and anatomic tumour location. METHODS We investigated MMR and BRAF V600E status of gastroesophaegal adenocarcinomas in patients who underwent gastrectomy or esophagectomy with extended (D2) lymphadenectomy at a single centre between 2011 and 2019. We correlated patterns of MMR expression in the overall cohort and in anatomic location-defined subgroups with treatment response and overall survival using multivariate analysis. RESULTS In all, 226 cases of gastroesophaegal adenocarcinoma (63 esophageal, 98 gastroesophageal junctional and 65 gastric) were included. The MMR-deficient (dMMR) immunophenotype was found in 28 tumours (12.3%) (15 junctional [15.3%], 13 gastric [20.0%] and none of the esophageal). The majority (25 [89%]) of dMMR cases showed MLH1/PMS2 loss without concurrent BRAF V600E mutation. Two MSH2/ MSH6-deficient gastric tumours and 1 MSH6-deficient junctional tumour were detected. The pathologic response to preoperative chemotherapy was comparable in the dMMR and MMR-proficient (pMMR) cohorts. However, dMMR status was associated with significantly longer median overall survival than pMMR status (5.8 yr v. 2.4 yr, hazard ratio [HR] 1.91, 95% confidence interval [CI] 1.06-3.46), particularly in junctional tumours (4.6 yr v. 1.9 yr, HR 2.97, 95% CI 1.27-6.94). CONCLUSION Our study shows that MMR status has at least prognostic value, which supports the need for biomarker testing in gastroesophageal adenocarcinomas, including junctional adenocarcinomas. This highlights the clinical significance of determining the MMR status in all adenocarcinomas of the upper gastrointestinal tract. Response to induction chemotherapy, however, was not influenced by MMR status.
Collapse
Affiliation(s)
- Gertruda Evaristo
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - Amit Katz
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - José L Ramírez-GarcíaLuna
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - Marianne S M Issac
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - Veena Sangwan
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - Duc-Vinh Thai
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - Nicholas Bertos
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - Marie-Christine Guiot
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - Sophie Camilleri-Broët
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - Victoria Marcus
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - Carmen Mueller
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - Jonathan Cools-Lartigue
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - Pierre O Fiset
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| | - Lorenzo E Ferri
- From the Department of Pathology, McGill University Health Centre, Montréal, Que. (Evaristo, Thai, Guiot, Camilleri-Broët, Marcus, Fiset); the Cancer Research Program and the LD MacLean Surgical Research Laboratories, Department of Surgery, Research Institute of the McGill University Health Centre, Montréal, Que. (Katz, Ramírez-GarcíaLuna, Sangwan, Mueller, Cools-Lartigue, Ferri); the Research Institute of the McGill University Health Centre, Montréal, Que. (Issac, Bertos); and the Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, McGill University Health Centre, Montreal General Hospital, Montréal, Que. (Sangwan, Mueller, Cools-Lartigue, Ferri)
| |
Collapse
|
52
|
Russo G, Pepe F, Pisapia P, Palumbo L, Nacchio M, Vigliar E, Pallante P, Parente P, Fassan M, Graziano P, Bellevicine C, Troncone G, Malapelle U, Iaccarino A. Microsatellite instability evaluation of patients with solid tumour: routine practice insight from a large series of Italian referral centre. J Clin Pathol 2023; 76:133-136. [PMID: 35545354 DOI: 10.1136/jclinpath-2022-208203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/13/2022] [Indexed: 01/24/2023]
Abstract
DNA mismatch repair complex is involved in the maintenance of DNA stability. In the recent years, a plethora of technical approaches for microsatellite instability (MSI) analysis emerged. Here, we review the results of our MSI status evaluation by adopting a customised workflow on microfluidic system obtained in 4 years of diagnostic routine practice. Data from MSI status were retrieved from our institutional archive covering the period from January 2017 to December 2021. Microfluidic analysis was carried out on microfluidic platform. Results were inspected with a proprietary software. Overall, microsatellite stability (MSS) and MSI-high (MSI-H) profile was detected in n=423/458 (92.36%) and n=35/458 (7.64%) patients with metastatic CRC (mCRC), respectively. In addition, n=78/86 (90.70%) and n=8/86 (9.30%) patients without CRC showed an MSS and MSI-H profile. This review highlights the suitability of microfluidic approach in patients with cancer for MSI testing.
Collapse
Affiliation(s)
- Gianluca Russo
- Public Health, University of Naples Federico II, Naples, Italy
| | - Francesco Pepe
- Public Health, University of Naples Federico II, Naples, Italy
| | | | - Lucia Palumbo
- Public Health, University of Naples Federico II, Naples, Italy
| | | | - Elena Vigliar
- Public Health, University of Naples Federico II, Naples, Italy
| | - Pierlorenzo Pallante
- Institute of Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", National Research Council, Naples, Italy
| | - Paola Parente
- Unit of Pathology, Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), Padua University Hospital, Padua, Italy.,Veneto Institute of Oncology, IOV IRCCS, Padua, Italy
| | - Paolo Graziano
- Unit of Pathology, Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | | | | | | | | |
Collapse
|
53
|
Walker R, Georgeson P, Mahmood K, Joo JE, Makalic E, Clendenning M, Como J, Preston S, Joseland S, Pope BJ, Hutchinson RA, Kasem K, Walsh MD, Macrae FA, Win AK, Hopper JL, Mouradov D, Gibbs P, Sieber OM, O'Sullivan DE, Brenner DR, Gallinger S, Jenkins MA, Rosty C, Winship IM, Buchanan DD. Evaluating Multiple Next-Generation Sequencing-Derived Tumor Features to Accurately Predict DNA Mismatch Repair Status. J Mol Diagn 2023; 25:94-109. [PMID: 36396080 PMCID: PMC10424255 DOI: 10.1016/j.jmoldx.2022.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/27/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
Abstract
Identifying tumor DNA mismatch repair deficiency (dMMR) is important for precision medicine. Tumor features, individually and in combination, derived from whole-exome sequenced (WES) colorectal cancers (CRCs) and panel-sequenced CRCs, endometrial cancers (ECs), and sebaceous skin tumors (SSTs) were assessed for their accuracy in detecting dMMR. CRCs (n = 300) with WES, where mismatch repair status was determined by immunohistochemistry, were assessed for microsatellite instability (MSMuTect, MANTIS, MSIseq, and MSISensor), Catalogue of Somatic Mutations in Cancer tumor mutational signatures, and somatic mutation counts. A 10-fold cross-validation approach (100 repeats) evaluated the dMMR prediction accuracy for i) individual features, ii) Lasso statistical model, and iii) an additive feature combination approach. Panel-sequenced tumors (29 CRCs, 22 ECs, and 20 SSTs) were assessed for the top performing dMMR predicting features/models using these three approaches. For WES CRCs, 10 features provided >80% dMMR prediction accuracy, with MSMuTect, MSIseq, and MANTIS achieving ≥99% accuracy. The Lasso model achieved 98.3% accuracy. The additive feature approach, with three or more of six of MSMuTect, MANTIS, MSIseq, MSISensor, insertion-deletion count, or tumor mutational signature small insertion/deletion 2 + small insertion/deletion 7 achieved 99.7% accuracy. For the panel-sequenced tumors, the additive feature combination approach of three or more of six achieved accuracies of 100%, 95.5%, and 100% for CRCs, ECs, and SSTs, respectively. The microsatellite instability calling tools performed well in WES CRCs; however, an approach combining tumor features may improve dMMR prediction in both WES and panel-sequenced data across tissue types.
Collapse
Affiliation(s)
- Romy Walker
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Peter Georgeson
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Khalid Mahmood
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Melbourne Bioinformatics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jihoon E Joo
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Enes Makalic
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, Victoria, Australia
| | - Mark Clendenning
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Julia Como
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Susan Preston
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Sharelle Joseland
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Bernard J Pope
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Melbourne Bioinformatics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ryan A Hutchinson
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Kais Kasem
- Department of Clinical Pathology, Medicine Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael D Walsh
- Sullivan Nicolaides Pathology, Bowen Hills, Queensland, Australia
| | - Finlay A Macrae
- Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Melbourne, Victoria, Australia; Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Aung K Win
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, Victoria, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, Victoria, Australia
| | - Dmitri Mouradov
- Personalized Oncology Division, The Walter and Eliza Hall Institute of Medial Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Peter Gibbs
- Personalized Oncology Division, The Walter and Eliza Hall Institute of Medial Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia; Department of Medical Oncology, Western Health, Melbourne, Victoria, Australia
| | - Oliver M Sieber
- Personalized Oncology Division, The Walter and Eliza Hall Institute of Medial Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia; Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Dylan E O'Sullivan
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Darren R Brenner
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada; Department of Cancer Epidemiology and Prevention Research, Alberta Health Services, Calgary, Alberta, Canada
| | - Steven Gallinger
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Mark A Jenkins
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, Victoria, Australia
| | - Christophe Rosty
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Envoi Specialist Pathologists, Brisbane, Queensland, Australia; University of Queensland, Brisbane, Queensland, Australia
| | - Ingrid M Winship
- Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Melbourne, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Melbourne, Victoria, Australia.
| |
Collapse
|
54
|
Pan S, Cox H, Willmott J, Mundt E, Gorringe H, Landon M, Bowles KR, Coffee B, Roa BB, Mancini-DiNardo D. Discordance between germline genetic findings and abnormal tumor immunohistochemistry staining of mismatch repair proteins in individuals with suspected Lynch syndrome. Front Oncol 2023; 13:1069467. [PMID: 36793599 PMCID: PMC9923021 DOI: 10.3389/fonc.2023.1069467] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/11/2023] [Indexed: 01/31/2023] Open
Abstract
Background and Aims Tumor immunohistochemical staining (IHC) of DNA mismatch repair (MMR) proteins is often used to guide germline genetic testing and variant classification for patients with suspected Lynch syndrome. This analysis examined the spectrum of germline findings in a cohort of individuals showing abnormal tumor IHC. Methods We assessed individuals with reported abnormal IHC findings and referred for testing with a six-gene syndrome-specific panel (n=703). Pathogenic variants (PVs) and variants of uncertain significance (VUS) in MMR genes were designated expected/unexpected relative to IHC results. Results The PV positive rate was 23.2% (163/703; 95% confidence interval [CI], 20.1%-26.5%); 8.0% (13/163; 95% CI, 4.3%-13.3%) of PV carriers had a PV in an unexpected MMR gene. Overall, 121 individuals carried VUS in MMR genes expected to be mutated based on IHC results. Based on independent evidence, in 47.1% (57/121; 95% CI, 38.0%-56.4%) of these individuals the VUSs were later reclassified as benign and in 14.0% (17/121; 95% CI, 8.4%-21.5%) of these individuals the VUSs were reclassified as pathogenic. Conclusions Among patients with abnormal IHC findings, IHC-guided single-gene genetic testing may miss 8% of individuals with Lynch syndrome. In addition, in patients with VUS identified in MMR genes predicted to be mutated by IHC, extreme caution must be taken when the IHC results are considered in variant classification.
Collapse
Affiliation(s)
- Shujuan Pan
- Myriad Genetic Laboratories, Inc., Salt Lake City, UT, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Park Y, Nam SK, Seo SH, Park KU, Oh HJ, Park YS, Suh YS, Ahn SH, Park DJ, Kim HH, Lee HS. Comprehensive Study of Microsatellite Instability Testing and Its Comparison With Immunohistochemistry in Gastric Cancers. J Gastric Cancer 2023; 23:264-274. [PMID: 37129151 PMCID: PMC10154139 DOI: 10.5230/jgc.2023.23.e5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/12/2022] [Accepted: 12/23/2022] [Indexed: 01/27/2023] Open
Abstract
PURPOSE In this study, polymerase chain reaction (PCR)-based microsatellite instability (MSI) testing was comprehensively analyzed and compared with immunohistochemistry (IHC) for mismatch repair (MMR) protein expression in patients with gastric cancer (GC). MATERIALS AND METHODS In 5,676 GC cases, PCR-based MSI testing using five microsatellites (BAT-26, BAT-25, D5S346, D2S123, and D17S250) and IHC for MLH1 were performed. Re-evaluation of MSI testing/MLH1 IHC and additional IHC for MSH2, MSH6, and PMS2 were performed in discordant/indeterminate cases. RESULTS Of the 5,676 cases, microsatellite stable (MSS)/MSI-low and intact MLH1 were observed in 5,082 cases (89.5%), whereas MSI-high (MSI-H) and loss of MLH1 expression were observed in 502 cases (8.8%). We re-evaluated the remaining 92 cases (1.6%) with a discordant/indeterminate status. Re-evaluation showed 1) 37 concordant cases (0.7%) (18 and 19 cases of MSI-H/MMR-deficient (dMMR) and MSS/MMR-proficient (pMMR), respectively), 2) 6 discordant cases (0.1%) (3 cases each of MSI-H/pMMR and MSS/dMMR), 3) 14 MSI indeterminate cases (0.2%) (1 case of dMMR and 13 cases of pMMR), and 4) 35 IHC indeterminate cases (0.6%) (22 and 13 cases of MSI-H and MSS, respectively). Finally, MSI-H or dMMR was observed in 549 cases (9.7%), of which 47 (0.8%) were additionally confirmed as MSI-H or dMMR by re-evaluation. Sensitivity was 99.3% for MSI testing and 95.4% for MMR IHC. CONCLUSIONS Considering the low incidence of MSI-H or dMMR, discordant/indeterminate results were occasionally identified in GCs, in which case complementary testing is required. These findings could help improve the accuracy of MSI/MMR testing in daily practice.
Collapse
Affiliation(s)
- Yujun Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Soo Kyung Nam
- Department of Interdisciplinary Program in Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Soo Hyun Seo
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Kyoung Un Park
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hyeon Jeong Oh
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Young Suk Park
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Sang-Hoon Ahn
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Do Joong Park
- Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Hyung-Ho Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hye Seung Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
56
|
Sorokin M, Rabushko E, Rozenberg JM, Mohammad T, Seryakov A, Sekacheva M, Buzdin A. Clinically relevant fusion oncogenes: detection and practical implications. Ther Adv Med Oncol 2022; 14:17588359221144108. [PMID: 36601633 PMCID: PMC9806411 DOI: 10.1177/17588359221144108] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 11/22/2022] [Indexed: 12/28/2022] Open
Abstract
Mechanistically, chimeric genes result from DNA rearrangements and include parts of preexisting normal genes combined at the genomic junction site. Some rearranged genes encode pathological proteins with altered molecular functions. Those which can aberrantly promote carcinogenesis are called fusion oncogenes. Their formation is not a rare event in human cancers, and many of them were documented in numerous study reports and in specific databases. They may have various molecular peculiarities like increased stability of an oncogenic part, self-activation of tyrosine kinase receptor moiety, and altered transcriptional regulation activities. Currently, tens of low molecular mass inhibitors are approved in cancers as the drugs targeting receptor tyrosine kinase (RTK) oncogenic fusion proteins, that is, including ALK, ABL, EGFR, FGFR1-3, NTRK1-3, MET, RET, ROS1 moieties. Therein, the presence of the respective RTK fusion in the cancer genome is the diagnostic biomarker for drug prescription. However, identification of such fusion oncogenes is challenging as the breakpoint may arise in multiple sites within the gene, and the exact fusion partner is generally unknown. There is no gold standard method for RTK fusion detection, and many alternative experimental techniques are employed nowadays to solve this issue. Among them, RNA-seq-based methods offer an advantage of unbiased high-throughput analysis of only transcribed RTK fusion genes, and of simultaneous finding both fusion partners in a single RNA-seq read. Here we focus on current knowledge of biology and clinical aspects of RTK fusion genes, related databases, and laboratory detection methods.
Collapse
Affiliation(s)
| | - Elizaveta Rabushko
- Moscow Institute of Physics and Technology,
Dolgoprudny, Moscow Region, Russia,I.M. Sechenov First Moscow State Medical
University, Moscow, Russia
| | | | - Tharaa Mohammad
- Moscow Institute of Physics and Technology,
Dolgoprudny, Moscow Region, Russia
| | | | - Marina Sekacheva
- I.M. Sechenov First Moscow State Medical
University, Moscow, Russia
| | - Anton Buzdin
- Moscow Institute of Physics and Technology,
Dolgoprudny, Moscow Region, Russia,I.M. Sechenov First Moscow State Medical
University, Moscow, Russia,Shemyakin-Ovchinnikov Institute of Bioorganic
Chemistry, Moscow, Russia,PathoBiology Group, European Organization for
Research and Treatment of Cancer (EORTC), Brussels, Belgium
| |
Collapse
|
57
|
DNA Mismatch Repair Proteins and BRAF V600E Detection by Immunohistochemistry in Colorectal Cancer Demonstrates Concordance with Next Generation Sequencing. JOURNAL OF MOLECULAR PATHOLOGY 2022. [DOI: 10.3390/jmp3040029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background and Aims: Multiple laboratory methods are used to screen patients with colorectal cancer (CRC) for mismatch repair (MMR) protein deficiency to identify possible Lynch syndrome patients. The goal of this study was to compare the agreement between ready-to-use immunohistochemistry (IHC) assays for MLH-1, PMS-2, MSH-2, MSH-6, and mutated BRAF at V600E and molecular methods in CRC cases. The inclusion of the BRAF V600E mutation testing is important for the identification of patients with sporadic CRC, as the BRAF V600E mutation is very rarely observed in patients with Lynch syndrome tumors. Methods: CRC cases were analyzed by ColoSeqTM tumor sequencing assay and VENTANA MMR IHC Panel that included anti-MLH1, anti-PMS2, anti-MSH2, anti-MSH6, and anti-BRAF V600E antibodies. Additionally, CRC cases with MLH1 IHC loss were evaluated for MLH1 promoter hypermethylation. Results: One hundred and eighteen cases were analyzed. The overall percent agreement (OPA) for each evaluated marker status compared to next-generation sequencing (NGS) exceeded 96%. Twenty-three cases were positive for the BRAF V600E mutation by IHC and NGS, and twenty cases showed loss of MLH1 protein and were positive for MLH1 hypermethylation. Samples with loss of MMR protein expression by IHC demonstrated genetic and/or epigenetic alterations that were consistent with the observed protein expression patterns. Conclusions: The results of this study indicate that ready-to-use IHC assays can correctly identify the loss of MMR proteins and the presence of mutated BRAF V600E protein, supporting the utility of the VENTANA MMR IHC Panel as an aid to stratify patients with sporadic CRC vs. potential Lynch syndrome.
Collapse
|
58
|
Molecular Targeted Therapy in Oncology Focusing on DNA Repair Mechanisms. Arch Med Res 2022; 53:807-817. [PMID: 36460545 DOI: 10.1016/j.arcmed.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/17/2022] [Indexed: 11/30/2022]
Abstract
DNA repair mechanisms are essential for maintaining cellular homeostasis. Malfunction of these repair mechanisms leads to cellular DNA mutations, carcinogenic transformation, and cell death. These same defects also create vulnerabilities that are relatively specific to cancer cells, and which could potentially be exploited to increase the therapeutic index of anticancer treatments and thereby improve patient outcomes. The targeted therapy based on inhibiting the DNA damage response (DDR) opens a new therapeutic landscape for patients with deficient DDR. Currently there are two DNA repair mechanisms that are used as targets for molecular therapies: Mitsmach Repair (MMR) and Homologous Recombination Repair (HRR). These molecular targets allow for immunotherapy treatments based on "checkpoint inhibitors" (ICIs) drugs and "PARP inhibitor" (PARPi) drugs in different solid tumors. In this review we will describe the state of the art of this interesting mechanism and explain the options for treatment based on these alterations. Moreover, many clinical trials are currently underway exploring better treatment options for dMMR and HRD patients with different solid tumours.
Collapse
|
59
|
Salman FG, Kankaya D, Özakıncı H, Şahin Y, Kubilay E, Süer E, Hayme S, Baltacı S. Role of PD-1/PD-L1-mediated tumour immune escape mechanism and microsatellite instability in the BCG failure of high-grade urothelial carcinomas. Turk J Med Sci 2022; 52:1802-1813. [PMID: 36945968 PMCID: PMC10390201 DOI: 10.55730/1300-0144.5526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 10/09/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Intravesical BCG treatment fails inexplicably in 30%-45% of patients for high-grade nonmuscle-invasive bladder cancer (NMIBC). We aimed to investigate the role of PD-1/PD-L1 interaction on BCG failure of high-grade NMIBC and to identify biomarkers for predicting BCG responsive cases. METHODS Thirty BCG responsive and 29 nonresponsive NMIBCs were included in the study. Expressions of PDL1(SP-263), MSH2, MSH6, PMS2, and MLH1 were evaluated on pre- and post-BCG transurethral resection (TUR-B) specimens by immunohistochemistry. PD-L1(SP-263) expression was categorised as negative/low, high. DNA mismatch repair protein (MMR) expressions were classified as "reduced" if ≤30% of nuclei stained, "preserved" if >30% of nuclei stained. Microsatellite instability (MSI) testing was performed by PCR using five mononucleotide markers. RESULTS Reduced DNA MMR protein expression was found to be significantly higher in the pretreatment biopsies of BCG-responsive group than the BCG nonresponsive tumour group (p = 0.022). PD-L1 expression did not show any significant difference between the pre- and posttreatment TUR-B specimens of the BCG nonresponsive tumour group or between the pretreatment TUR-B specimens of BCG nonresponsive and the BCG responsive groups (p = 0.508, p = 0.708, respectively). DISCUSSION Immune escape of tumour cells by PD-1/PD-L1 interaction does not seem to have any role in BCG failure of NMIBCs. Reduced MMR expression may help to determine cases that will respond well to BCG therapy. A better antitumour activity of BCG in NMIBCs with reduced MMR expression may be related to the ongoing accumulation of cancer neoantigens in correlation with increased tumour mutation load as a result of DNA repair defects.
Collapse
Affiliation(s)
- Fadime Gül Salman
- Department of Pathology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Duygu Kankaya
- Department of Pathology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Hilal Özakıncı
- Department of Pathology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Yasemin Şahin
- Department of Pathology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Eralp Kubilay
- Department of Urology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Evren Süer
- Department of Urology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Serhat Hayme
- Department of Biostatistics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Sümer Baltacı
- Department of Urology, Faculty of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
60
|
Qin S, Li J, Zhong H, Jin C, Chen L, Yuan X, Fan Q, Chen K, Cao P, Xiao J, Jiang D, Zhang T, Zhang H, Wang X, Wang W, Han L, Wang Q, Zhu J. Serplulimab, a novel anti-PD-1 antibody, in patients with microsatellite instability-high solid tumours: an open-label, single-arm, multicentre, phase II trial. Br J Cancer 2022; 127:2241-2248. [PMID: 36261583 PMCID: PMC9726893 DOI: 10.1038/s41416-022-02001-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 09/07/2022] [Accepted: 09/23/2022] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Microsatellite instability-high/mismatch repair-deficient (MSI-H/dMMR) tumours have a high response rate to immunotherapy. Antitumour activity and safety of serplulimab, a novel humanised anti-PD-1 monoclonal antibody, were evaluated in this phase II study. METHODS In this ongoing, single-arm, open-label, phase II trial, patients with previously treated unresectable or metastatic MSI-H/dMMR solid tumours received intravenous serplulimab 3 mg/kg every 2 weeks for up to 52 cycles. The primary endpoint was objective response rate (ORR) assessed by an independent radiological review committee per Response Evaluation Criteria in Solid Tumors v1.1. Secondary endpoints included additional efficacy measures, safety, and tolerability. RESULTS As of 9 January 2021, 108 patients were enrolled, and 68 patients with confirmed MSI-H solid tumours were included in the main efficacy analysis population (MEAP). The median follow-up duration in the MEAP was 7.7 months, with an ORR of 38.2% (95% confidence interval, 26.7-50.8). Of the 108 patients, grade ≥3 treatment-emergent adverse events were reported in 53 (49.1%) patients; immune-related adverse events occurred in 52 (48.1%) patients. CONCLUSIONS Serplulimab demonstrates a durable antitumour effect and a manageable safety profile in previously treated patients with MSI-H solid tumours. Serplulimab is a promising tissue-agnostic treatment for previously treated MSI-H solid tumours. TRIAL REGISTRATION NCT03941574.
Collapse
Affiliation(s)
- Shukui Qin
- Department of Oncology, Qinhuai Medical Area, Eastern Theater General Hospital of PLA China, Nanjing, China
| | - Jin Li
- Department of Oncology, Tongji University Shanghai East Hospital, Shanghai, China.
| | - Haijun Zhong
- Department of Abdominal Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Chuan Jin
- Department of Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Lili Chen
- Department of Hematology and Oncology, Taizhou First People's Hospital, Taizhou, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qingxia Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kehe Chen
- Department of Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Peiguo Cao
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jianjun Xiao
- Chemotherapeutic Department, Zhongshan City People's Hospital, Zhongshan, China
| | - Da Jiang
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tao Zhang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongyu Zhang
- Cancer Center, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, China
| | - Xicheng Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Wei Wang
- Department of Gastrointestinal Oncology, The First People's Hospital of Foshan, Foshan, China
| | - Lin Han
- Shanghai Henlius Biotech, Inc., Shanghai, China
| | - Qingyu Wang
- Shanghai Henlius Biotech, Inc., Shanghai, China
| | - Jun Zhu
- Shanghai Henlius Biotech, Inc., Shanghai, China
| |
Collapse
|
61
|
Detecting mismatch repair deficiency in solid neoplasms: immunohistochemistry, microsatellite instability, or both? Mod Pathol 2022; 35:1515-1528. [PMID: 35668150 DOI: 10.1038/s41379-022-01109-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 12/25/2022]
Abstract
In managing patients with solid tumors, the value of detecting the status of tumor DNA mismatch repair function is widely recognized. Mismatch repair protein immunohistochemistry and molecular microsatellite instability testing constitute the two major test modalities currently in use, yet each is associated with caveats and limitations that can be consequential. Most notably, the traditional approach of defining mismatch repair protein immunohistochemistry abnormality by complete loss of staining in all tumor cells is evolving. Partial or clonal loss is becoming recognized as a manifestation of gene abnormality; in some cases, such clonal loss is associated with germline pathogenic variants. The current criteria and cutoff values for defining microsatellite instability-high are developed primarily according to colorectal tumors. Non-colorectal cases, and occasionally even colorectal tumors, that are mismatch repair-deficient by immunohistochemistry but not microsatellite instability-high by current standards are being recognized. Emerging data suggest that these immunohistochemistry abnormal / non-microsatellite instability-high cases warrant further genetic workup for Lynch syndrome detection. Whether these tumors respond to immunotherapy is a question still to be addressed. It is imperative that pathologists as well as clinicians and investigators be aware of such intricacies regarding routine immunohistochemistry and microsatellite instability testing and the results they generate. This review summarizes our current understanding of the advantages and limitations of these tests and offer our view on what constitutes the most optimal strategy in test selection and how best to utilize case context to enhance the interpretation of the test results.
Collapse
|
62
|
Catalano M, Francesco Iannone L, Cosso F, Generali D, Mini E, Roviello G. Combining inhibition of immune checkpoints and PARP: rationale and perspectives in cancer treatment. Expert Opin Ther Targets 2022; 26:923-936. [PMID: 36519314 DOI: 10.1080/14728222.2022.2158813] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Genomic instability resulting from the inability of cells to repair DNA damage is a breeding ground for immune checkpoint inhibitors (ICIs) and targeted treatments. Poly (ADP-ribose) polymerase inhibitors (PARPi) interfere with the efficient repair of DNA single-strand break damage inducing, mainly in tumors with existing defects in double strand DNA repair system, synthetic lethality. AREAS COVERED By amplifying the DNA damage and inducing immunogenic cell death PARPi leads tumor neoantigens to increase, upregulation of programmed death-ligand 1, and modulation of the tumor microenvironment facilitating a more intense antitumor immune response. In this review, we reported the immunological role of PARPi and the rational use of the combination with ICIs, evaluating data from combination clinical trials and discussing perspectives. EXPERT OPINION Several prospective combination studies to overcome existing limitations to PARPi and ICI single agents are currently ongoing. The identification of the different resistance mechanisms to PARPi and ICI as well as the development of accurate and predictive biomarkers of response should be a priority to identify the patients who may most benefit from this combination. Similarly, clarifying the role and interaction between the DNA damage repair pathways and the tumor immune microenvironment would increase success of the combination.
Collapse
Affiliation(s)
- Martina Catalano
- School of Human Health Sciences, University of Florence, Florence, Italy
| | - Luigi Francesco Iannone
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Federica Cosso
- School of Human Health Sciences, University of Florence, Florence, Italy
| | - Daniele Generali
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Enrico Mini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
63
|
Flecchia C, Zaanan A, Lahlou W, Basile D, Broudin C, Gallois C, Pilla L, Karoui M, Manceau G, Taieb J. MSI colorectal cancer, all you need to know. Clin Res Hepatol Gastroenterol 2022; 46:101983. [PMID: 35732266 DOI: 10.1016/j.clinre.2022.101983] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/18/2022] [Indexed: 02/04/2023]
Abstract
Colorectal cancer management has been dramatically impacted by molecular profiling these last years. Among these molecular subgroups, patients with microsatellite instability (MSI) are of particular interest, owing to the prognostic and predictive value of this tumor biomarker. This review article explains the molecular abnormalities underlying MSI phenotype and the consequences of such molecular abnormalities on carcinogenesis, genetic instability and immune infiltration. It details the diagnostic methods for identifying MSI colorectal cancer patients and describes how the prognostic and theranostic values of this marker are impacting treatment decision-making for these patients in 2022.
Collapse
Affiliation(s)
- Clémence Flecchia
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, AP-HP, Paris-Cité University, Paris, France
| | - Aziz Zaanan
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, AP-HP, Paris-Cité University, Paris, France
| | - Widad Lahlou
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, AP-HP, Paris-Cité University, Paris, France
| | - Debora Basile
- Department of Oncology, San Bortolo General Hospital, AULSS8 Berica, Vicenza, Italy
| | - Chloé Broudin
- Department of Pathology, European Georges Pompidou Hospital, AP-HP, Paris, France
| | - Claire Gallois
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, AP-HP, Paris-Cité University, Paris, France
| | - Lorenzo Pilla
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, AP-HP, Paris-Cité University, Paris, France
| | - Mehdi Karoui
- Department of Surgery, European Georges Pompidou Hospital, AP-HP, Paris, France
| | - Gilles Manceau
- Department of Surgery, European Georges Pompidou Hospital, AP-HP, Paris, France
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, AP-HP, Paris-Cité University, Paris, France.
| |
Collapse
|
64
|
Reitsam NG, Märkl B, Dintner S, Waidhauser J, Vlasenko D, Grosser B. Concurrent loss of MLH1, PMS2 and MSH6 immunoexpression in digestive system cancers indicating a widespread dysregulation in DNA repair processes. Front Oncol 2022; 12:1019798. [PMID: 36387226 PMCID: PMC9643848 DOI: 10.3389/fonc.2022.1019798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/04/2022] [Indexed: 11/24/2022] Open
Abstract
Immunohistochemical analysis of mismatch repair (MMR) protein expression is widely used to identify tumors with a deficient MMR (dMMR). MMR proteins (MLH1/PMS2 and MSH2/MSH6) work as functional heterodimers, which usually leads to the loss of expression in only one functional MMR heterodimer. Recently, there have been studies showing the simultaneous loss of immunoexpression in proteins of both heterodimers. Yet, this phenomenon has been rarely investigated. In this study, we retrospectively considered cases of different digestive system cancers (gastric cancer, ampullary cancer, small bowel cancer, colorectal cancer), which were immunohistochemically tested for dMMR within a 4-year period at our university hospital (n=352). Of the 103 cases showing dMMR, 5 cases (1.4% of all, 5.1% of dMMR cases) showed a concurrent loss of MLH1, PMS2 and MSH6 immunoexpression, whereas in the other 98 dMMR cases only one MMR heterodimer was affected. MLH1-/PMS2-/MSH6- cancer cases almost arose throughout the entire digestive tract: from the gastric antrum to the left colic flexur. To provide a comprehensive molecular characterization of this MLH1-/PMS2-/MSH6- immunophenotype, tumors were analyzed for microsatellite instability, MLH1 promotor hypermethylation and BRAF exon 15 status. Furthermore, we performed next-generation sequencing focusing on genes related to DNA repair. Here, we could detect pathogenic germline variants as well as multiple sporadic mutations in different genes involved in MMR and homologous recombination repair (HRR) respectively. The affected MMR/HRR-related genes were: ATM, BARD1, BRCA1, CDK12, CHEK1, CHEK2, FANCA, MLH1, MSH6, PALB2, TP53. Considering the biologic function of HRR/MMR proteins as potential drug targets and the low frequency of most of these mutations in digestive system cancers in general, their common occurrence in our MLH1-/PMS2-/MSH6- cases seems to be even more noteworthy, highlighting the need for recognition, awareness and further investigation of this unusual IHC staining pattern.
Collapse
Affiliation(s)
- Nic Gabriel Reitsam
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Augsburg, Germany
- *Correspondence: Nic Gabriel Reitsam, ;
| | - Bruno Märkl
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Sebastian Dintner
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Johanna Waidhauser
- Department of Hematology and Oncology, University Medical Center Augsburg / University Hospital of Augsburg, Augsburg, Germany
| | - Dmytro Vlasenko
- General, Visceral and Transplantation Surgery, University Hospital of Augsburg, Augsburg, Germany
| | - Bianca Grosser
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Augsburg, Germany
| |
Collapse
|
65
|
Biallelic PMS2 Mutations in a Family with Uncommon Clinical and Molecular Features. Genes (Basel) 2022; 13:genes13111953. [PMID: 36360190 PMCID: PMC9690098 DOI: 10.3390/genes13111953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/20/2022] [Accepted: 10/23/2022] [Indexed: 11/29/2022] Open
Abstract
We describe a patient with constitutional mismatch repair-deficiency (CMMR-D) in whom the syndrome started at age 10 with the development of multiple adenomas in the large bowel. In the successive 25 years, four malignancies developed in different organs (rectum, ileum, duodenum, and lymphoid tissue). The patient had biallelic constitutional pathogenic variants in the PMS2 gene. We speculate that besides the PMS2 genotype, alterations of other genes might have contributed to the development of the complex phenotype. In the nuclear family, both parents carried different PMS2 germline mutations. They appeared in good clinical condition and did not develop polyps or cancer. The index case had a brother who died at age three of lymphoblastic leukemia, and a sister who was affected by sarcoidosis. Tumor tissue showed diffuse DNA microsatellite instability. A complete absence of immunoreactivity was observed for the PMS2 protein both in the tumors and normal tissues. Next-generation sequencing and multiple ligation-dependent probe amplification analyses revealed biallelic PMS2 germline pathogenic variants in the proband (genotype c.[137G>T];[(2174+1_2175-1)_(*160_?)del]), and one of the two variants was present in both parents—c.137G>T in the father and c.(2174+1-2175-1)_(*160_?)del in the mother—as well as c.137G>T in the sister. Moreover, Class 3 variants of MSH2 (c.1787A>G), APC (c.1589T>C), and CHEK2 (c.331G>T) genes were also detected in the proband. In conclusion, the recognition of CMMR-D may sometimes be difficult; however, the possible role of constitutional alterations of other genes in the development of the full-blown phenotype should be investigated in more detail.
Collapse
|
66
|
Xie Y, Jiao X, Zeng M, Fan Z, Li X, Yuan Y, Zhang Q, Xia Y. Clinical Significance of Fusobacterium nucleatum and Microsatellite Instability in Evaluating Colorectal Cancer Prognosis. Cancer Manag Res 2022; 14:3021-3036. [PMID: 36262751 PMCID: PMC9576466 DOI: 10.2147/cmar.s382464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/04/2022] [Indexed: 02/05/2023] Open
Abstract
Objective Both genetic and microbial factors play important roles in colorectal cancer (CRC) development. The effects of Fusobacterium nucleatum (F. nucleatum) and microsatellite instability (MSI) on CRC prognosis require more clinical evidence. We aimed to investigate the role of F. nucleatum and MSI as biomarkers in predicting the prognosis of CRC. Methods CRC patients in various TNM stages were enrolled. MSI status and F. nucleatum were detected by immunohistochemical staining of formalin-fixed paraffin-embedded (FFPE) specimens. The associations between MSI status and F. nucleatum and clinical parameters were analyzed. Results MSI tumors were more frequently observed in the colon than in the rectum. Cancerous tissues had higher levels of F. nucleatum than adjacent noncancerous tissues. There were no significant differences in F. nucleatum abundance in different age, sex, tumor stage, location, and tumor marker groups. MSI status was associated with tumor location and stage. Survival analyses revealed that disease-free survival (DFS) was significantly longer in the F. nucleatum-negative, younger age, and TNM stage I-II groups (p< 0.05), and age, advanced TNM stage (III and IV), and F. nucleatum status were independent factors for poor prognosis. Multivariate Cox regression and receiver operating characteristic (ROC) curve analyses showed that conventional tumor biomarkers of CRC had more prognostic value than F. nucleatum and MSI. Conclusion Age, advanced TNM stage, and F. nucleatum positivity were independent factors of poor prognosis, suggesting that F. nucleatum and MSI may contribute to the identification of new strategies for the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Yanxuan Xie
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, People’s Republic of China
| | - Xiaoyang Jiao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, 515041, People’s Republic of China
| | - Mi Zeng
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, 515041, People’s Republic of China
| | - Zhiqiang Fan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, 515041, People’s Republic of China
| | - Xin Li
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, 515041, People’s Republic of China
| | - Yumeng Yuan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, 515041, People’s Republic of China
| | - Qiaoxin Zhang
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, People’s Republic of China
| | - Yong Xia
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, People’s Republic of China
| |
Collapse
|
67
|
Atjimakul T, Wattanapaisal P, Suwiwat S, Wanichsuwan W, Hanprasertpong J. Microsatellite instability and oncological outcomes in Thai patients with endometrial cancer. J OBSTET GYNAECOL 2022; 42:3117-3123. [PMID: 35930016 DOI: 10.1080/01443615.2022.2106557] [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: 01/06/2023]
Abstract
Here, we determined the frequency of microsatellite instability (MSI) and the impact of MSI-high (MSI-H) on clinical outcomes of Thai patients with endometrial cancer (EC). Tissue samples of 110 Thai patients with EC, who had undergone surgical staging, were tested for mismatch repair (MMR) gene deficiency, and the patients were grouped into MSI-H and MSI-stable (MSI-S) groups; 24.5% had MSI-H. Unlike MSI-S group patients, MSI-H group patients had synchronous and metachronous cancer. They showed better 3-year disease-free survival (DFS) than those in the MSI-S group (p=.182; 92.3% vs. 82.6%). The 3-year overall survival was 96.2% in MSI-H and 86.4% in MSI-S groups (p=.163). Multivariate analyses showed lower uterine involvement (p=.004), myometrial invasion ≥50% (p=.032), lymphovascular space invasion (p<.001) and MSI-S (p=.006) as prognostic factors for DFS. Our study showed that the prevalence of MMR gene deficiency in Thai patients with EC is common and associated with better outcomes.Impact StatementWhat is already known on this subject? Microsatellite instability (MSI) occurs in approximately 20-40% of endometrial cancer (EC) cases. MSI analysis in EC can identify patients at higher risk of hereditary nonpolyposis colorectal cancer and those having prognostic factors. Additionally, it is predictive of immune checkpoint inhibitor treatment. However, current evidence shows a correlation between clinicopathological characteristics and EC prognosis. Studies on EC and MSI status effect on survival outcome have yielded inconsistent results regarding the pathological significance of MSI in such malignancies.What do the results of this study add? The prevalence of mismatch repair (MMR) gene deficiency in Thai patients with EC is common (24.5%) and associated with better outcomes.What are the implications of these findings for clinical practice and/or further research? This study highlights the prevalence and impact of MSI on oncological outcomes in patients with EC in a low-incidence country. Future studies should focus on the detection of germline mutation to understand the accurate prevalence of Lynch syndrome in Thai patients with EC.
Collapse
Affiliation(s)
- Thiti Atjimakul
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Panote Wattanapaisal
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Supaporn Suwiwat
- Department of Pathology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Worrawit Wanichsuwan
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Jitti Hanprasertpong
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand.,Department of Research and Medical Innovation, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| |
Collapse
|
68
|
Combination of Tumor Mutational Burden and DNA Damage Repair Gene Mutations with Stromal/Immune Scores Improved Prognosis Stratification in Patients with Lung Adenocarcinoma. JOURNAL OF ONCOLOGY 2022; 2022:6407344. [PMID: 36262349 PMCID: PMC9576425 DOI: 10.1155/2022/6407344] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 07/24/2022] [Indexed: 12/24/2022]
Abstract
Background Both the tumor environment and the genomic landscape of lung cancer may shape patient responses to treatments, including immunotherapy, but their joint impacts on lung adenocarcinoma (LUAD) prognosis are underexplored. Methods RNA sequencing data and whole-exome sequencing results were downloaded from the TCGA database, and only LUAD-related data were included in this study. Based on gene expression data, the ESTIMATE algorithm was used to estimate stromal and immune scores, and CIBERSORT analysis was used for quantification of the relative abundances of immune cells. Somatic mutations were used for calculating tumor mutation burden (TMB). Specific mutations in genes involved in DNA damage repair (DDR) pathways were identified. The individual and joint associations of stromal and immune score, TMB, and DDR gene mutations with 5-year survival were analyzed by the Kaplan–Meier method and multivariate Cox model. Results LUAD patients with a high (>highest 25%) stromal or immune score had prolonged survival as compared to those with a low (<lowest 25%) score (log-rank P=0.05 and 0.035, respectively). Patients with both high stromal and immune scores had the most favorable survival. Although the survival differences between patients with high (>highest 25%) and low (<lowest 25%) TMB, or between patients with mutant- and wild-type DDR genes were not statistically significant, a survival benefit from high TMB or DDR gene mutations was observed in patients with high stromal or immune scores. Conclusion A comprehensive evaluation of transcriptomic signatures and genomic biomarkers may provide a novel avenue for improving prognosis stratification in LUAD.
Collapse
|
69
|
Clinical Testing for Mismatch Repair in Neoplasms Using Multiple Laboratory Methods. Cancers (Basel) 2022; 14:cancers14194550. [PMID: 36230473 PMCID: PMC9559284 DOI: 10.3390/cancers14194550] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/11/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Background: A deficiency in DNA mismatch repair function in neoplasms can be assessed by an immunohistochemical (IHC) analysis of the deficiency/loss of the mismatch repair proteins (dMMR) or by PCR-based methods to assess high microsatellite instability (MSI-H). In some cases, however, there is a discrepancy between the IHC and MSI analyses. Several studies have addressed the issue of discrepancy between IHC and MSI deficiency assessment, but there are limited studies that also incorporate genetic/epigenetic alterations. Methods: In this single-institution retrospective chart-review study, we reviewed 706 neoplasms assessed between 2015 and 2021. All eligible neoplasms were assessed by IHC testing, MSI analysis by PCR-based assay, and tumor-normal paired next-generation sequencing (NGS) analysis. Eighty percent of neoplasms with MLH1 protein loss had a concurrent MLH1 promoter methylation analysis. Mutation data for MMR genes, IHC, MSI analysis, and tumor histology were correlated with each other. Results: Fifty-eight (8.2%) of 706 neoplasms had MSI-H by PCR and/or dMMR by IHC. Of the 706 analyzed neoplasms, 688 neoplasms (98%) had concordant results: MSI-H/dMMR (n = 44), microsatellite-stable (MSS)/proficient MMR (pMMR) (n = 625), and MSI-Low (L)/pMMR (n = 19). Of the remaining 18 neoplasms, 9 had a major discordance: MSS/loss of MSH2 and MSH6 (n = 3), MSS/loss of MSH6 (n = 2), MSS/Loss of MLH1 and PMS2 (n = 1), and MSI-High/pMMR (n = 3). In total, 57% of cases with dMMR and 61% of cases with MSI-H had a null mutation of an MMR gene mutation (or methylation of the MLH1 promoter), whereas this figure was 1% for neoplasms with a normal IHC or MSI pattern (p < 0.001). Among 9 cases with major discordance between MSI and IHC, only 3 cases (33%) had an underlying genetic/epigenetic etiology, whereas 37 (76%) of 49 cases with MSI-H and/or dMMR and without major discordance had an underlying genetic abnormality (p = 0.02). Discussion: For most neoplasms, IHC and PCR-based MSI testing results are concordant. In addition, an underlying genetic abnormality (a null mutation of an MMR gene or MLH1 promoter methylation) was attributable to dMMR and/or MSI-H findings. For neoplasms with major discordance in IHC and MSI testing, the addition and integration of NGS results and MLH1 promoter methylation analyses can be beneficial for resolving borderline cases, thereby facilitating patient management.
Collapse
|
70
|
Tanaka T, Takehara K, Yamashita N, Okazawa-Sakai M, Kuraoka K, Teramoto N, Taguchi K, Yamashiro K, Kato H, Mizunoe T, Suzuki R, Yamamoto D, Ueki A, Saito T. Frequency and clinical features of deficient mismatch repair in ovarian clear cell and endometrioid carcinoma. J Gynecol Oncol 2022; 33:e67. [PMID: 36032025 PMCID: PMC9428302 DOI: 10.3802/jgo.2022.33.e67] [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: 05/17/2021] [Revised: 04/20/2022] [Accepted: 06/12/2022] [Indexed: 11/30/2022] Open
Abstract
Objective To clarify the frequency of deficient mismatch repair (dMMR) in Japanese ovarian cancer patients, we examined microsatellite instability (MSI) status and immunohistochemistry (IHC) subtypes, including endometrioid carcinoma (EMC), clear cell carcinoma (CCC), or a mixture of both (Mix). Methods We registered 390 patients who were diagnosed with EMC/CCC/Mix between 2006 and 2015 and treated at seven participating facilities. For 339 patients confirmed eligible by the Central Pathological Review Board, MSI, IHC, and MutL homolog 1 methylation analyses were conducted. The tissues of patients with Lynch syndrome (LS)-related cancer histories, such as colorectal and endometrial cancer, were also investigated. Results MSI-high (MSI-H) status was observed in 2/217 CCC (0.9%), 10/115 EMC (8.7%), and 1/4 Mix (25%). Additionally, loss of MMR protein expression (LoE-MMR) was observed in 5/219 (2.3%), 16/115 (14.0%), and 1/4 (25%) patients with CCC, EMC, and Mix, respectively. Both MSI-H and LoE-MMR were found significantly more often in EMC (p<0.001). The median (range) ages of patients with MMR expression and LoE-MMR were 54 (30–90) and 46 (22–76) (p=0.002), respectively. In the multivariate analysis, advanced stage and histological type were identified as prognostic factors. Conclusion The dMMR rate for EMC/CCC was similar to that reported in Western countries. In Japan, it is assumed that the dMMR frequency is higher because of the increased proportion of CCC. The deficient mismatch repair (dMMR) rate in Japanese patients is currently unknown. This study determined the frequency of dMMR in Japanese ovarian cancer patients. The dMMR rate was similar to that reported in Western countries. We identified potential criteria for implementing microsatellite instability and immunohistochemistry analyses in Lynch syndrome screening.
Collapse
Affiliation(s)
- Tamaki Tanaka
- Department of Perinatology and Gynecology, Kagawa University Graduate School of Medicine, Kida, Japan.,Department of Gynecologic Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Kazuhiro Takehara
- Department of Gynecologic Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan.
| | - Natsumi Yamashita
- Department of Clinical Research Center, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Mika Okazawa-Sakai
- Department of Gynecologic Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Kazuya Kuraoka
- Department of Diagnostic Pathology, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Norihiro Teramoto
- Department of Pathology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Kenichi Taguchi
- Department of Pathology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | | | - Hidenori Kato
- Department of Gynecologic Oncology, National Hospital Organization Hokkaido Cancer Center, Sapporo, Japan
| | - Tomoya Mizunoe
- Department of Obstetrics and Gynecology, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Rie Suzuki
- Department of Obstetrics and Gynecology, National Defense Medical Collage Hospital, Tokorozawa, Japan
| | - Dan Yamamoto
- Department of Gynecology, National Hospital Organization Fukuyama Medical Center, Fukuyama, Japan
| | - Arisa Ueki
- Department of Clinical Genetic Oncology, Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Toshiaki Saito
- Gynecology Service, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| |
Collapse
|
71
|
Bounous VE, Robba E, Perotto S, Pasini B, Tomasi Cont N, Ricci MT, Ditto A, Vitellaro M, Raspagliesi F, Biglia N. Gynecological Cancers in Lynch Syndrome: A Comparison of the Histological Features with Sporadic Cases of the General Population. J Clin Med 2022; 11:jcm11133689. [PMID: 35806973 PMCID: PMC9267402 DOI: 10.3390/jcm11133689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/21/2022] Open
Abstract
Introduction: About 5% of endometrial cancers (ECs) are attributed to an inherited predisposition, for which Lynch syndrome (LS) accounts for the majority of cases. Women with LS have a 40−60% predicted lifetime risk of developing EC, in addition to a 40−80% lifetime risk of developing colorectal cancer and other cancers. In this population, the lifetime risk of developing ovarian cancer (OC) is 10−12%. Object: to compare the histopathological features of LS-associated EC and OC with sporadic cancers in order to evaluate whether there are differences in terms of age at diagnosis, site of occurrence in the uterus, histological type, stage at diagnosis, and tumor grading. Materials and methods: we compared data obtained from 96 patients with LS-associated gynecological cancers (82 with EC and 14 with OC) to a control group (CG) of 209 patients who developed sporadic EC, and a CG of 187 patients with sporadic OC. Results: The mean age at diagnosis of LS-associated EC and OC was much lower than in the control groups. In both groups with EC, the endometrioid histotype was the most frequently occurring histotype. However, among LS women there was a significantly higher incidence of clear cell tumors (11% versus 2.4% in the CG, p = 0.0001). Similar to the sporadic cancer cases, most of the LS-associated ECs presented at an early stage (89% of cases at FIGO I-II stage). In the LS group, the tumor frequently involved only the inner half of the endometrium (77% of cases, p < 0.01). In the LS group, 7.3% of ECs were localized to the lower uterine segment (LUS), whereas no cancer developed in the LUS in the CG. No serous OCs were diagnosed in the LS group (versus 45.5% in the CG, p = 0.0009). Most of the LS-associated OCs presented at an early stage (85% of cases at FIGO I-II stages, p < 0.01). Conclusion: LS-associated EC and OC seem to have peculiar features, occurring at a younger age and at an earlier stage. In LS, EC less frequently involves the outer half of the endometrium, with a more frequent occurrence in the LUS. The presence of clear cell EC was more frequently observed, whereas in OC, the predominant histotype was endometrioid.
Collapse
Affiliation(s)
- Valentina Elisabetta Bounous
- Academic Division of Obstetrics and Gynecology—A.O. Ordine Mauriziano, University of Turin, 10128 Turin, Italy; (E.R.); (N.B.)
- Correspondence:
| | - Elisabetta Robba
- Academic Division of Obstetrics and Gynecology—A.O. Ordine Mauriziano, University of Turin, 10128 Turin, Italy; (E.R.); (N.B.)
| | | | - Barbara Pasini
- Department of Genetics, Biology and Biochemistry, University of Turin, 10128 Turin, Italy;
| | | | - Maria Teresa Ricci
- Unit of Hereditary Digestive Tract Tumors, Fondazione IRCCs—National Cancer Institute, 20133 Milan, Italy; (M.T.R.); (M.V.)
| | - Antonino Ditto
- Division of Gynecologic Oncology, Fondazione IRCCs—National Cancer Institute, 20133 Milan, Italy; (A.D.); (F.R.)
| | - Marco Vitellaro
- Unit of Hereditary Digestive Tract Tumors, Fondazione IRCCs—National Cancer Institute, 20133 Milan, Italy; (M.T.R.); (M.V.)
| | - Francesco Raspagliesi
- Division of Gynecologic Oncology, Fondazione IRCCs—National Cancer Institute, 20133 Milan, Italy; (A.D.); (F.R.)
| | - Nicoletta Biglia
- Academic Division of Obstetrics and Gynecology—A.O. Ordine Mauriziano, University of Turin, 10128 Turin, Italy; (E.R.); (N.B.)
| |
Collapse
|
72
|
Adeleke S, Haslam A, Choy A, Diaz-Cano S, Galante JR, Mikropoulos C, Boussios S. Microsatellite instability testing in colorectal patients with Lynch syndrome: lessons learned from a case report and how to avoid such pitfalls. Per Med 2022; 19:277-286. [PMID: 35708161 DOI: 10.2217/pme-2021-0128] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We present the case of a patient with Lynch syndrome and metastatic colorectal carcinoma (mCRC). The initial immunohistochemistry (IHC) test for deficient mismatch repair gave a false negative result. However, the same mutation has accurately has been detected with IHC in other cancers with microsatellite instability (MSI) This supports the determining role of somatic missense mutations in MMR IHC. MSI-PCR testing confirmed MSI and the patient benefited from nivolumab with a complete metabolic response. We explain the rationale for immunotherapy in mCRC, current testing strategies and discuss future developments in MSI testing. We advocate for upfront testing using both IHC and MSI-PCR to direct therapy in mCRC, and a greater understanding of IHC and MSI-PCR testing pitfalls.
Collapse
Affiliation(s)
- Sola Adeleke
- High Dimensional Neurology Group, UCL Queen's Square Institute of Neurology, London, WC1N 3BG, UK.,Department of Oncology, Guy's & St Thomas' Hospital, London, UK.,School of Cancer & Pharmaceutical Sciences, King's College London, Strand, London, WC2R 2LS, UK
| | - Aidan Haslam
- South Bristol Academy, Dolphin House, Bristol Royal Infirmary, Marlborough Street, BS2 8HW, UK
| | - Adrian Choy
- Department of Oncology, Oxford University Hospitals, NHS Foundation Trust, Headington, Oxford, OX3 7DQ, UK
| | - Salvador Diaz-Cano
- Department of Cellular Pathology, University Hospitals Birmingham NHS Foundation Trust, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK.,Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent, ME7 5NY, UK
| | - Joao R Galante
- Maidstone Hospital, Hermitage Lane, Maidstone, Kent, ME16 9QQ, UK
| | - Christos Mikropoulos
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent, ME7 5NY, UK
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent, ME7 5NY, UK.,King's College London, Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, London, SE1 9RT, UK.,AELIA Organization, 9th Km Thessaloniki, Thermi, Thessaloniki, 57001, Greece
| |
Collapse
|
73
|
Vasseur D, Sassi H, Bayle A, Tagliamento M, Besse B, Marzac C, Arbab A, Auger N, Cotteret S, Aldea M, Blanc-Durand F, Géraud A, Gazzah A, Loriot Y, Hollebecque A, Martín-Romano P, Ngo-Camus M, Nicotra C, Ponce S, Sakkal M, Caron O, Smolenschi C, Micol JB, Italiano A, Rouleau E, Lacroix L. Next-Generation Sequencing on Circulating Tumor DNA in Advanced Solid Cancer: Swiss Army Knife for the Molecular Tumor Board? A Review of the Literature Focused on FDA Approved Test. Cells 2022; 11:cells11121901. [PMID: 35741030 PMCID: PMC9221453 DOI: 10.3390/cells11121901] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/01/2022] [Accepted: 06/10/2022] [Indexed: 11/16/2022] Open
Abstract
FDA-approved next-generation sequencing assays based on cell-free DNA offers new opportunities in a molecular-tumor-board context thanks to the noninvasiveness of liquid biopsy, the diversity of analyzed parameters and the short turnaround time. It gives the opportunity to study the heterogeneity of the tumor, to elucidate complex resistance mechanisms and to adapt treatment strategies. However, lowering the limit of detection and increasing the panels' size raise new questions in terms of detection of incidental germline alterations, occult malignancies and clonal hematopoiesis of indeterminate potential mutations. In this review, after a technological discussion and description of the common problematics encountered, we establish recommendations in properly using these FDA-approved tests in a molecular-tumor-board context.
Collapse
Affiliation(s)
- Damien Vasseur
- Medical Biology and Pathology Department, Gustave Roussy, F-94805 Villejuif, France; (H.S.); (C.M.); (A.A.); (N.A.); (S.C.); (E.R.); (L.L.)
- AMMICa UAR3655/US23, Gustave Roussy, F-94805 Villejuif, France
- Correspondence:
| | - Hela Sassi
- Medical Biology and Pathology Department, Gustave Roussy, F-94805 Villejuif, France; (H.S.); (C.M.); (A.A.); (N.A.); (S.C.); (E.R.); (L.L.)
| | - Arnaud Bayle
- Drug Development Department (DITEP), Gustave Roussy, F-94805 Villejuif, France; (A.B.); (A.G.); (P.M.-R.); (M.N.-C.); (C.N.); (S.P.); (A.I.)
- Oncostat U1018, Inserm, Université Paris-Saclay, Équipe Labellisée Ligue Contre le Cancer, F-94805 Villejuif, France
| | - Marco Tagliamento
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (M.T.); (B.B.); (M.A.); (A.G.); (Y.L.); (A.H.); (O.C.)
| | - Benjamin Besse
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (M.T.); (B.B.); (M.A.); (A.G.); (Y.L.); (A.H.); (O.C.)
| | - Christophe Marzac
- Medical Biology and Pathology Department, Gustave Roussy, F-94805 Villejuif, France; (H.S.); (C.M.); (A.A.); (N.A.); (S.C.); (E.R.); (L.L.)
| | - Ahmadreza Arbab
- Medical Biology and Pathology Department, Gustave Roussy, F-94805 Villejuif, France; (H.S.); (C.M.); (A.A.); (N.A.); (S.C.); (E.R.); (L.L.)
| | - Nathalie Auger
- Medical Biology and Pathology Department, Gustave Roussy, F-94805 Villejuif, France; (H.S.); (C.M.); (A.A.); (N.A.); (S.C.); (E.R.); (L.L.)
| | - Sophie Cotteret
- Medical Biology and Pathology Department, Gustave Roussy, F-94805 Villejuif, France; (H.S.); (C.M.); (A.A.); (N.A.); (S.C.); (E.R.); (L.L.)
| | - Mihaela Aldea
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (M.T.); (B.B.); (M.A.); (A.G.); (Y.L.); (A.H.); (O.C.)
| | - Félix Blanc-Durand
- Gynecological Cancer Unit, Department of Medicine, Gustave Roussy, F-94805 Villejuif, France;
| | - Arthur Géraud
- Drug Development Department (DITEP), Gustave Roussy, F-94805 Villejuif, France; (A.B.); (A.G.); (P.M.-R.); (M.N.-C.); (C.N.); (S.P.); (A.I.)
| | - Anas Gazzah
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (M.T.); (B.B.); (M.A.); (A.G.); (Y.L.); (A.H.); (O.C.)
| | - Yohann Loriot
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (M.T.); (B.B.); (M.A.); (A.G.); (Y.L.); (A.H.); (O.C.)
| | - Antoine Hollebecque
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (M.T.); (B.B.); (M.A.); (A.G.); (Y.L.); (A.H.); (O.C.)
| | - Patricia Martín-Romano
- Drug Development Department (DITEP), Gustave Roussy, F-94805 Villejuif, France; (A.B.); (A.G.); (P.M.-R.); (M.N.-C.); (C.N.); (S.P.); (A.I.)
| | - Maud Ngo-Camus
- Drug Development Department (DITEP), Gustave Roussy, F-94805 Villejuif, France; (A.B.); (A.G.); (P.M.-R.); (M.N.-C.); (C.N.); (S.P.); (A.I.)
| | - Claudio Nicotra
- Drug Development Department (DITEP), Gustave Roussy, F-94805 Villejuif, France; (A.B.); (A.G.); (P.M.-R.); (M.N.-C.); (C.N.); (S.P.); (A.I.)
| | - Santiago Ponce
- Drug Development Department (DITEP), Gustave Roussy, F-94805 Villejuif, France; (A.B.); (A.G.); (P.M.-R.); (M.N.-C.); (C.N.); (S.P.); (A.I.)
| | - Madona Sakkal
- Dermatology Unit, Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (M.S.); (C.S.)
| | - Olivier Caron
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (M.T.); (B.B.); (M.A.); (A.G.); (Y.L.); (A.H.); (O.C.)
| | - Cristina Smolenschi
- Dermatology Unit, Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (M.S.); (C.S.)
| | | | - Antoine Italiano
- Drug Development Department (DITEP), Gustave Roussy, F-94805 Villejuif, France; (A.B.); (A.G.); (P.M.-R.); (M.N.-C.); (C.N.); (S.P.); (A.I.)
| | - Etienne Rouleau
- Medical Biology and Pathology Department, Gustave Roussy, F-94805 Villejuif, France; (H.S.); (C.M.); (A.A.); (N.A.); (S.C.); (E.R.); (L.L.)
- AMMICa UAR3655/US23, Gustave Roussy, F-94805 Villejuif, France
| | - Ludovic Lacroix
- Medical Biology and Pathology Department, Gustave Roussy, F-94805 Villejuif, France; (H.S.); (C.M.); (A.A.); (N.A.); (S.C.); (E.R.); (L.L.)
- AMMICa UAR3655/US23, Gustave Roussy, F-94805 Villejuif, France
| |
Collapse
|
74
|
Orr C, Wang C, Firat C, Connell LC, Sheehan MR, Vakiani E, Stadler ZK, Shia J. Primary Clonal Loss of Mismatch Repair Protein on Immunohistochemistry: A Pattern of Abnormality That Warrants Genetic Workup. JCO Precis Oncol 2022; 6:e2200111. [PMID: 35700411 DOI: 10.1200/po.22.00111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Christine Orr
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Chiyun Wang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Canan Firat
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Louise C Connell
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Margaret R Sheehan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Efsevia Vakiani
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
75
|
Microsatellite Instability Assessment by Immunohistochemistry in Acute Myeloid Leukemia: A Reappraisal and Review of the Literature. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:e386-e391. [PMID: 34980577 DOI: 10.1016/j.clml.2021.12.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Microsatellite instability (MSI) is caused by defects in DNA mismatch repair (MMR) components. Inactivation of any MMR gene(s), including hMLH1, hMSH2, hMSH6, and hPMS2, can result in MSI. Immunohistochemistry (IHC) is a sensitive and specific screening tool for MSI that can detect loss of expression of one or more MMR components. Of the four MMR markers, hMLH1 and hMSH2 are considered most informative of MSI status. There has been renewed interest in MSI status in view of its favorable association with response to immune checkpoint inhibitors in some cancers. MMR expression patterns in acute myeloid leukemia (AML) have not been evaluated systematically. METHODS We used clinically-validated IHC assays to assess the expression of hMLH1, hMSH2, hMSH6, and/or hPMS2 in formalin-fixed paraffin-embedded tissue sections of bone marrow core biopsies from patients diagnosed with AML. Mutation profiling was performed using next-generation sequencing to assess for mutations in MMR genes. RESULTS The study group included 236 patients with AML, including a cohort treated on a clinical trial of azacitidine and nivolumab (NCT02397720). In addition, hMSH6, and/or hPMS2 expression was assessed in 99 AML patients with diploid karyotype. All patients, except two, had retained expression of all MMR markers assessed: One patient from the azacytidine+nivolumab group had zonal patchy loss of staining of hMLH1 and, to a lesser extent, a similar staining pattern of hMSH2; and one patient from the AML with diploid karyotype group had loss of hMSH2 but retained expression of hMLH1, hMSH6 and hPMS2. In addition, a retrospective analysis on a separate cohort of 139 patients with primary AML, on which next generation sequencing profiling was performed, identified 14 cases with alterations in MMR genes. CONCLUSION AND REMARKS MMR loss is a rare event in AML, thus does not appear to underlie response patterns to anti-PD1 therapy.
Collapse
|
76
|
Cherri S, Oneda E, Noventa S, Melocchi L, Zaniboni A. Microsatellite instability and chemosensitivity in solid tumours. Ther Adv Med Oncol 2022; 14:17588359221099347. [PMID: 35620236 PMCID: PMC9127927 DOI: 10.1177/17588359221099347] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 04/21/2022] [Indexed: 01/01/2023] Open
Abstract
The use of biomarkers that influence a targeted choice in cancer treatments is the future of medical oncology. Within this scenario, in recent years, an important role has been played by knowledge of microsatellite instability (MSI), a molecular fingerprint that identifies defects in the mismatch repair system. This knowledge has changed clinical practice in the adjuvant setting of colon cancer, and its role in the neoadjuvant setting in gastric tumours is becoming increasingly interesting, as well as in endometrial cancers in both early and advanced diseases. Furthermore, it has undoubtedly conditioned the first lines of treatment in the metastatic setting in different types of cancers. The incidence of MSI is different in different cancer types, as well as in early cancers versus metastatic disease. Knowing the incidence of MSI in the various histologies can provide insight into the potential use of this biomarker considering its prognostic value, especially in the early stages, and its predictive role with respect to treatment response. In particular, MSI can guide the choice of chemotherapy treatments in the adjuvant setting of colon and perioperative setting in gastric tumours, which could lead to immunotherapy treatments in these patients in both the early stages of the disease and the metastatic setting where the response to immunotherapy drugs in diseases with MSI is now well established. In this review, we focus on colon, gastric and endometrial cancers, and we briefly discuss other cancer types where MSI could have a potential role in oncological treatment decisions.
Collapse
Affiliation(s)
- Sara Cherri
- Department of Clinical Oncology, Fondazione Poliambulanza, Via bissolati 57, 25124, Brescia, Italy
| | - Ester Oneda
- Department of Clinical Oncology, Fondazione Poliambulanza, Brescia, Italy
| | - Silvia Noventa
- Department of Clinical Oncology, Fondazione Poliambulanza, Brescia, Italy
| | - Laura Melocchi
- Department of Anatomical Pathology, Fondazione Poliambulanza, Brescia, Italy
| | - Alberto Zaniboni
- Department of Clinical Oncology, Fondazione Poliambulanza, Brescia, Italy
| |
Collapse
|
77
|
Nozawa Y, Ishida K, Maiko N, Takada-Owada A, Onozaki M, Takaoka M, Matsuyama K, Sakuraoka Y, Nakazato Y, Kubota K. Mixed adenoneuroendocrine carcinoma of the non-ampullary duodenum with mismatch repair deficiency: a rare case report. Med Mol Morphol 2022; 55:258-266. [PMID: 35596001 PMCID: PMC9374788 DOI: 10.1007/s00795-022-00324-x] [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: 03/17/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022]
Abstract
A non-ampullary duodenal mixed adenoneuroendocrine carcinoma (MANEC), consisting of a conventional adenocarcinoma and a neuroendocrine carcinoma (NEC), is exceedingly rare. Moreover, mismatch repair (MMR) deficient tumors have recently attracted attention. The patient, a 75-year-old woman with epigastric pain and nausea, was found to have a type 2 tumor of the duodenum, which was diagnosed on biopsy as a poorly differentiated carcinoma. A pancreaticoduodenectomy specimen showed a well-defined 50 × 48 mm tumor in the duodenal bulb, which was morphologically composed of glandular, sheet-like, and pleomorphic components. The glandular component was a tubular adenocarcinoma, showing a MUC5AC-positive gastric type. The sheet-like component consisted of homogenous tumor cells, with chromogranin A and synaptophysin diffusely positive, and a Ki-67 index of 72.8%. The pleomorphic component was diverse and prominent atypical tumor cells proliferated, focally positive for chromogranin A, diffusely positive for synaptophysin, and the Ki-67 index was 67.1%. The sheet-like and pleomorphic components were considered NEC, showing aberrant expression of p53, retinoblastoma, and p16. Notably, all three components were deficient in MLH1 and PMS2. We diagnosed a non-ampullary duodenal MANEC with MMR deficiency. This tumor has a unique morphology and immunohistochemical profile, and is valuable for clarifying the tumorigenesis mechanism of a non-ampullary duodenal MANEC.
Collapse
Affiliation(s)
- Yumi Nozawa
- Department of Diagnostic Pathology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Kazuyuki Ishida
- Department of Diagnostic Pathology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan. .,Department of Pathology, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan.
| | - Niki Maiko
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Atsuko Takada-Owada
- Department of Diagnostic Pathology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Masato Onozaki
- Department of Diagnostic Pathology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Mina Takaoka
- Department of Diagnostic Pathology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Kinichi Matsuyama
- Department of Pathology, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Yuhki Sakuraoka
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Yoshimasa Nakazato
- Department of Diagnostic Pathology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Keiichi Kubota
- Second Department of Surgery, Dokkyo Medical University, Mibu, Tochigi, Japan
| |
Collapse
|
78
|
Herz AL, Wisser S, Kohlruss M, Slotta-Huspenina J, Jesinghaus M, Grosser B, Steiger K, Novotny A, Hapfelmeier A, Schmidt T, Gaida MM, Weichert W, Keller G. Elevated microsatellite instability at selected tetranucleotide (EMAST) repeats in gastric cancer: a distinct microsatellite instability type with potential clinical impact? JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2022; 8:233-244. [PMID: 35099128 PMCID: PMC8977279 DOI: 10.1002/cjp2.257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 12/21/2022]
Abstract
We investigated the clinical impact of elevated microsatellite instability at selected tetranucleotide (EMAST) repeats in the context of neoadjuvant chemotherapy (CTx) in gastric/gastro‐oesophageal adenocarcinomas. We analysed 583 resected tumours (272 without and 311 after CTx) and 142 tumour biopsies before CTx. If at least two or three of the five tetranucleotide repeat markers tested showed instability, the tumours were defined as EMAST (2+) or EMAST (3+), respectively. Expression of mismatch repair proteins including MSH3 was analysed using immunohistochemistry. Microsatellite instability (MSI) and Epstein–Barr virus (EBV) positivity were determined using standard assays. EMAST (2+) and (3+) were detected in 17.8 and 11.5% of the tumours, respectively. The frequency of EMAST (2+) or (3+) in MSI‐high (MSI‐H) tumours was 96.2 or 92.5%, respectively, demonstrating a high overlap with this molecular subtype, and the association of EMAST and MSI status was significant (each overall p < 0.001). EMAST (2+ or 3+) alone in MSI‐H and EBV‐negative tumours demonstrated only a statistically significant association of EMAST (2+) positivity and negative lymph node status (42.3% in EMAST (2+) and 28.8% in EMAST negative, p = 0.045). EMAST alone by neither definition was significantly associated with overall survival (OS) of the patients. The median OS for EMAST (2+) patients was 40.0 months (95% confidence interval [CI] 16.4–63.6) compared with 38.7 months (95% CI 26.3–51.1) for the EMAST‐negative group (p = 0.880). The median OS for EMAST (3+) patients was 46.7 months (95% CI 18.2–75.2) and 38.7 months (95% CI 26.2–51.2) for the negative group (p = 0.879). No statistically significant association with response to neoadjuvant CTx was observed (p = 0.992 and p = 0.433 for EMAST (2+) and (3+), respectively). In conclusion, our results demonstrate a nearly complete intersection between MSI‐H and EMAST and they indicate that EMAST alone is not a distinct instability type associated with noticeable clinico‐pathological characteristics of gastric carcinoma patients.
Collapse
Affiliation(s)
- Anna-Lina Herz
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Sarah Wisser
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Meike Kohlruss
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Julia Slotta-Huspenina
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Moritz Jesinghaus
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany.,Institute of Pathology, University Hospital Marburg, Marburg, Germany
| | - Bianca Grosser
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany.,Institute of Pathology and Molecular Diagnostics, University Hospital Augsburg, Augsburg, Germany
| | - Katja Steiger
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany.,German Cancer Consortium [DKTK], Partner Site Munich, Institute of Pathology, Munich, Germany
| | - Alexander Novotny
- Department of Surgery, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Hapfelmeier
- Institute for AI and Informatics in Medicine, Technical University of Munich, Munich, Germany.,Institute of General Practice and Health Services Research, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas Schmidt
- Department of Surgery, University of Heidelberg, Heidelberg, Germany.,Department of Surgery, Universitätsklinikum Köln, Köln, Germany
| | - Matthias M Gaida
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Wilko Weichert
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany.,German Cancer Consortium [DKTK], Partner Site Munich, Institute of Pathology, Munich, Germany
| | - Gisela Keller
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
79
|
Lawrence L, Longacre T, Saleem A, Kunder C. Percent Agreement Between Immunohistochemistry and Next-Generation Sequencing in Testing Patients for Mismatch Repair Deficiency. Appl Immunohistochem Mol Morphol 2022; 30:345-349. [PMID: 35285457 DOI: 10.1097/pai.0000000000001018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 01/17/2022] [Indexed: 11/25/2022]
Abstract
The presence of mismatch repair deficiency is frequently assessed in gastrointestinal and gynecologic neoplasms by surgical pathologists using immunohistochemical methods. Targeted next-generation sequencing (NGS) covering some genes in the mismatch repair complex is used with increasing frequency, however, the percent positive and negative agreement of immunohistochemical methods and NGS of mismatch repair genes is not well-described in the literature. We sought to compare performance of immunohistochemistry (IHC) and NGS of mismatch repair genes on our institutional targeted panel. We evaluated the concordance of immunohistochemical and panel-based gene sequencing methods in a retrospective cohort study of patients evaluated at our center with both immunohistochemical and panel-based sequencing. Our NGS panel covers only MLH1 and MSH2, whereas our immunohistochemical panel assesses for expression of MLH1, PMS2, MSH2, and MSH6. We identified 68 unique patients with both immunohistochemical evaluation of mismatch repair protein expression and NGS panel sequencing, of which 67 were suitable for analysis given the patterns of immunohistochemical loss of expression observed. The percent positive agreement for NGS with IHC was 50%, albeit with very rare positive cases (n=2/4). Percent negative agreement was also high at 100% (n=63/63). One case with loss of MLH1, PMS2, and MSH6 expression by IHC and no pathogenic variants by NGS exhibited MLH1 promoter hypermethylation. Percent negative agreement between immunohistochemical and NGS gene sequencing is high, although firm conclusions regarding percent positive agreement between NGS and IHC are limited by low numbers of positive cases in our cohort. In general, we consider the findings to support continued use of immunohistochemical methods to screen for the presence of mismatch repair deficiency and consider additional testing by NGS likely to add little diagnostic value in the context of intact immunohistochemical expression of mismatch repair proteins.
Collapse
Affiliation(s)
- Lauren Lawrence
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | | | | | | |
Collapse
|
80
|
Zito Marino F, Amato M, Ronchi A, Panarese I, Ferraraccio F, De Vita F, Tirino G, Martinelli E, Troiani T, Facchini G, Pirozzi F, Perrotta M, Incoronato P, Addeo R, Selvaggi F, Lucido FS, Caraglia M, Savarese G, Sirica R, Casillo M, Lieto E, Auricchio A, Cardella F, Docimo L, Galizia G, Franco R. Microsatellite Status Detection in Gastrointestinal Cancers: PCR/NGS Is Mandatory in Negative/Patchy MMR Immunohistochemistry. Cancers (Basel) 2022; 14:cancers14092204. [PMID: 35565332 PMCID: PMC9102010 DOI: 10.3390/cancers14092204] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Microsatellite instability (MSI) detection has a high impact on eligibility for immune checkpoint inhibitors in gastrointestinal cancers. The appropriate detection of MSI represents the major critical issue in clinical practice, thus a better understanding of the limits related to MSI testing is needed to avoid misinterpretations. This study addresses the discordance between IHC and PCR/NGS testing in a large retrospective series of colorectal and gastric cancers in order to improve diagnosis. Our findings show a disagreement between negative/patchy expression IHC and PCR/NGS results, suggesting that molecular testing is mandatory in this subset of tumors. Abstract Background: Microsatellite instability (MSI) is a predictive biomarker for immune checkpoint inhibitors. The main goal was to investigate the discordance between IHC and PCR/NGS for MSI testing in gastrointestinal cancers. Methods: Two series were analyzed through IHC for mismatch-repair-system proteins (MMRP) and PCR, with one series of 444 colorectal cancers (CRC) and the other of 176 gastric cancers (GC). All cases with discordant results between IHC and PCR were analyzed by NGS. IHC staining was evaluated as follows: proficient MMR (pMMR), with all MMR positive; deficient MMR (dMMR), with the loss of one heterodimer; and cases with the loss/patchy expression of one MMR (lo-paMMR). Cases with instability in at least two markers by PCR were MSI-high (MSI-H) and with instability in one marker, MSI-low (MSI-L). Cases without instability were evaluated as microsatellite-stable (MSS). Results: In the CRC cohort, 15 out of 444 cases were dMMR and 46 lo-paMMR. Among the 15 dMMR, 13 were MSI-H and 2 MSS. Among the 46 lo-paMMR, 13 were MSI-H and 33 were MSS. In the GC cohort, 13 out of 176 cases were dMMR and 6 cases lo-paMMR. Among the 13 dMMR, 12 were MSI-H and only 1 was MSS. All six lo-paMMR cases were MSS. All NGS results were in agreement with PCR. Conclusions: In clinical practice, MMR–IHC could be used as a screening test and additional molecular analysis is mandatory exclusively in cases carrying loss/patchy MMR-IHC.
Collapse
Affiliation(s)
- Federica Zito Marino
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Martina Amato
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Andrea Ronchi
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Iacopo Panarese
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Franca Ferraraccio
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
| | - Ferdinando De Vita
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Giuseppe Tirino
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Erika Martinelli
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Teresa Troiani
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Gaetano Facchini
- Medical Oncology Unit, SM delle Grazie Hospital, 80078 Pozzuoli, Naples, Italy;
| | - Felice Pirozzi
- General Surgery Unit, Santa Maria delle Grazie Hospital, 80078 Pozzuoli, Naples, Italy;
| | - Michele Perrotta
- Hepatology and Interventional Ultrasound Unit, San Giovanni di Dio Hospital, 80027 Frattamaggiore, Naples, Italy;
| | - Pasquale Incoronato
- Medical Oncology Unit, ASL Napoli 2 Nord Hospital, 80014 Giugliano, Naples, Italy;
| | - Raffaele Addeo
- Medical Oncology Unit, San Giovanni di Dio Hospital, 80027 Frattamaggiore, Naples, Italy;
| | - Francesco Selvaggi
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (F.S.); (F.S.L.); (L.D.)
| | - Francesco Saverio Lucido
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (F.S.); (F.S.L.); (L.D.)
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.D.V.); (G.T.); (E.M.); (T.T.); (M.C.)
| | - Giovanni Savarese
- AMES, Centro Polidiagnostico Strumentale srl, 80013 Casalnuovo, Naples, Italy; (G.S.); (R.S.); (M.C.)
| | - Roberto Sirica
- AMES, Centro Polidiagnostico Strumentale srl, 80013 Casalnuovo, Naples, Italy; (G.S.); (R.S.); (M.C.)
| | - Marika Casillo
- AMES, Centro Polidiagnostico Strumentale srl, 80013 Casalnuovo, Naples, Italy; (G.S.); (R.S.); (M.C.)
| | - Eva Lieto
- Department of Translational Medical Science, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (E.L.); (A.A.); (F.C.)
| | - Annamaria Auricchio
- Department of Translational Medical Science, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (E.L.); (A.A.); (F.C.)
| | - Francesca Cardella
- Department of Translational Medical Science, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (E.L.); (A.A.); (F.C.)
| | - Ludovico Docimo
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy; (F.S.); (F.S.L.); (L.D.)
| | - Gennaro Galizia
- Department of Surgical Sciences, School of Medicine, Second University of Naples, Place Miraglia, 3th Building, West Side, 4th Floor, 80138 Naples, Italy;
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.Z.M.); (M.A.); (A.R.); (I.P.); (F.F.)
- Correspondence: ; Tel.: +39-0815664000
| |
Collapse
|
81
|
Curtius K, Gupta S, Boland CR. Review article: Lynch Syndrome-a mechanistic and clinical management update. Aliment Pharmacol Ther 2022; 55:960-977. [PMID: 35315099 DOI: 10.1111/apt.16826] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/02/2021] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Lynch syndrome (LS) is an autosomal dominant familial condition caused by a pathogenic variant (PV) in a DNA mismatch repair gene, which then predisposes carriers to various cancers. AIM To review the pathogenesis, clinical presentation, differential diagnosis and clinical strategies for detection and management of LS. METHODS A narrative review synthesising knowledge from published literature, as well as current National Comprehensive Cancer Network guidelines for management of LS was conducted. RESULTS LS tumours are characterised by unique pathogenesis, ultimately resulting in hypermutation, microsatellite instability and high immunogenicity that has significant implications for cancer risk, clinical presentation, treatment and surveillance. LS is one of the most common hereditary causes of cancer, and about 1 in 279 individuals carry a PV in an LS gene that predisposes to associated cancers. Individuals with LS have increased risks for colorectal, endometrial and other cancers, with significant variation in lifetime risk by LS-associated gene. CONCLUSIONS As genetic testing becomes more widespread, the number of individuals identified with LS is expected to increase in the population. Understanding the pathogenesis of LS informs current strategies for detection and clinical management, and also guides future areas for clinical innovation. Unravelling the mechanisms by which these tumours evolve may help to more precisely tailor management by the gene involved.
Collapse
Affiliation(s)
- Kit Curtius
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Samir Gupta
- Section of Gastroenterology, San Diego Veterans Affairs Healthcare System, San Diego, CA, USA.,Division of Gastroenterology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - C Richard Boland
- Division of Gastroenterology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
82
|
Universal Immunohistochemistry for Lynch Syndrome: A Systematic Review and Meta-analysis of 58,580 Colorectal Carcinomas. Clin Gastroenterol Hepatol 2022; 20:e496-e507. [PMID: 33887476 DOI: 10.1016/j.cgh.2021.04.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Lynch syndrome is a form of hereditary colorectal cancer (CRC) caused by pathogenic germline variants (PV) in DNA mismatch repair (MMR) genes. Currently, many Western countries perform universal immunohistochemistry testing on CRC to increase the identification of Lynch syndrome patients and their relatives. For a clear understanding of health benefits and costs, data on its outcomes are required: proportions of Lynch syndrome, sporadic MMR-deficient (MMRd) cases, and unexplained MMRd cases. METHODS Ovid Medline, Embase, and Cochrane CENTRAL were searched for studies reporting on universal MMR immunohistochemistry, followed by MMR germline analysis, until March 20, 2020. Proportions were calculated, subgroup analyses were performed based on age and diagnostics used, and random effects meta-analyses were conducted. Quality was assessed using the Joanna Briggs Critical Appraisal Tool for Prevalence Studies. RESULTS Of 2723 identified articles, 56 studies covering 58,580 CRCs were included. In 6.22% (95% CI, 5.08%-7.61%; I2 = 96%) MMRd was identified. MMR germline PV was present in 2.00% (95% CI, 1.59%-2.50%; I2 = 92%), ranging from 1.80% to 7.27% based on completeness of diagnostics and age restriction. Immunohistochemistry outcomes were missing in 11.81%, and germline testing was performed in 76.30% of eligible patients. In 7 studies, including 6848 CRCs completing all diagnostic stages, germline PV and biallelic somatic MMR inactivation were found in 3.01% and 1.75%, respectively; 0.61% remained unexplained MMRd. CONCLUSIONS Age, completeness, and type of diagnostics affect the percentage of MMR PV and unexplained MMRd percentages. Complete diagnostics explain almost all MMRd CRCs, reducing the amount of subsequent multigene panel testing. This contributes to optimizing testing and surveillance in MMRd CRC patients and relatives.
Collapse
|
83
|
Gatius S, Velasco A, Varela M, Cuatrecasas M, Jares P, Setaffy L, Bonhomme B, Santon A, Lindemann K, Croce S, Davidson B, Lax S, Palacios J, Matias-Guiu X. Comparison of the Idylla™ MSI assay with the Promega™ MSI Analysis System and immunohistochemistry on formalin-fixed paraffin-embedded tissue of endometrial carcinoma: results from an international, multicenter study. Virchows Arch 2022; 480:1031-1039. [DOI: 10.1007/s00428-022-03291-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/10/2022] [Accepted: 01/30/2022] [Indexed: 12/11/2022]
|
84
|
Kim H, Lim KY, Park JW, Kang J, Won JK, Lee K, Shim Y, Park CK, Kim SK, Choi SH, Kim TM, Yun H, Park SH. Sporadic and Lynch syndrome-associated mismatch repair-deficient brain tumors. J Transl Med 2022; 102:160-171. [PMID: 34848827 PMCID: PMC8784316 DOI: 10.1038/s41374-021-00694-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/09/2022] Open
Abstract
Mismatch repair-deficient (MMRD) brain tumors are rare among primary brain tumors and can be induced by germline or sporadic mutations. Here, we report 13 MMRD-associated (9 sporadic and 4 Lynch syndrome) primary brain tumors to determine clinicopathological and molecular characteristics and biological behavior. Our 13 MMRD brain tumors included glioblastoma (GBM) IDH-wildtype (n = 9) including 1 gliosarcoma, astrocytoma IDH-mutant WHO grade 4 (n = 2), diffuse midline glioma (DMG) H3 K27M-mutant (n = 1), and pleomorphic xanthoastrocytoma (PXA) (n = 1). Next-generation sequencing using a brain tumor-targeted gene panel, microsatellite instability (MSI) testing, Sanger sequencing for germline MMR gene mutation, immunohistochemistry of MMR proteins, and clinicopathological and survival analysis were performed. There were many accompanying mutations, suggesting a high tumor mutational burden (TMB) in 77%, but TMB was absent in one case of GBM, IDH-wildtype, DMG, and PXA, respectively. MSH2, MLH1, MSH6, and PMS2 mutations were found in 31%, 31%, 31% and 7% of patients, respectively. MSI-high and MSI-low were found in 50% and 8% of these gliomas, respectively and 34% was MSI-stable. All Lynch syndrome-associated GBMs had MSI-high. In addition, 77% (10/13) had histopathologically multinucleated giant cells. The progression-free survival tended to be poorer than the patients with no MMRD gliomas, but the number and follow-up duration of our patients were insufficient to get statistical significance. In the present study, we found that the most common MMRD primary brain tumor was GBM IDH-wildtype. The genetic profile of MMRD GBM was different from that of conventional GBM. MMRD gliomas with TMB and MSI-H may be sensitive to immunotherapy but resistant to temozolomide. Our findings can help develop better treatment options.
Collapse
Affiliation(s)
- Hyunhee Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ka Young Lim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin Woo Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeongwan Kang
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae Kyung Won
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kwanghoon Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yumi Shim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung-Ki Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung-Hong Choi
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hongseok Yun
- Department of Genomic Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Institute of Neuroscience, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
85
|
Scheiderer A, Riedinger C, Kimball K, Kilgore L, Orucevic A. Reporting Subclonal Immunohistochemical Staining of Mismatch Repair Proteins in Endometrial Carcinoma in the Times of Ever-Changing Guidelines. Arch Pathol Lab Med 2022; 146:1114-1121. [PMID: 34979565 DOI: 10.5858/arpa.2021-0201-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT.— The current College of American Pathologists reporting guideline for mismatch repair protein (MMRP) immunohistochemistry for Lynch syndrome (LS) screening considers the presence of any positive nuclear staining as intact MMRP expression. This would include tumors with combined areas of subclonal retention and loss of MMRP staining. OBJECTIVE.— To evaluate the clinical significance of reporting subclonal staining patterns of MMRP immunohistochemistry in endometrial carcinoma. DESIGN.— We retrospectively reviewed 455 consecutive MMRP immunohistochemistry results of endometrial carcinoma in hysterectomy specimens from 2012 through 2017 and identified cases with subclonal MMRP staining. These results were correlated with the patient's personal and family history of LS-associated carcinoma, MLH1 promoter methylation status, and LS genetic testing. RESULTS.— Subclonal staining of MMRP was seen in 48 of 455 cases (10.5%) on review. Thirty cases demonstrated isolated subclonal staining and were reported by pathologists as follows: subclonal (n = 5), complete MMRP loss (n = 4), and intact MMRP (n = 21). Eighteen cases had subclonal staining in combination with complete loss of other MMRP. Cases reported as subclonal or complete MMRP loss had appropriate clinical follow-up. Two of 2 cases with isolated subclonal MSH6 loss tested positive for LS. One of 3 cases with isolated subclonal MLH1/PMS2 loss was negative for MLH1 promoter methylation; LS genetic testing was not performed because of cost. CONCLUSIONS.— Our study reveals that LS germline mutation can be detected in endometrial carcinoma patients whose tumors display sole subclonal MMRP staining. Our results stress the importance of reporting subclonal staining patterns to ensure appropriate clinical follow-up.
Collapse
Affiliation(s)
- Ashley Scheiderer
- From the Department of Pathology (Scheiderer, Orucevic), University of Tennessee Medical Center, Knoxville
| | - Courtney Riedinger
- Department of Obstetrics and Gynecology (Riedinger, Kimball, Kilgore), University of Tennessee Medical Center, Knoxville
| | - Kristopher Kimball
- Department of Obstetrics and Gynecology (Riedinger, Kimball, Kilgore), University of Tennessee Medical Center, Knoxville
| | - Larry Kilgore
- Department of Obstetrics and Gynecology (Riedinger, Kimball, Kilgore), University of Tennessee Medical Center, Knoxville
| | - Amila Orucevic
- From the Department of Pathology (Scheiderer, Orucevic), University of Tennessee Medical Center, Knoxville
| |
Collapse
|
86
|
Kurpiel B, Thomas MS, Mubeen M, Ring KL, Modesitt SC, Moskaluk CA, Mills AM. MLH1/PMS2-deficient Endometrial Carcinomas in a Universally Screened Population: MLH1 Hypermethylation and Germline Mutation Status. Int J Gynecol Pathol 2022; 41:1-11. [PMID: 33577226 DOI: 10.1097/pgp.0000000000000767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
MLH1/PMS2 loss due to epigenetic hypermethylation of the MLH1 promoter is the most common cause of mismatch repair deficiency in endometrial carcinoma, and typically provides reassurance against an associated germline mutation. To further characterize the genetic features of MLH1/PMS2-deficient endometrial cancers, the departmental database was searched for cases with dual MLH1/PMS2 loss and retained MSH2/6 expression which underwent MLH1 hypermethylation testing. Genetic testing results were obtained when available. One hundred seventeen endometrial cancers met inclusion criteria: 100 (85%) were MLH1-hypermethylated, 3 (3%) were low-level/borderline, 7 (6%) were nonmethylated, and 7 (6%) were insufficient for testing. Sixteen cases (12 MLH1-hypermethylated, 3 nonmethylated, and 1 insufficient for testing) underwent germline testing, 6 of which (37.5%) demonstrated germline variants of unknown significance (VUS) (MSH6, PMS2, POLD1, BRIP1, RAD51D, CHEK2) but no known deleterious mutations. Notably, however, the patients harboring the MSH6 and PMS2 germline VUS had clinical features concerning for Lynch syndrome. One nonmethylated, germline-normal case underwent somatic tumor testing, and demonstrated a somatic MLH1 mutation. In summary, MLH1-hypermethylation accounts for the vast majority of MLH1/PMS2-deficient cancers in a universally screened population, although MLH1 somatic and germline mutations can occur. Occasionally, patients with MLH1-hypermethlated tumors also bear germline VUS in other mismatch repair genes as well as genes implicated in other hereditary cancer syndromes, but their clinical relevance is unclear. Family and personal cancer histories must always be evaluated to determine the need for germline testing in women with loss of MLH1/PMS2, even in the setting of hypermethylation.
Collapse
|
87
|
Jaffrelot M, Farés N, Brunac AC, Laurenty AP, Danjoux M, Grand D, Icher S, Meilleroux J, Mery E, Buscail E, Maulat C, Toulas C, Vande Perre P, Chipoulet E, Bonnet D, Staub A, Guimbaud R, Selves J. An unusual phenotype occurs in 15% of mismatch repair-deficient tumors and is associated with non-colorectal cancers and genetic syndromes. Mod Pathol 2022; 35:427-437. [PMID: 34545179 PMCID: PMC8860743 DOI: 10.1038/s41379-021-00918-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 08/21/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023]
Abstract
Immunohistochemistry (IHC) and/or MSI-PCR (microsatellite instability-polymerase chain reaction) tests are performed routinely to detect mismatch repair deficiency (MMR-D). Classical MMR-D tumors present a loss of MLH1/PMS2 or MSH2/MSH6 with MSI-High. Other profiles of MMR-D tumors have been described but have been rarely studied. In this study, we established a classification of unusual MMR-D tumors and determined their frequency and clinical impact. All MMR-D tumors identified between 2007 and 2017 were selected. Any profile besides the classical MMR-D phenotype was defined as unusual. For patients with unusual MMR-D tumors, IHC, and PCR data were reviewed, the tumor mutation burden (TMB) was evaluated and clinical and genetic features were collected. Of the 4948 cases of MMR testing, 3800 had both the available IHC and MSI-PCR results and 585 of these had MMR-D. After reviewing the IHC and PCR, 21% of the cases initially identified as unusual MMR-D were reclassified, which resulted in a final identification of 89 unusual MMR-D tumors (15%). Unusual MMR-D tumors were more often associated with non-CRC than classical MMR-D tumors. Unusual MMR-D tumors were classified into four sub-groups: i) isolated loss of PMS2 or MSH6, ii) classical loss of MLH1/PMS2 or MSH2/MSH6 without MSI, iii) four MMR proteins retained with MSI and, iv) complex loss of MMR proteins, with clinical characteristics for each sub-group. TMB-high or -intermediate was shown in 96% of the cancers studied (24/25), which confirmed MMR deficiency. Genetic syndromes were identified in 44.9% (40/89) and 21.4% (106/496) of patients with unusual and classical MMR-D tumors, respectively (P < 0.001). Five patients treated with an immune checkpoint inhibitor (ICI) had a prolonged clinical benefit. Our classification of unusual MMR-D phenotype helps to identify MMR deficiency. Unusual MMR-D phenotype occurs in 15% of MMR-D tumors. A high frequency of genetic syndromes was noted in these patients who could benefit from ICI.
Collapse
Affiliation(s)
- Marion Jaffrelot
- grid.411175.70000 0001 1457 2980Department of Digestive Oncology, Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Nadim Farés
- grid.411175.70000 0001 1457 2980Department of Digestive Oncology, Centre Hospitalier Universitaire (CHU), Toulouse, France ,grid.15781.3a0000 0001 0723 035XUniversité Fédérale Toulouse Midi-Pyrénées, Université Toulouse III Paul Sabatier, INSERM, CRCT, 31330 Toulouse, France ,grid.411175.70000 0001 1457 2980Department of Digestive Surgery, Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Anne Cécile Brunac
- grid.411175.70000 0001 1457 2980Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse; Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Anne Pascale Laurenty
- grid.411175.70000 0001 1457 2980Department of Digestive Oncology, Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Marie Danjoux
- grid.411175.70000 0001 1457 2980Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse; Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - David Grand
- grid.411175.70000 0001 1457 2980Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse; Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Samira Icher
- grid.411175.70000 0001 1457 2980Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse; Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Julie Meilleroux
- grid.411175.70000 0001 1457 2980Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse; Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Eliane Mery
- grid.411175.70000 0001 1457 2980Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse; Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Etienne Buscail
- grid.411175.70000 0001 1457 2980Department of Oncogenetics, Institut Universitaire du Cancer-Oncopole de Toulouse, Institut Claudius Regaud and Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Charlotte Maulat
- grid.411175.70000 0001 1457 2980Department of Oncogenetics, Institut Universitaire du Cancer-Oncopole de Toulouse, Institut Claudius Regaud and Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Christine Toulas
- grid.411175.70000 0001 1457 2980Department of Oncogenetics, Institut Universitaire du Cancer-Oncopole de Toulouse, Institut Claudius Regaud and Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Pierre Vande Perre
- grid.411175.70000 0001 1457 2980Department of Oncogenetics, Institut Universitaire du Cancer-Oncopole de Toulouse, Institut Claudius Regaud and Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Edith Chipoulet
- grid.411175.70000 0001 1457 2980Department of Oncogenetics, Institut Universitaire du Cancer-Oncopole de Toulouse, Institut Claudius Regaud and Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Delphine Bonnet
- grid.411175.70000 0001 1457 2980Department of Oncogenetics, Institut Universitaire du Cancer-Oncopole de Toulouse, Institut Claudius Regaud and Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Anne Staub
- grid.411175.70000 0001 1457 2980Department of Oncogenetics, Institut Universitaire du Cancer-Oncopole de Toulouse, Institut Claudius Regaud and Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Rosine Guimbaud
- grid.411175.70000 0001 1457 2980Department of Digestive Oncology, Centre Hospitalier Universitaire (CHU), Toulouse, France ,grid.15781.3a0000 0001 0723 035XUniversité Fédérale Toulouse Midi-Pyrénées, Université Toulouse III Paul Sabatier, INSERM, CRCT, 31330 Toulouse, France ,grid.411175.70000 0001 1457 2980Department of Digestive Surgery, Centre Hospitalier Universitaire (CHU), Toulouse, France
| | - Janick Selves
- Université Fédérale Toulouse Midi-Pyrénées, Université Toulouse III Paul Sabatier, INSERM, CRCT, 31330, Toulouse, France. .,Department of Digestive Surgery, Centre Hospitalier Universitaire (CHU), Toulouse, France.
| |
Collapse
|
88
|
Ijsselsteijn R, van Hees S, Drost M, Jansen JG, de Wind N. Induction of mismatch repair deficiency, compromised DNA damage signaling and compound hypermutagenesis by a dietary mutagen in a cell-based model for Lynch Syndrome. Carcinogenesis 2021; 43:160-169. [PMID: 34919656 PMCID: PMC8947211 DOI: 10.1093/carcin/bgab108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/21/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
The prevalent cancer predisposition Lynch syndrome (LS, OMIM #120435) is caused by an inherited heterozygous defect in any of the four core DNA mismatch repair (MMR) genes MSH2, MSH6, MLH1 or PMS2. MMR repairs errors by the replicative DNA polymerases in all proliferating tissues. Its deficiency, following somatic loss of the wild type copy, results in a spontaneous mutator phenotype that underlies the rapid development of, predominantly, colorectal cancer (CRC) in LS. Here we have addressed the hypothesis that aberrant responses of intestinal stem cells to diet-derived mutagens may be causally involved in the restricted cancer tropism of LS. To test this we have generated a panel of isogenic mouse embryonic stem (mES) cells with heterozygous or homozygous disruption of multiple MMR genes and investigated their responses to the common dietary mutagen and carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP). Our data reveal that PhIP can inactivate the wild type allele of heterozygous mES cells via the induction of either loss of heterozygosity (LOH) or intragenic mutations. Moreover, while protective DNA damage signaling (DDS) is compromised, PhIP induces more mutations in Msh2, Mlh1, Msh6 or Pms2-deficient mES cells than in wild type cells. Combined with their spontaneous mutator phenotypes, this results in a compound hypermutator phenotype. Together, these results indicate that dietary mutagens may promote CRC development in LS at multiple levels, providing a rationale for dietary modifications in the management of LS.
Collapse
Affiliation(s)
- Robbert Ijsselsteijn
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Sandrine van Hees
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Mark Drost
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jacob G Jansen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Niels de Wind
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
89
|
Olave MC, Graham RP. Mismatch repair deficiency: The what, how and why it is important. Genes Chromosomes Cancer 2021; 61:314-321. [PMID: 34837268 DOI: 10.1002/gcc.23015] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/15/2022] Open
Abstract
The mismatch repair system is a major pathway that functions in the maintenance of genomic integrity. It is involved in mitotic and meiotic recombination, apoptosis, immunoglobulin gene rearrangement, somatic hypermutation, and other processes. Deficiencies in mismatch repair give rise to hypermutability and the phenomenon called microsatellite instability. Detection of deficient mismatch repair function or microsatellite instability is used diagnostically, predictively, and prognostically. Specifically, deficient mismatch repair function is used for screening of Lynch syndrome, determining patients who are likely to respond to immune checkpoint inhibition, and to contributes to an understanding of which cancer patients may pursue a more aggressive clinical course. Microsatellite instability can be evaluated directly by polymerase chain reaction (PCR) or indirectly by assessment of mismatch repair protein expression using immunohistochemistry (IHC), and mismatch repair function using next-generation sequencing assays which evaluates homopolymer indels. In this article, we provide a concise practical review on mismatch repair deficiency (MMR-d)/microsatellite instability (MSI), focusing on clinical testing, different testing methods, interpretation of findings, the predictive, and prognostic utility of MSI.
Collapse
Affiliation(s)
- Maria C Olave
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Rondell P Graham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
90
|
Ando Y, Kumamoto K, Matsukawa H, Ishikawa R, Suto H, Oshima M, Kamada H, Morishita A, Kobara H, Matsunaga T, Haba R, Masaki T, Suzuki Y, Okano K. Low prevalence of biliary tract cancer with defective mismatch repair genes in a Japanese hospital-based population. Oncol Lett 2021; 23:4. [PMID: 34820003 PMCID: PMC8607234 DOI: 10.3892/ol.2021.13122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
Recent studies have reported that immune checkpoint inhibitors are effective against various defective mismatch repair (dMMR)/microsatellite instability-high (MSI-H) cancers. A limited number of reports are available on the frequency of dMMR/MSI-H carcinoma in biliary tract cancer (BTC), describing its clinicopathological characteristics and prognosis. The latter carcinoma is also associated with Lynch syndrome (LS). The present study was performed to investigate the frequency of patients with dMMR/MSI-H in BTC and the clinical characteristics of BTC with dMMR/MSI-H in a single institution in Japan. A total of 116 patients with BTC who underwent curative surgical resection at Kagawa University Hospital between January 2008 and December 2017 were included. The protein expression levels of the mismatch repair (MMR) genes [mutL homolog 1 (MLH1), mismatch repair endonuclease PMS2 (PMS2), MutS homolog (MSH)2 and MSH6] were assessed by immunohistochemistry (IHC) using formalin-fixed paraffin-embedded tissue specimens. Subsequently, MSI testing was performed on patients who exhibited loss of MMR protein expression. Loss of expression of one or more proteins was detected in five cases (4.3%). Loss of MLH1/PMS2 expression was observed in one case of intrahepatic cholangiocarcinoma, whereas loss of PMS2 expression was noted in one case of perihilar cholangiocarcinoma. Loss of MSH2/MSH6 and MSH6 expression was noted in two cases of distal cholangiocarcinoma and loss of PMS2 expression in one case of ampullary carcinoma. Out of the five patients, two demonstrated MSI-H. Microsatellite stability was observed in two cases and for one case, no data were available. Two MSI-H cases were patients with loss of expression of MLH1/PMS2 and MSH2/MSH6. None of the five patients exhibited a past medical history or family history of suspected LS. The frequency of dMMR in BTC was ~5%, which was similar to that reported by similar studies performed in other countries. In the present study, IHC appeared to be more useful than MSI testing for detecting MMR abnormalities with regards to the detection rate. Furthermore, there may only be a limited number of patients with BTCs who are likely to benefit from the therapeutic effects of treatment with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Yasuhisa Ando
- Department of Gastroenterological Surgery, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Kensuke Kumamoto
- Department of Gastroenterological Surgery, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Hiroyuki Matsukawa
- Department of Gastroenterological Surgery, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Ryou Ishikawa
- Department of Diagnostic Pathology, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Hironobu Suto
- Department of Gastroenterological Surgery, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Minoru Oshima
- Department of Gastroenterological Surgery, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Hideki Kamada
- Department of Gastroenterology and Neurology, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Toru Matsunaga
- Department of Diagnostic Pathology, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Reiji Haba
- Department of Diagnostic Pathology, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Yasuyuki Suzuki
- Department of Gastroenterological Surgery, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Keiichi Okano
- Department of Gastroenterological Surgery, Kagawa University, Miki, Kagawa 761-0793, Japan
| |
Collapse
|
91
|
Sorokin M, Rabushko E, Efimov V, Poddubskaya E, Sekacheva M, Simonov A, Nikitin D, Drobyshev A, Suntsova M, Buzdin A. Experimental and Meta-Analytic Validation of RNA Sequencing Signatures for Predicting Status of Microsatellite Instability. Front Mol Biosci 2021; 8:737821. [PMID: 34888350 PMCID: PMC8650122 DOI: 10.3389/fmolb.2021.737821] [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: 07/07/2021] [Accepted: 10/19/2021] [Indexed: 01/16/2023] Open
Abstract
Microsatellite instability (MSI) is an important diagnostic and prognostic cancer biomarker. In colorectal, cervical, ovarian, and gastric cancers, it can guide the prescription of chemotherapy and immunotherapy. In laboratory diagnostics of susceptible tumors, MSI is routinely detected by the size of marker polymerase chain reaction products encompassing frequent microsatellite expansion regions. Alternatively, MSI status is screened indirectly by immunohistochemical interrogation of microsatellite binding proteins. RNA sequencing (RNAseq) profiling is an emerging source of data for a wide spectrum of cancer biomarkers. Recently, three RNAseq-based gene signatures were deduced for establishing MSI status in tumor samples. They had 25, 15, and 14 gene products with only one common gene. However, they were developed and tested on the incomplete literature of The Cancer Genome Atlas (TCGA) sampling and never validated experimentally on independent RNAseq samples. In this study, we, for the first time, systematically validated these three RNAseq MSI signatures on the literature colorectal cancer (CRC) (n = 619), endometrial carcinoma (n = 533), gastric cancer (n = 380), uterine carcinosarcoma (n = 55), and esophageal cancer (n = 83) samples and on the set of experimental CRC RNAseq samples (n = 23) for tumors with known MSI status. We found that all three signatures performed well with area under the curve (AUC) ranges of 0.94-1 for the experimental CRCs and 0.94-1 for the TCGA CRC, esophageal cancer, and uterine carcinosarcoma samples. However, for the TCGA endometrial carcinoma and gastric cancer samples, only two signatures were effective with AUC 0.91-0.97, whereas the third signature showed a significantly lower AUC of 0.69-0.88. Software for calculating these MSI signatures using RNAseq data is included.
Collapse
Affiliation(s)
- Maksim Sorokin
- Laboratory For Clinical and Genomic Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- OmicsWay Corp., Walnut, CA, United States
| | - Elizaveta Rabushko
- Laboratory For Clinical and Genomic Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Victor Efimov
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
- Oncobox Ltd., Moscow, Russia
| | - Elena Poddubskaya
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Marina Sekacheva
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexander Simonov
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
- Oncobox Ltd., Moscow, Russia
| | - Daniil Nikitin
- Oncobox Ltd., Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Aleksey Drobyshev
- Laboratory For Clinical and Genomic Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maria Suntsova
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anton Buzdin
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- OmicsWay Corp., Walnut, CA, United States
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
92
|
Tonello M, Baratti D, Sammartino P, Di Giorgio A, Robella M, Sassaroli C, Framarini M, Valle M, Macrì A, Graziosi L, Coccolini F, Lippolis PV, Roberta G, Deraco M, Biacchi D, Santullo F, Vaira M, Di Lauro K, D'Acapito F, Carboni F, Giuffrè G, Donini A, Fugazzola P, Faviana P, Lorena S, Scapinello A, Del Bianco P, Sommariva A. Microsatellite and RAS/RAF Mutational Status as Prognostic Factors in Colorectal Peritoneal Metastases Treated with Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy (HIPEC). Ann Surg Oncol 2021; 29:3405-3417. [PMID: 34783946 DOI: 10.1245/s10434-021-11045-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/20/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) leads to prolonged survival for selected patients with colorectal (CRC) peritoneal metastases (PM). This study aimed to analyze the prognostic role of micro-satellite (MS) status and RAS/RAF mutations for patients treated with CRS. METHODS Data were collected from 13 Italian centers with PM expertise within a collaborative group of the Italian Society of Surgical Oncology. Clinical and pathologic variables and KRAS/NRAS/BRAF mutational and MS status were correlated with overall survival (OS) and disease-free survival (DFS). RESULTS The study enrolled 437 patients treated with CRS-HIPEC. The median OS was 42.3 months [95% confidence interval (CI), 33.4-51.2 months], and the median DFS was 13.6 months (95% CI, 12.3-14.9 months). The local (peritoneal) DFS was 20.5 months (95% CI, 16.4-24.6 months). In addition to the known clinical factors, KRAS mutations (p = 0.005), BRAF mutations (p = 0.01), and MS status (p = 0.04) were related to survival. The KRAS- and BRAF-mutated patients had a shorter survival than the wild-type (WT) patients (5-year OS, 29.4% and 26.8% vs 51.5%, respectively). The patients with micro-satellite instability (MSI) had a longer survival than the patients with micro-satellite stability (MSS) (5-year OS, 58.3% vs 36.7%). The MSI/WT patients had the best prognosis. The MSS/WT and MSI/mutated patients had similar survivals, whereas the MSS/mutated patients showed the worst prognosis (5-year OS, 70.6%, 48.1%, 23.4%; p = 0.0001). In the multivariable analysis, OS was related to the Peritoneal Cancer Index [hazard ratio (HR), 1.05 per point], completeness of cytoreduction (CC) score (HR, 2.8), N status (HR, 1.6), signet-ring (HR, 2.4), MSI/WT (HR, 0.5), and MSS/WT-MSI/mutation (HR, 0.4). Similar results were obtained for DFS. CONCLUSION For patients affected by CRC-PM who are eligible for CRS, clinical and pathologic criteria need to be integrated with molecular features (KRAS/BRAF mutation). Micro-satellite status should be strongly considered because MSI confers a survival advantage over MSS, even for mutated patients.
Collapse
Affiliation(s)
- Marco Tonello
- Unit of Surgical Oncology of the Esophagus and Digestive Tract, Surgical Oncology Department, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Dario Baratti
- Peritoneal Surface Malignancy Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo Sammartino
- Cytoreductive Surgery and HIPEC Unit, Department of Surgery "Pietro Valdoni", Sapienza University of Rome, Rome, Italy
| | - Andrea Di Giorgio
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Manuela Robella
- Surgical Oncology Unit, Candiolo Cancer Institute, Candiolo, Turin, Italy
| | - Cinzia Sassaroli
- Colorectal Surgical Oncology, Abdominal Oncology Department, Fondazione Giovanni Pascale" IRCCS, Naples, Italy
| | - Massimo Framarini
- General and Oncologic Surgery, Morgagni-Pierantoni Hospital, AUSL Romagna, Forlì, Italy
| | - Mario Valle
- Peritoneal Malignancies Unit, INT "Regina Elena", Rome, Italy
| | - Antonio Macrì
- Peritoneal Surface Malignancy and Soft Tissue Sarcoma Program, University of Messina, Messina, Italy
| | - Luigina Graziosi
- General and Emergency Surgery Department, Santa Maria della Misericordia Hospital, University of Perugia, Perugia, Italy
| | - Federico Coccolini
- General Emergency and Trauma Surgery, Bufalini Hospital, Cesena, Italy.,General Emergency and Trauma Surgery, Pisa University Hospital, Pisa, Italy
| | - Piero Vincenzo Lippolis
- General and Peritoneal Surgery, Department of Surgery, Hospital University Pisa (AOUP), Pisa, Italy
| | - Gelmini Roberta
- General and Oncological Surgery Unit, AOU of Modena University of Modena and Reggio Emilia, Modena, Italy
| | - Marcello Deraco
- Peritoneal Surface Malignancy Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Daniele Biacchi
- Cytoreductive Surgery and HIPEC Unit, Department of Surgery "Pietro Valdoni", Sapienza University of Rome, Rome, Italy
| | - Francesco Santullo
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marco Vaira
- Surgical Oncology Unit, Candiolo Cancer Institute, Candiolo, Turin, Italy
| | - Katia Di Lauro
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Fabrizio D'Acapito
- General and Oncologic Surgery, Morgagni-Pierantoni Hospital, AUSL Romagna, Forlì, Italy
| | - Fabio Carboni
- Peritoneal Malignancies Unit, INT "Regina Elena", Rome, Italy
| | - Giuseppe Giuffrè
- Department of Human Pathology in Adult and Developmental Age 'Gaetano Barresi', Section of Pathology, University of Messina, Messina, Italy
| | - Annibale Donini
- General and Emergency Surgery Department, Santa Maria della Misericordia Hospital, University of Perugia, Perugia, Italy
| | - Paola Fugazzola
- General Emergency and Trauma Surgery, Bufalini Hospital, Cesena, Italy
| | - Pinuccia Faviana
- Pathological Anatomy III, Laboratory Medicine Department, Hospital University Pisa (AOUP), Pisa, Italy
| | - Sorrentino Lorena
- General and Oncological Surgery Unit, AOU of Modena University of Modena and Reggio Emilia, Modena, Italy
| | | | - Paola Del Bianco
- Clinical Research Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Antonio Sommariva
- Unit of Surgical Oncology of the Esophagus and Digestive Tract, Surgical Oncology Department, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.
| |
Collapse
|
93
|
Buza N. Immunohistochemistry in gynecologic carcinomas: Practical update with diagnostic and clinical considerations based on the 2020 WHO classification of tumors. Semin Diagn Pathol 2021; 39:58-77. [PMID: 34750021 DOI: 10.1053/j.semdp.2021.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/28/2021] [Indexed: 02/06/2023]
Abstract
This review provides an update on immunohistochemistry applications-diagnostic, prognostic, and predictive-in the pathology evaluation of gynecologic carcinomas. The 5th edition of the WHO Classification of Female Genital Tumors introduced important changes in the diagnostic classification of lower genital tract, endometrial, and ovarian carcinomas, with major influence on the routine pathology practice. Lower genital tract carcinomas and their precursor lesions are now classified based on their human papillomavirus (HPV)-associated and HPV-independent pathogenesis, reflecting the clinically significant prognostic differences and impacting the therapeutic decision-making. Immunohistochemical markers have an increasing role in the pathology evaluation of endometrial carcinomas: in addition to their traditional use in the differential diagnosis and histologic subtyping, they have also been recently advocated for prognostic classification as surrogates for the TCGA (The Cancer Genome Atlas) molecular groups. New entities - mesonephric-like adenocarcinoma and gastric (gastrointestinal)-type mucinous adenocarcinoma of the endometrium - have also been added and often require immunostains for diagnostic confirmation. Ovarian carcinomas frequently show overlapping morphologic patterns and heterogeneous appearance within the same tumor, necessitating immunohistochemical work-up. Beyond diagnostic applications, there is increasing clinical demand for screening of inherited cancer syndromes, prediction of prognosis and guiding targeted therapy. Practical issues and pitfalls related to mismatch repair protein immunohistochemistry, HER2, and PD-L1 testing are also discussed.
Collapse
Affiliation(s)
- Natalia Buza
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
94
|
Escobar D, Bushara O, Sun L, Liao J, Yang GY. Clinicopathologic Characteristics of FBXW7-Mutated Colorectal Adenocarcinoma and Association with Aberrant Beta-catenin Localization. Hum Pathol 2021; 119:51-58. [PMID: 34717891 DOI: 10.1016/j.humpath.2021.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/06/2021] [Indexed: 11/04/2022]
Abstract
Oncogenic mutations in the adenomatous polyposis coli (APC)/Wnt signaling pathway are well documented. The FBXW7 gene (F-Box And WD Repeat Domain Containing 7) encodes a member of the ubiquitin-proteasome complex that is more recently described to antagonize the oncogenic Wnt pathway by promoting the degradation of β-catenin encoded by CTNNB1 gene. The pathologic significance of FBXW7 mutation in colorectal carcinoma (CRC) remains underreported. In this study, we report the clinicopathologic and β-catenin immunohistochemical features of a single-institution cohort (83 cases) of FBXW7-mutated CRC compared to CTNNB1-mutated CRC. FBXW7-mutated CRC was more common in older patients (p=0.031) and in the left/distal colon (p=0.022). Immunohistochemical analysis revealed that aberrant nuclear/cytoplasmic β-catenin localization was identified in a significantly high proportion of FBXW7-mutated CRCs. When compared to CTNNB1-mutated CRC, FBXW7-mutated CRC showed a significantly higher proportion of MSI-stable tumors with intact expression of DNA mismatch repair proteins, and had significantly more frequent co-occurrence of missense TP53 and KRAS mutations. The most frequently mutated FBXW7 residues/hotspots were located within the WD repeat domains (aa 378-659), which were also associated with aberrant nuclear/cytoplasmic localization of β-catenin protein. Our results indicate the unique pathologic characteristics of FBXW7 mutated CRC with frequent co-occurrence of missense mutant TP53, and KRAS. The mutated FBXW7 residues/hotspots and its association with aberrant nuclear/cytoplasmic β-catenin localization further support the oncogenic role of FBXW7 in colon carcinogenesis.
Collapse
Affiliation(s)
- David Escobar
- Department of Pathology, Northwestern University, Feinberg School of Medicine
| | - Omar Bushara
- Department of Pathology, Northwestern University, Feinberg School of Medicine
| | - Leyu Sun
- Department of Pathology, Northwestern University, Feinberg School of Medicine
| | - Jie Liao
- Department of Pathology, Northwestern University, Feinberg School of Medicine
| | - Guang-Yu Yang
- Department of Pathology, Northwestern University, Feinberg School of Medicine.
| |
Collapse
|
95
|
Olkinuora AP, Peltomäki PT, Aaltonen LA, Rajamäki K. From APC to the genetics of hereditary and familial colon cancer syndromes. Hum Mol Genet 2021; 30:R206-R224. [PMID: 34329396 PMCID: PMC8490010 DOI: 10.1093/hmg/ddab208] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 11/12/2022] Open
Abstract
Hereditary colorectal cancer (CRC) syndromes attributable to high penetrance mutations represent 9-26% of young-onset CRC cases. The clinical significance of many of these mutations is understood well enough to be used in diagnostics and as an aid in patient care. However, despite the advances made in the field, a significant proportion of familial and early-onset cases remains molecularly uncharacterized and extensive work is still needed to fully understand the genetic nature of CRC susceptibility. With the emergence of next-generation sequencing and associated methods, several predisposition loci have been unraveled, but validation is incomplete. Individuals with cancer-predisposing mutations are currently enrolled in life-long surveillance, but with the development of new treatments, such as cancer vaccinations, this might change in the not so distant future for at least some individuals. For individuals without a known cause for their disease susceptibility, prevention and therapy options are less precise. Herein, we review the progress achieved in the last three decades with a focus on how CRC predisposition genes were discovered. Furthermore, we discuss the clinical implications of these discoveries and anticipate what to expect in the next decade.
Collapse
Affiliation(s)
- Alisa P Olkinuora
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00014 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00014 Helsinki, Finland
| | - Päivi T Peltomäki
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00014 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00014 Helsinki, Finland
| | - Lauri A Aaltonen
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00014 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00014 Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, 00014 Helsinki, Finland
| | - Kristiina Rajamäki
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00014 Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
96
|
Laycock A, Kang A, Ang S, Texler M, Bentel J. Lack of correlation between MSH3 immunohistochemistry and microsatellite analysis for the detection of elevated microsatellite alterations at selected tetranucleotide repeats (EMAST) in colorectal cancers. Hum Pathol 2021; 118:9-17. [PMID: 34537247 DOI: 10.1016/j.humpath.2021.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/02/2021] [Accepted: 09/10/2021] [Indexed: 12/24/2022]
Abstract
Immunohistochemical evaluation of mismatch repair protein (MMR) expression is an important screening tool in diagnostic pathology, where it is routinely used to identify subsets of colorectal cancers (CRCs) with either inherited or sporadic forms of microsatellite instability (MSI). MSH3 is not included in current MMR panels, although aberrant MSH3 expression is reported to occur in 40-60% of CRCs and is associated with elevated microsatellite alterations at selected tetranucleotide repeats (EMAST) and a worse prognosis. In this study, we applied MSH3 immunohistochemistry and tetranucleotide MSI analysis to a cohort of 250 unselected CRCs to evaluate the potential use of the methods in routine practice. Partial, complete, and focal loss of nuclear MSH3 and its cytoplasmic mislocalization were evident in 67% of tumors, whereas MSI was evident in two to six of a panel of six tetranucleotide repeats in 46% of cases. However, concordance between MSH3 immunohistochemistry and tetranucleotide MSI results was only 61%, indicating the unsuitability of this combination of tests in routine pathology practice. MSH3 immunostaining was compromised in areas of tissue crush and autolysis, which are common in biopsy and surgical samples, potentially mitigating against its routine use. Although tetranucleotide MSI is clearly evident in a subset of CRCs, further development of validated sets of tetranucleotide repeats and either MSH3 or other immunohistochemical markers will be required to include EMAST testing in the routine evaluation of CRCs in clinical practice.
Collapse
Affiliation(s)
- Andrew Laycock
- PathWest Anatomical Pathology, Fiona Stanley Hospital, Perth, 6150 Western Australia, Australia; University of Notre Dame, Fremantle, 6160 Western Australia, Australia; Curtin University, Perth, 6102 Western Australia, Australia.
| | - Alexandra Kang
- PathWest Anatomical Pathology, Fiona Stanley Hospital, Perth, 6150 Western Australia, Australia
| | - Sophia Ang
- Clinical Services, Fiona Stanley Hospital, Perth, 6150 Western Australia, Australia
| | - Michael Texler
- PathWest Anatomical Pathology, Fiona Stanley Hospital, Perth, 6150 Western Australia, Australia
| | - Jacqueline Bentel
- PathWest Anatomical Pathology, Fiona Stanley Hospital, Perth, 6150 Western Australia, Australia
| |
Collapse
|
97
|
Sensitive detection of microsatellite instability in tissues and liquid biopsies: Recent developments and updates. Comput Struct Biotechnol J 2021; 19:4931-4940. [PMID: 34527197 PMCID: PMC8433064 DOI: 10.1016/j.csbj.2021.08.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 12/16/2022] Open
Abstract
Microsatellite instability (MSI), a phenotype displayed as deletions/insertions of repetitive genomic sequences, has drawn great attention due to its application in cancer including diagnosis, prognosis and immunotherapy response prediction. Several methods have been developed for the detection of MSI, facilitating the MSI classification of cancer patients. In view of recent interest in minimally-invasive detection of MSI via liquid biopsy samples, which requires methods with high sensitivity to identify small fractions of altered DNA in the presence of large amount of wild type copies, sensitive MSI detection approaches are emerging. Here we review the available MSI detection methods and their detection limits and focus on recently developed next-generation-sequencing based approaches and bioinformatics algorithms available for MSI analysis in various cancer types.
Collapse
|
98
|
Liu H, Ye Z, Yang T, Xie H, Duan T, Li M, Wu M, Song B. Predictive Value of Metabolic Parameters Derived From 18F-FDG PET/CT for Microsatellite Instability in Patients With Colorectal Carcinoma. Front Immunol 2021; 12:724464. [PMID: 34512653 PMCID: PMC8426433 DOI: 10.3389/fimmu.2021.724464] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/02/2021] [Indexed: 02/05/2023] Open
Abstract
Background Microsatellite instability (MSI) is one of the important factors that determine the effectiveness of immunotherapy in colorectal cancer (CRC) and serves as a prognostic biomarker for its clinical outcomes. Purpose To investigate whether the metabolic parameters derived from18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) can predict MSI status in patients with CRC. Materials and Methods A retrospective analysis was performed on CRC patients who underwent 18F-FDG PET/CT examination before surgery between January 2015 and April 2021. The metabolic 18F-FDG PET/CT parameters of the primary CRC lesion were calculated and recorded with different thresholds, including the maximum, peak, and mean standardized uptake value (SUVmax, SUVpeak, and SUVmean), as well as the metabolic tumor volume (MTV) and the total lesion glycolysis (TLG). The status of MSI was determined by immunohistochemical assessment. The difference of quantitative parameters between MSI and microsatellite stability (MSS) groups was assessed, and the receiver operating characteristic (ROC) analyses with area under ROC curves (AUC) was used to evaluate the predictive performance of metabolic parameters. Results A total of 44 patients (24 men and 20 women; mean ± standard deviation age: 71.1 ± 14.2 years) were included. There were 14 patients in the MSI group while there were 30 in the MSS group. MTV30%, MTV40%, MTV50%, and MTV60%, as well as TLG50% and TLG60% showed significant difference between two groups (all p-values <0.05), among which MTV50% demonstrated the highest performance in the prediction of MSI, with an AUC of 0.805 [95% confidence interval (CI): 0.657-0.909], a sensitivity of 92.9% (95% CI: 0.661-0.998), and a specificity of 66.7% (95% CI: 0.472-0.827). Patients' age and MTV50% were significant predictive indicators of MSI in multivariate logistic regression. Conclusion The metabolic parameters derived from18F-FDG PET/CT were able to preoperatively predict the MSI status in CRC, with MTV50% demonstrating the highest predictive performance. PET/CT imaging could serve as a noninvasive tool in the guidance of immunotherapy and individualized treatment in CRC patients.
Collapse
Affiliation(s)
- Hao Liu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Department of Nuclear Medicine, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China
| | - Zheng Ye
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongjun Xie
- Department of Nuclear Medicine, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China
| | - Ting Duan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Mou Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Min Wu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
99
|
Sugimoto R, Endo M, Osakabe M, Toya Y, Yanagawa N, Matsumoto T, Sugai T. Immunohistochemical Analysis of Mismatch Repair Gene Proteins in Early Gastric Cancer Based on Microsatellite Status. Digestion 2021; 102:691-700. [PMID: 33053554 DOI: 10.1159/000510679] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/05/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND Microsatellite instability (MSI) is a major pathway involved in gastric carcinogenesis and is observed in 10-20% of early gastric cancers (EGCs). Early detection of EGCs with an MSI-high phenotype would be useful for elucidating the mechanisms of gastric carcinogenesis and improving outcomes in patients with GC. OBJECTIVE We explored the usefulness of immunohistochemical expression of mismatch repair (MMR) proteins, including MLH1, PMS2, MSH2, and MSH6 in EGC. METHODS We examined the expression of 4 MMR proteins using immunohistochemistry in 119 patients with EGC based on MS status, as determined by polymerase chain reaction-microsatellite analysis. In addition, methylation of the MLH1 gene was quantified by pyrosequencing. RESULTS EGCs were classified into 46 MSI-high phenotypes and 73 microsatellite stable (MSS) phenotypes. Although loss of MLH1 expression was associated with loss of PMS2 expression in the MSI-high phenotype, discordant cases of loss of expression between MLH1 and PMS2 were found (MLH1 [-]/PMS2 [+], 3 cases). Loss of MLH1/PMS2 expression was observed in 2 of 73 MSS phenotypes. Loss of MSH2/MSH6 expression was found in 4 of 46 MSI-high phenotypes, whereas loss of MSH2/MSH6 expression was not detected in the MSS phenotype. In addition, loss of MLH1 expression was correlated with methylation of MLH1. However, there were discordant cases in which loss of MLH1 expression was not accompanied by methylation of MLH1. CONCLUSION Although immunostaining of MMR proteins could help predict MSI in EGCs, immunostaining did not have the same value as genetic testing for determination of MSI.
Collapse
Affiliation(s)
- Ryo Sugimoto
- Department of Molecular Diagnostic Pathology, School of Medicine, Iwate Medical University, Shiwa, Japan
| | - Masaki Endo
- Department of Internal Medicine, Division of Gastroenterology, School of Medicine, Iwate Medical University, Shiwa, Japan
| | - Mitsumasa Osakabe
- Department of Molecular Diagnostic Pathology, School of Medicine, Iwate Medical University, Shiwa, Japan
| | - Yosuke Toya
- Department of Internal Medicine, Division of Gastroenterology, School of Medicine, Iwate Medical University, Shiwa, Japan
| | - Naoki Yanagawa
- Department of Molecular Diagnostic Pathology, School of Medicine, Iwate Medical University, Shiwa, Japan
| | - Takayuki Matsumoto
- Department of Internal Medicine, Division of Gastroenterology, School of Medicine, Iwate Medical University, Shiwa, Japan
| | - Tamotsu Sugai
- Department of Molecular Diagnostic Pathology, School of Medicine, Iwate Medical University, Shiwa, Japan,
| |
Collapse
|
100
|
Han X, Zhang S, Zhou DC, Wang D, He X, Yuan D, Li R, He J, Duan X, Wendl MC, Ding L, Niu B. MSIsensor-ct: microsatellite instability detection using cfDNA sequencing data. Brief Bioinform 2021; 22:bbaa402. [PMID: 33461213 PMCID: PMC8424396 DOI: 10.1093/bib/bbaa402] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
MOTIVATION Microsatellite instability (MSI) is a promising biomarker for cancer prognosis and chemosensitivity. Techniques are rapidly evolving for the detection of MSI from tumor-normal paired or tumor-only sequencing data. However, tumor tissues are often insufficient, unavailable, or otherwise difficult to procure. Increasing clinical evidence indicates the enormous potential of plasma circulating cell-free DNA (cfNDA) technology as a noninvasive MSI detection approach. RESULTS We developed MSIsensor-ct, a bioinformatics tool based on a machine learning protocol, dedicated to detecting MSI status using cfDNA sequencing data with a potential stable MSIscore threshold of 20%. Evaluation of MSIsensor-ct on independent testing datasets with various levels of circulating tumor DNA (ctDNA) and sequencing depth showed 100% accuracy within the limit of detection (LOD) of 0.05% ctDNA content. MSIsensor-ct requires only BAM files as input, rendering it user-friendly and readily integrated into next generation sequencing (NGS) analysis pipelines. AVAILABILITY MSIsensor-ct is freely available at https://github.com/niu-lab/MSIsensor-ct. SUPPLEMENTARY INFORMATION Supplementary data are available at Briefings in Bioinformatics online.
Collapse
Affiliation(s)
- Xinyin Han
- Computer Network Information Center, Chinese Academy of Sciences, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Shuying Zhang
- Computer Network Information Center, Chinese Academy of Sciences, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Daniel Cui Zhou
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Dongliang Wang
- ChosenMed Technology (Beijing) Co., Ltd., Beijing 100176, China
| | - Xiaoyu He
- Computer Network Information Center, Chinese Academy of Sciences, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Danyang Yuan
- Computer Network Information Center, Chinese Academy of Sciences, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100190, China
| | - Ruilin Li
- Computer Network Information Center, Chinese Academy of Sciences, Beijing 100190, China
| | - Jiayin He
- Computer Network Information Center, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaohong Duan
- ChosenMed Technology (Beijing) Co., Ltd., Beijing 100176, China
| | - Michael C Wendl
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
- Department of Genetics, Washington University in St. Louis, St. Louis, MO 63108, USA
- Department of Mathematics, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Li Ding
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
- Department of Genetics, Washington University in St. Louis, St. Louis, MO 63108, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63108, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Beifang Niu
- Computer Network Information Center, Chinese Academy of Sciences, Beijing 100190, China
- University of the Chinese Academy of Sciences, Beijing 100190, China
- ChosenMed Technology (Beijing) Co., Ltd., Beijing 100176, China
| |
Collapse
|