1
|
Lin MT, Christenson ES, Pallavajjala A, Eshleman JR. Highly sensitive and specific markers for detection of mismatch repair deficiency by next-generation sequencing. Am J Clin Pathol 2025:aqaf026. [PMID: 40318191 DOI: 10.1093/ajcp/aqaf026] [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: 11/23/2024] [Accepted: 03/16/2025] [Indexed: 05/07/2025] Open
Abstract
OBJECTIVE To identify exonic markers that could improve analytic performance characteristics of next-generation sequencing (NGS) in detecting mismatch repair deficiency (dMMR) using colorectal cancer (CRC) as a model. METHODS Coding sequences of a target NGS panel (~1.13 megabase) were compared between dMMR CRC and mismatch repair-proficient (pMMR) CRC in a training cohort (41 dMMR CRCs and 213 pMMR CRCs) and a validation cohort (33 dMMR CRCs and 307 pMMR CRCs) with documented mismatch repair status by immunohistochemical and/or microsatellite instability assays. RESULTS The dMMR CRC cases showed significantly higher insertion/deletion (indel) mutations within exonic homopolymers (homo-indels), occurring predominantly within longer repeats of 5 to 10 nucleotides (92%, P < .0001), rather than shorter repeats of 2 to 4 nucleotides seen in pMMR CRC (62%). Homo-indels in dMMR CRC were not random. Hotspot loci were consistent between the training and validation cohorts. The dMMR defined by indels within homopolymers of 5 or more nucleotides, homopolymers of 7 or more nucleotides, or a panel of hotspots all showed 100% sensitivity and specificity with a range of cutoffs. CONCLUSIONS We propose that this approach allows one to identify highly sensitive and specific markers for detecting dMMR CRC by NGS alone. Further studies are warranted to test whether these markers are applicable to non-CRC neoplasms.
Collapse
Affiliation(s)
- Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Eric S Christenson
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Aparna Pallavajjala
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - James R Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| |
Collapse
|
2
|
Nicka CM, Green DC, Tsongalis GJ, Tafe LJ. Detection of Microsatellite Instability in Endometrial Carcinoma Using a Novel Homopolymer Assay. Int J Surg Pathol 2025:10668969241311500. [PMID: 40033864 DOI: 10.1177/10668969241311500] [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: 03/05/2025]
Abstract
Approximately 30% of endometrial cancers are associated with microsatellite instability (MSI) caused by deficiencies in the DNA mismatch repair (MMR) genes (dMMR). MMR testing by immunohistochemistry for MMR proteins and MSI testing by polymerase chain reaction (PCR) are routinely utilized to screen patients with colorectal cancer and endometrial cancer for Lynch syndrome and, more recently, to identify patients eligible for immunotherapy. The Biocartis Idylla™ MSI assay is a fully automated, cartridge-based real-time PCR assay. The assay uses as little as one formalin-fixed paraffin-embedded (FFPE) tumor section and is designed to detect seven novel MSI biomarkers consisting of short homopolymers located in ACVR2A, BTBD7, DIDO1, MRE11, RYR3, SEC31A and SULF2 genes. Mutation in two of these markers is considered MSI-H. FFPE of 35 ECs (25 dMMR and 10 microsatellite stable (MSS)) were used in this study. When tumor content was ≤20% on a slide, macrodissection was performed. The overall percent agreement with MMR IHC was 97% (31/32) with sensitivity = 96% and specificity = 100%. Pre-analytic evaluation of the manufacturer's recommended 20% tumor content cut-off is essential to ensure valid results. The Idylla MSI assay offers several advantages over other PCR-based assays including minimal hands-on time, rapid turn-around-time, no requirement for a paired normal sample and the use of FFPE directly without an extraction step.
Collapse
Affiliation(s)
- Catherine M Nicka
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Donald C Green
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Gregory J Tsongalis
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
- The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Laura J Tafe
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
- The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| |
Collapse
|
3
|
Mendiola M, Heredia-Soto V, Ruz-Caracuel I, Baillo A, Ramon-Patino JL, Berjon A, Escudero FJ, Pelaez-Garcia A, Hernandez A, Feliu J, Hardisson D, Redondo A. Performance of the Idylla microsatellite instability test in endometrial cancer. Mol Cell Probes 2024; 77:101976. [PMID: 39069012 DOI: 10.1016/j.mcp.2024.101976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
CONTEXT DNA mismatch repair (MMR) deficiency (dMMR) testing is now recommended in endometrial cancer. Defect identification in the molecules participating in this pathway, or the presence of microsatellite instability, are commonly employed for this purpose. Novel methods are continuously evolving to report dMMR/microsatellite instability and to easily perform routine diagnoses. OBJECTIVE The main aim of this study was to compare the concordance of the Idylla microsatellite instability test for the identification of dMMR endometrial cancer samples defined by immunohistochemistry and MMR genomic status. DESIGN We applied the Idylla MSI test to 126 early-stage endometrial cancer cases with MMR testing by immunohistochemistry and genomic characterization (methylation in MLH1 and sequence alterations in MLH1, PMS2, MSH2 and MSH6). Individual markers and overall specific performance indicators were explored. RESULTS The Idylla platform achieved a higher global concordance rate with MMR genomic status than with immunohistochemistry (75 % and 66 %, respectively). Sensitivity and specificity are also higher (75 % vs 66 % and 96 % vs 90 %, respectively). Clustering analysis split the patients into 2 well-differentiated clusters, the pMMR and the dMMR group, represented by MLH1/PMS2 loss and the MLH1 methylated promoter. Overall, immunohistochemistry and MMR genomic status identified more dMMR cases than did the Idylla test, although correlations were improved with a modified Idylla test cut-off. CONCLUSIONS Performance of the Idylla test was better correlated with MMR genomic status than MMR immunohistochemistry status, which improved with a modified test cut-off. Further studies are needed to confirm the cut-off accuracy.
Collapse
Affiliation(s)
- Marta Mendiola
- Molecular Pathology and Therapeutic Targets Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046, Madrid, Spain; Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Victoria Heredia-Soto
- Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain; Translational Oncology Research Laboratory, Hospital La Paz Institute for Health Research (IdiPAZ), 28046, Madrid, Spain
| | - Ignacio Ruz-Caracuel
- Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain; Department of Pathology, La Paz University Hospital, 28046, Madrid, Spain
| | - Amparo Baillo
- Mathematics Department, Autonomous University of Madrid, 28049, Madrid, Spain
| | | | - Alberto Berjon
- Molecular Pathology and Therapeutic Targets Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046, Madrid, Spain; Department of Pathology, La Paz University Hospital, 28046, Madrid, Spain
| | - Francisco Javier Escudero
- Translational Oncology Research Laboratory, Hospital La Paz Institute for Health Research (IdiPAZ), 28046, Madrid, Spain
| | - Alberto Pelaez-Garcia
- Molecular Pathology and Therapeutic Targets Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046, Madrid, Spain
| | - Alicia Hernandez
- Department of Obstetrics and Gynecology, La Paz University Hospital, 28046, Madrid, Spain; School of Medicine, Autonomous University of Madrid, 28046, Madrid, Spain
| | - Jaime Feliu
- Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain; Translational Oncology Research Laboratory, Hospital La Paz Institute for Health Research (IdiPAZ), 28046, Madrid, Spain; Department of Medical Oncology, La Paz University Hospital, 28046, Madrid, Spain; School of Medicine, Autonomous University of Madrid, 28046, Madrid, Spain; Cátedra UAM-ANGEM, Faculty of Medicine, Autonomous University of Madrid, 28046, Madrid, Spain
| | - David Hardisson
- Molecular Pathology and Therapeutic Targets Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046, Madrid, Spain; Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28029, Madrid, Spain; Department of Pathology, La Paz University Hospital, 28046, Madrid, Spain; School of Medicine, Autonomous University of Madrid, 28046, Madrid, Spain
| | - Andres Redondo
- Translational Oncology Research Laboratory, Hospital La Paz Institute for Health Research (IdiPAZ), 28046, Madrid, Spain; Department of Medical Oncology, La Paz University Hospital, 28046, Madrid, Spain; School of Medicine, Autonomous University of Madrid, 28046, Madrid, Spain; Cátedra UAM-ANGEM, Faculty of Medicine, Autonomous University of Madrid, 28046, Madrid, Spain.
| |
Collapse
|
4
|
Wang J, Zhang Z, Liu H, Liu N, Hu Y, Guo W, Li X. Identification of 8 candidate microsatellite instability loci in colorectal cancer and validation of the ACVR2A mechanism in the tumor progression. Sci Rep 2024; 14:14145. [PMID: 38898042 PMCID: PMC11187151 DOI: 10.1038/s41598-024-62753-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
This study probes the utility of biomarkers for microsatellite instability (MSI) detection and elucidates the molecular dynamics propelling colorectal cancer (CRC) progression. We synthesized a primer panel targeting 725 MSI loci, informed by The Cancer Genome Atlas (TCGA) and ancillary databases, to construct an amplicon library for next-generation sequencing (NGS). K-means clustering facilitated the distillation of 8 prime MSI loci, including activin A receptor type 2A (ACVR2A). Subsequently, we explored ACVR2A's influence on CRC advancement through in vivo tumor experiments and hematoxylin-eosin (HE) staining. Transwell assays gauged ACVR2A's role in CRC cell migration and invasion, while colony formation assays appraised cell proliferation. Western blotting illuminated the impact of ACVR2A suppression on CRC's PI3K/AKT/mTOR pathway protein expressions under hypoxia. Additionally, ACVR2A's influence on CRC-induced angiogenesis was quantified via angiogenesis assays. K-means clustering of NGS data pinpointed 32 MSI loci specific to tumor and DNA mismatch repair deficiency (dMMR) tissues. ACVR2A emerged as a pivotal biomarker, discerning MSI-H tissues with 90.97% sensitivity. A curated 8-loci set demonstrated 100% sensitivity and specificity for MSI-H detection in CRC. In vitro analyses corroborated ACVR2A's critical role, revealing its suppression of CRC proliferation, migration, and invasion. Moreover, ACVR2A inhibition under CRC-induced hypoxia markedly escalated MMP3, CyclinA, CyclinD1, and HIF1α protein expressions, alongside angiogenesis, by triggering the PI3K/AKT/mTOR cascade. The 8-loci ensemble stands as the optimal marker for MSI-H identification in CRC. ACVR2A, a central element within this group, deters CRC progression, while its suppression amplifies PI3K/AKT/mTOR signaling and angiogenesis under hypoxic stress.
Collapse
Affiliation(s)
- Jingyu Wang
- Molecular Oncology R&D Department, Guangzhou Wondfo Biotechnology Co.,LTD., Guangzhou, China
| | - Zhe Zhang
- Molecular Oncology R&D Department, Guangzhou Wondfo Biotechnology Co.,LTD., Guangzhou, China
| | - Hui Liu
- Molecular Oncology R&D Department, Guangzhou Wondfo Biotechnology Co.,LTD., Guangzhou, China
| | - Nian Liu
- Molecular Oncology R&D Department, Guangzhou Wondfo Biotechnology Co.,LTD., Guangzhou, China
| | - Yucheng Hu
- Molecular Oncology R&D Department, Guangzhou Wondfo Biotechnology Co.,LTD., Guangzhou, China
| | - Wenjuan Guo
- Molecular Oncology R&D Department, Guangzhou Wondfo Biotechnology Co.,LTD., Guangzhou, China.
| | - Xiangzhao Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Nádorvári ML, Lotz G, Kulka J, Kiss A, Tímár J. Microsatellite instability and mismatch repair protein deficiency: equal predictive markers? Pathol Oncol Res 2024; 30:1611719. [PMID: 38655493 PMCID: PMC11036414 DOI: 10.3389/pore.2024.1611719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024]
Abstract
Current clinical guidelines recommend mismatch repair (MMR) protein immunohistochemistry (IHC) or molecular microsatellite instability (MSI) tests as predictive markers of immunotherapies. Most of the pathological guidelines consider MMR protein IHC as the gold standard test to identify cancers with MMR deficiency and recommend molecular MSI tests only in special circumstances or to screen for Lynch syndrome. However, there are data in the literature which suggest that the two test types may not be equal. For example, molecular epidemiology studies reported different rates of deficient MMR (dMMR) and MSI in various cancer types. Additionally, direct comparisons of the two tests revealed relatively frequent discrepancies between MMR IHC and MSI tests, especially in non-colorectal and non-endometrial cancers and in cases with unusual dMMR phenotypes. There are also scattered clinical data showing that the efficacy of immune checkpoint inhibitors is different if the patient selection was based on dMMR versus MSI status of the cancers. All these observations question the current dogma that dMMR phenotype and genetic MSI status are equal predictive markers of the immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
6
|
Bae H, Rha SE, Kim H, Kang J, Shin YR. Predictive Value of Magnetic Resonance Imaging in Risk Stratification and Molecular Classification of Endometrial Cancer. Cancers (Basel) 2024; 16:921. [PMID: 38473283 DOI: 10.3390/cancers16050921] [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: 12/01/2023] [Revised: 01/27/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
This study evaluated the magnetic resonance imaging (MRI) findings of endometrial cancer (EC) patients and identified differences based on risk group and molecular classification. The study involved a total of 175 EC patients. The MRI data were retrospectively reviewed and compared based on the risk of recurrence. Additionally, the associations between imaging phenotypes and genomic signatures were assessed. The low-risk and non-low-risk groups (intermediate, high-intermediate, high, metastatic) showed significant differences in tumor diameter (p < 0.001), signal intensity and heterogeneity on diffusion-weighted imaging (DWI) (p = 0.003), deep myometrial invasion (involvement of more than 50% of the myometrium), cervical invasion (p < 0.001), extrauterine extension (p = 0.002), and lymphadenopathy (p = 0.003). Greater diffusion restriction and more heterogeneity on DWI were exhibited in the non-low-risk group than in the low-risk group. Deep myometrial invasion, cervical invasion, extrauterine extension, lymphadenopathy, recurrence, and stage discrepancy were more common in the non-low-risk group (p < 0.001). A significant difference in microsatellite stability status was observed in the heterogeneity of the contrast-enhanced T1-weighted images (p = 0.027). However, no significant differences were found in MRI parameters related to TP53 mutation. MRI features can be valuable predictors for differentiating risk groups in patients with EC. However, further investigations are needed to explore the imaging markers based on molecular classification.
Collapse
Affiliation(s)
- Hanna Bae
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 14662, Republic of Korea
| | - Sung Eun Rha
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 14662, Republic of Korea
| | - Hokun Kim
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 14662, Republic of Korea
| | - Jun Kang
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 14662, Republic of Korea
| | - Yu Ri Shin
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 14662, Republic of Korea
| |
Collapse
|
7
|
Nádorvári ML, Kenessey I, Kiss A, Barbai T, Kulka J, Rásó E, Tímár J. Comparison of standard mismatch repair deficiency and microsatellite instability tests in a large cancer series. J Transl Med 2024; 22:150. [PMID: 38350968 PMCID: PMC10863158 DOI: 10.1186/s12967-024-04960-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/07/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND The tumor-agnostic indication of immune checkpoint inhibitors to treat cancers with mismatch repair deficiency (dMMR)/microsatellite instability (MSI) increased the demand for such tests beyond Lynch syndrome. International guideline recommendations accept immunohistochemistry (IHC) for dMMR or molecular techniques (PCR or NGS) for MSI status determinations considering the two tests are equal, although there are scattered reports contradicting to this presumption. MATERIALS AND METHODS Here we have directly compared four protein MMR immunohistochemistry (IHC) to MSI Pentaplex PCR test in a large cancer patient cohort (n = 1306) of our diagnostic center where the two tests have been run parallel in 703 cases. RESULTS In this study we have found a high discrepancy rate (19.3%) of the two tests which was independent of the tumor types. The MSI PCR sensitivity for MMR IHC status was found to be very low resulting in a relatively low positive and negative predicting values. As a consequence, the correlation of the two tests was low (kappa < 0.7). During analysis of the possible contributing factors of this poor performance, we have excluded low tumor percentage of the samples, but identified dMMR phenotypes (classic versus non-classic or unusual) as possible contributors. CONCLUSION Although our cohort did not include samples with identified technical errors, our data strongly support previous reports that unidentified preanalytical factors might have the major influence on the poor performance of the MSI PCR and MMR IHC. Furthermore, the case is open whether the two test types are equally powerful predictive markers of immunotherapies.
Collapse
Affiliation(s)
- Maja L Nádorvári
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - István Kenessey
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - András Kiss
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Tamás Barbai
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Janina Kulka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Erzsébet Rásó
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
8
|
Bacher JW, Udho EB, Strauss EE, Vyazunova I, Gallinger S, Buchanan DD, Pai RK, Templeton AS, Storts DR, Eshleman JR, Halberg RB. A Highly Sensitive Pan-Cancer Test for Microsatellite Instability. J Mol Diagn 2023; 25:806-826. [PMID: 37544360 PMCID: PMC10629437 DOI: 10.1016/j.jmoldx.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 08/08/2023] Open
Abstract
Microsatellite instability (MSI) is an evolving biomarker for cancer detection and treatment. MSI was first used to identify patients with Lynch syndrome, a hereditary form of colorectal cancer (CRC), but has recently become indispensable in predicting patient response to immunotherapy. To address the need for pan-cancer MSI detection, a new multiplex assay was developed that uses novel long mononucleotide repeat (LMR) markers to improve sensitivity. A total of 469 tumor samples from 20 different cancer types, including 319 from patients with Lynch syndrome, were tested for MSI using the new LMR MSI Analysis System. Results were validated by using deficient mismatch repair (dMMR) status according to immunohistochemistry as the reference standard and compared versus the Promega pentaplex MSI panel. The sensitivity of the LMR panel for detection of dMMR status by immunohistochemistry was 99% for CRC and 96% for non-CRC. The overall percent agreement between the LMR and Promega pentaplex panels was 99% for CRC and 89% for non-CRC tumors. An increased number of unstable markers and the larger size shifts observed in dMMR tumors using the LMR panel increased confidence in MSI determinations. The LMR MSI Analysis System expands the spectrum of cancer types in which MSI can be accurately detected.
Collapse
Affiliation(s)
- Jeffery W Bacher
- R&D Clinical Diagnostics, Promega Corporation, Madison, Wisconsin; Department of Medicine, University of Wisconsin, Madison, Wisconsin.
| | - Eshwar B Udho
- R&D Clinical Diagnostics, Promega Corporation, Madison, Wisconsin
| | | | - Irina Vyazunova
- R&D Clinical Diagnostics, Promega Corporation, Madison, Wisconsin
| | - Steven Gallinger
- Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, Victoria, Australia; Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Rish K Pai
- Health Science Research, Mayo Clinic, Scottsdale, Arizona
| | | | - Douglas R Storts
- R&D Clinical Diagnostics, Promega Corporation, Madison, Wisconsin
| | - James R Eshleman
- School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Richard B Halberg
- Department of Medicine, University of Wisconsin, Madison, Wisconsin; Department of Oncology, McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.
| |
Collapse
|
9
|
Mendiola M, Heredia-Soto V, Ruz-Caracuel I, Baillo A, Ramon-Patino JL, Escudero FJ, Miguel M, Pelaez-Garcia A, Hernandez A, Feliu J, Hardisson D, Redondo A. Comparison of Methods for Testing Mismatch Repair Status in Endometrial Cancer. Int J Mol Sci 2023; 24:14468. [PMID: 37833916 PMCID: PMC10572657 DOI: 10.3390/ijms241914468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Approximately 20-30% of endometrial carcinomas (EC) are characterized by mismatch repair (MMR) deficiency (dMMR) or microsatellite instability (MSI), and their testing has become part of the routine diagnosis. The aim of this study was to establish and compare the MMR status using various approaches. Immunohistochemistry (IHC), PCR-based MSI, and the detection of defects in the four key MMR genes (MLH1, PMS2, MSH2, and MSH6) via methylation-specific multiplex ligation-dependent probe amplification (MLPA) and targeted next-generation sequencing (NGS) were performed. MSH3 expression was also evaluated. A set of 126 early-stage EC samples were analyzed, 53.2% of which were dMMR and 46.8% of which were proficient MMR (pMMR) as determined using IHC, whereas 69.3% were classified as microsatellite stable, while 8.8% and 21.9% were classified MSI-low (MSI-L) and MSI-high (MSI-H), respectively. In total, 44.3% of the samples showed genetic or epigenetic alterations in one or more genes; MLH1 promoter methylation was the most common event. Although acceptable concordance was observed, there were overall discrepancies between the three testing approaches, mainly associated with the dMMR group. IHC had a better correlation with MMR genomic status than the MSI status determined using PCR. Further studies are needed to establish solid conclusions regarding the best MMR assessment technique for EC.
Collapse
Affiliation(s)
- Marta Mendiola
- Molecular Pathology and Therapeutic Targets Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain; (M.M.); (A.P.-G.); (D.H.)
- Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.H.-S.); (J.F.)
| | - Victoria Heredia-Soto
- Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.H.-S.); (J.F.)
- Translational Oncology Research Laboratory, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
| | - Ignacio Ruz-Caracuel
- Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.H.-S.); (J.F.)
- Department of Pathology, La Paz University Hospital, 28046 Madrid, Spain
| | - Amparo Baillo
- Mathematics Department, Autonomous University of Madrid, 28049 Madrid, Spain;
| | | | - Francisco Javier Escudero
- Translational Oncology Research Laboratory, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
| | - Maria Miguel
- Molecular Pathology and Therapeutic Targets Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain; (M.M.); (A.P.-G.); (D.H.)
| | - Alberto Pelaez-Garcia
- Molecular Pathology and Therapeutic Targets Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain; (M.M.); (A.P.-G.); (D.H.)
| | - Alicia Hernandez
- Department of Obstetrics and Gynecology, La Paz University Hospital, 28046 Madrid, Spain;
- Faculty of Medicine, Autonomous University of Madrid, 28046 Madrid, Spain
| | - Jaime Feliu
- Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.H.-S.); (J.F.)
- Translational Oncology Research Laboratory, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
- Department of Medical Oncology, La Paz University Hospital, 28046 Madrid, Spain;
- Faculty of Medicine, Autonomous University of Madrid, 28046 Madrid, Spain
- Cátedra UAM-ANGEM, Faculty of Medicine, Autonomous University of Madrid, 28046 Madrid, Spain
| | - David Hardisson
- Molecular Pathology and Therapeutic Targets Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain; (M.M.); (A.P.-G.); (D.H.)
- Center for Biomedical Research in the Cancer Network (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.H.-S.); (J.F.)
- Department of Pathology, La Paz University Hospital, 28046 Madrid, Spain
- Faculty of Medicine, Autonomous University of Madrid, 28046 Madrid, Spain
| | - Andres Redondo
- Translational Oncology Research Laboratory, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
- Department of Medical Oncology, La Paz University Hospital, 28046 Madrid, Spain;
- Faculty of Medicine, Autonomous University of Madrid, 28046 Madrid, Spain
- Cátedra UAM-ANGEM, Faculty of Medicine, Autonomous University of Madrid, 28046 Madrid, Spain
| |
Collapse
|
10
|
Malapelle U, Orsulic S. Editorial: Molecular pathology and computational image analyses in gynecologic malignancies. Front Oncol 2022; 12:1082220. [PMID: 36479087 PMCID: PMC9720375 DOI: 10.3389/fonc.2022.1082220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/04/2022] [Indexed: 11/22/2022] Open
|
11
|
Salvati A, Carnevali I, Alexandrova E, Facchi S, Ronchi S, Libera L, Sahnane N, Memoli D, Lamberti J, Amabile S, Pepe S, Tarallo R, Sessa F, Weisz A, Tibiletti MG, Rizzo F. Targeted molecular profiling of epithelial ovarian cancer from Italian BRCA wild-type patients with a BRCA and PARP pathways gene panel. Exp Mol Pathol 2022; 128:104833. [PMID: 36165864 DOI: 10.1016/j.yexmp.2022.104833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 09/01/2022] [Accepted: 09/15/2022] [Indexed: 12/15/2022]
Abstract
Ovarian cancer (OC) is the fifth most common type of cancer in women and the fourth most common cause of cancer death in women. Identification of pathogenic variants in OC tissues has an important clinical significance for therapeutic and prevention purposes. This study aims to evaluate the mutational profile of a patient cohort, negative for BRCA1/2 germinal variants and Mismatch Repair defects, using next-generation sequencing (NGS) approach on DNA from formalin-fixed paraffin-embedded samples. We used a custom NGS panel, targeting 34 cancer-related genes, mainly of the BRCA and PARP pathways, and analyzed NGS data to identify somatic and germline variants in Italian patients affected by primary epithelial ovarian cancer. We analyzed 75 epithelial ovarian cancer tissues and identified 54 pathogenic variants and 56 variants of unknown significance. TP53 was characterized by the highest mutational rate, occurring in 55% of tested epithelial ovarian cancers (EOCs). Interestingly, a subset of 8 EOCs showed pathogenic variants of homologous recombination pathway, which could be sensitive to PARP-inhibitor therapies. Germline analysis of actionable genes revealed 4 patients carrier of pathogenic germline variants respectively of RAD51C (2 patients), RAD51D, and PALB2. Molecular profiling of EOCs using our custom NGS panel has enabled the detection of both somatic and germline variants, allowing the selection of patients suitable for targeted therapies, and the identification of high-risk OC families that can benefit from genetic counseling and testing.
Collapse
Affiliation(s)
- Annamaria Salvati
- Medical Genomics Program and Division of Oncology, AOU'S. Giovanni di Dio e Ruggi d'Aragona' Università di Salerno, 84131 Salerno, Rete Oncologica Campana, Italy; Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081 Baronissi, Italy
| | - Ileana Carnevali
- Department of Pathology, Ospedale di Circolo - ASST Settelaghi and Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery of the University of Insubria, 21100 Varese, Italy
| | - Elena Alexandrova
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081 Baronissi, Italy
| | - Sofia Facchi
- Department of Pathology, Ospedale di Circolo - ASST Settelaghi and Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery of the University of Insubria, 21100 Varese, Italy
| | - Susanna Ronchi
- Department of Pathology, Ospedale di Circolo - ASST Settelaghi and Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery of the University of Insubria, 21100 Varese, Italy
| | - Laura Libera
- Department of Pathology, Ospedale di Circolo - ASST Settelaghi and Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery of the University of Insubria, 21100 Varese, Italy
| | - Nora Sahnane
- Department of Pathology, Ospedale di Circolo - ASST Settelaghi and Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery of the University of Insubria, 21100 Varese, Italy
| | - Domenico Memoli
- Medical Genomics Program and Division of Oncology, AOU'S. Giovanni di Dio e Ruggi d'Aragona' Università di Salerno, 84131 Salerno, Rete Oncologica Campana, Italy; Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081 Baronissi, Italy
| | - Jessica Lamberti
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081 Baronissi, Italy
| | - Sonia Amabile
- Medical Genomics Program and Division of Oncology, AOU'S. Giovanni di Dio e Ruggi d'Aragona' Università di Salerno, 84131 Salerno, Rete Oncologica Campana, Italy
| | - Stefano Pepe
- Medical Genomics Program and Division of Oncology, AOU'S. Giovanni di Dio e Ruggi d'Aragona' Università di Salerno, 84131 Salerno, Rete Oncologica Campana, Italy
| | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081 Baronissi, Italy; Genome Research Center for Health - CRGS, Campus of Medicine of the University of Salerno, 84081 Baronissi, SA, Italy
| | - Fausto Sessa
- Department of Pathology, Ospedale di Circolo - ASST Settelaghi and Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery of the University of Insubria, 21100 Varese, Italy
| | - Alessandro Weisz
- Medical Genomics Program and Division of Oncology, AOU'S. Giovanni di Dio e Ruggi d'Aragona' Università di Salerno, 84131 Salerno, Rete Oncologica Campana, Italy; Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081 Baronissi, Italy; Genome Research Center for Health - CRGS, Campus of Medicine of the University of Salerno, 84081 Baronissi, SA, Italy.
| | - Maria Grazia Tibiletti
- Department of Pathology, Ospedale di Circolo - ASST Settelaghi and Research Center for the Study of Hereditary and Familial Tumors, Department of Medicine and Surgery of the University of Insubria, 21100 Varese, Italy.
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, 84081 Baronissi, Italy; Genome Research Center for Health - CRGS, Campus of Medicine of the University of Salerno, 84081 Baronissi, SA, Italy.
| |
Collapse
|