Advertisement
Research Article|Articles in Press

Microsatellite Instability in Non-Endometrioid Ovarian Epithelial Tumors: A study of 400 cases Comparing Immunohistochemistry, PCR, and NGS Based Testing with Mutation Status of MMR Genes.

Open AccessPublished:May 24, 2023DOI:https://doi.org/10.1016/j.trsl.2023.05.004

      ABSTRACT

      Testing of microsatellite instability is not only used as a triage for possible Lynch syndrome, but also to predict immunotherapy treatment response. The aim of this study was to assess the frequency of mismatch repair deficiency (MMR-D) / microsatellite instability (MSI) in 400 cases of non-endometrioid ovarian tumors (high-grade serous, low-grade serous, mucinous and clear cell), to compare different methodological approaches of testing, and to assess the optimal approach for next generation sequencing (NGS) MSI testing. For all tumors, we evaluated immunohistochemical (IHC) expression of MMR proteins and assessed microsatellite markers by PCR-based method. Except for high-grade serous carcinoma, we correlated the findings of IHC and PCR with NGS-based MSI testing. We compared the results with somatic and germline mutation in MMR genes. Among the whole cohort, seven MMR-D cases, all clear cell carcinomas (CCC), were found. On PCR analysis, six cases were MSI-high and one was MSS. In all cases, mutation of an MMR gene was found; in two cases, the mutation was germline (Lynch syndrome). An additional five cases with a mutation in MMR gene(s) with MSS status and without MMR-D were identified. We further utilized sequence capture NGS for MSI testing. Employing 53 microsatellite loci provided high sensitivity and specificity. Our study shows that MSI occurs in 7% of CCC while it is rare or absent in other non-endometrioid ovarian neoplasms. Lynch syndrome was present in 2% of patients with CCC. However, some cases with MSH6 mutation can evade all testing methods, including IHC, PCR, and NGS-MSI.

      Abbreviations:

      CAP (College of American Pathologists), CCC (clear cell carcinoma), ESMO (European Society for Medical Oncology), FFPE (formalin-fixed and paraffin-embedded), HGSC (high-grade serous carcinoma), IHC (immunohistochemical), LGSC (low-grade serous carcinoma), LS (Lynch syndrome), MBT (mucinous borderline tumor), MC (mucinous carcinoma), MMR (mismatch repair), MMR-D (mismatch repair deficiency), MMR-P (proficient mismatch repair), mSBT (micropapillary subtype of serous borderline tumor), MSI (microsatellite instability), MSI-H (high microsatellite instability), mut/Mb (mutations per megabase), NGS (next generation sequencing), PCR (polymerase chain reaction), PCR-FA (polymerase chain reaction and fragment analysis), TCGA (The Cancer Genome Atlas), TMAs (tissue microarrays), TMB (tumor mutation burden)

      INTRODUCTION

      Testing of colorectal and endometrial carcinomas for mismatch repair (MMR) protein expression by immunohistochemistry (IHC) or for microsatellite instability (MSI) by PCR is a well-established screening method for Lynch syndrome (LS) [
      • Hampel H
      • Frankel W
      • Panescu J
      • et al.
      Screening for Lynch syndrome (hereditary nonpolyposis colorectal cancer) among endometrial cancer patients.
      ,

      Kim SR, Tone A, Kim RH, et al. Performance characteristics of screening strategies to identify Lynch syndrome in women with ovarian cancer. Cancer-Am Cancer Soc. 2020.

      ,
      • Lynch HT
      • Lynch PM
      • Lanspa SJ
      • et al.
      Review of the Lynch syndrome: history, molecular genetics, screening, differential diagnosis, and medicolegal ramifications.
      ,
      • Rambau PF
      • Duggan MA
      • Ghatage P
      • et al.
      Significant frequency of MSH2/MSH6 abnormality in ovarian endometrioid carcinoma supports histotype-specific Lynch syndrome screening in ovarian carcinomas.
      ,
      • Ryan N
      • Wall J
      • Crosbie EJ
      • et al.
      Lynch syndrome screening in gynaecological cancers: results of an international survey with recommendations for uniform reporting terminology for mismatch repair immunohistochemistry results.
      ]. Some ovarian carcinomas are also associated with LS but the optimal screening strategy to identify LS in ovarian cancer patients has not been determined, yet [

      Kim SR, Tone A, Kim RH, et al. Performance characteristics of screening strategies to identify Lynch syndrome in women with ovarian cancer. Cancer-Am Cancer Soc. 2020.

      ,
      • Concin N
      • Matias-Guiu X
      • Vergote I
      • et al.
      ESGO/ESTRO/ESP guidelines for the management of patients with endometrial carcinoma.
      ]. Currently, the significance of MSI testing is increasing in a broad spectrum of tumors. MMR deficiency (MMR-D) and/or presence of high MSI (MSI-H) are predictors of a favorable response to immune checkpoint inhibitor therapy in solid tumors [
      • Le DT
      • Uram JN
      • Wang H
      • et al.
      PD-1 Blockade in Tumors with Mismatch-Repair Deficiency.
      ,
      • Bartley AN
      • Mills AM
      • Konnick E
      • et al.
      Mismatch Repair and Microsatellite Instability Testing for Immune Checkpoint Inhibitor Therapy: Guideline From the College of American Pathologists in Collaboration With the Association for Molecular Pathology and Fight Colorectal Cancer.
      ]. In parallel with predictive testing in the broad spectrum of solid tumors, the knowledge about non-canonical neoplasms which can be associated with LS is increasing [
      • Latham A
      • Srinivasan P
      • Kemel Y
      • et al.
      Microsatellite Instability Is Associated With the Presence of Lynch Syndrome Pan-Cancer.
      ].
      Methods used for MSI testing have been mostly validated for screening purposes in colorectal and endometrial carcinomas. However, due to technical or biological reasons there are challenges in MSI testing even in colorectal and endometrial cancer. As such, immunohistochemical testing of the mismatch repair machinery may give different results for a given germline or somatic mismatch repair gene missense mutation [
      • Hechtman JF
      • Rana S
      • Middha S
      • et al.
      Retained mismatch repair protein expression occurs in approximately 6% of microsatellite instability-high cancers and is associated with missense mutations in mismatch repair genes.
      ,
      • Adeleke S
      • Haslam A
      • Choy A
      • et al.
      Microsatellite instability testing in colorectal patients with Lynch syndrome: lessons learned from a case report and how to avoid such pitfalls.
      ]. However, in other tumors the value of different approaches is not clear. The three main methods for MSI testing are immunohistochemistry (IHC), PCR based approaches and NGS testing. According to the European Society for Medical Oncology (ESMO) recommendation on MSI testing for immunotherapy in cancer, the preferable method of testing in the first line is IHC with 4 antibodies (MLH1, PMS2, MSH2 and MSH6) [
      • Luchini C
      • Bibeau F
      • Ligtenberg MJL
      • et al.
      ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: a systematic review-based approach.
      ]. PCR based approaches are for second line testing, in cases in which IHC is not evaluable or the results are inconclusive. However, these recommendations are for tumors belonging to the spectrum of cancer well-known to be associated with LS, including colorectal, endometrial, small intestine, urothelial, central nervous system and sebaceous gland neoplasms. For other tumor types, there are insufficient data to inform a recommendation. The ESMO comment on NGS MSI testing is that it has the potential to become the method of choice for all tumor types.
      In contrast, a recent College of American Pathologists (CAP) guideline for testing for possible immunotherapy with immune checkpoint inhibitors, considers IHC and PCR as equal and states that one of these methods should be used for colorectal, small bowel and gastroesophageal carcinoma [
      • Bartley AN
      • Mills AM
      • Konnick E
      • et al.
      Mismatch Repair and Microsatellite Instability Testing for Immune Checkpoint Inhibitor Therapy: Guideline From the College of American Pathologists in Collaboration With the Association for Molecular Pathology and Fight Colorectal Cancer.
      ]. This guideline states that for endometrial carcinoma, IHC is favored over PCR testing while for other tumor types, the optimal method of testing has not been established, so far. NGS may be used for colorectal cancer, but a validated MSI NGS assay is needed. For small bowel, gastroesophageal and endometrial carcinoma MMR-IHC, MSI-PCR and MMR-IHC, respectively, should be preferred over NGS. NGS is currently not the method of choice for primary isolated MSI testing. However, MSI could be evaluated by NGS panel testing together with the assessment of the mutation profile of the tumor and tumor mutation burden (TMB). However, the set of microsatellite loci analyzed by NGS have been established for colorectal and endometrial carcinomas and can differ in other tumors. Therefore, the optimal NGS approach covering all solid tumors has not been assessed, yet. Despite the recommendation for IHC testing using all 4 antibodies, first-line testing for only 2 MMR proteins (PMS2 and MSH6) can be used, if required completed by second-line testing of MLH1 and MSH2 [
      • Aiyer KTS
      • Doeleman T
      • Ryan NA
      • et al.
      Validity of a two-antibody testing algorithm for mismatch repair deficiency testing in cancer; a systematic literature review and meta-analysis.
      ].
      A recently developed rapid digital PCR technique for microsatellite instability has been successfully used for colorectal, endometrial and gastric carcinomas and also a small number of ovarian carcinomas without histological characterization. This novel technique is easy to handle and fast and turned out comparable to other PCR based techniques and immunohistochemistry [
      • Gatius S
      • Velasco A
      • Varela M
      • et al.
      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.
      ,
      • Velasco A
      • Tokat F
      • Bonde J
      • et al.
      Multi-center real-world comparison of the fully automated Idylla microsatellite instability assay with routine molecular methods and immunohistochemistry on formalin-fixed paraffin-embedded tissue of colorectal cancer.
      ,
      • Farmkiss L
      • Hopkins I
      • Jones M.
      Idylla microsatellite instability assay versus mismatch repair immunohistochemistry: a retrospective comparison in gastric adenocarcinoma.
      ,
      • Samaison L
      • Uguen A.
      Idylla MSI test combined with immunohistochemistry is a valuable and cost effective strategy to search for microsatellite instable tumors of noncolorectal origin.
      ,
      • Pecriaux A
      • Favre L
      • Calderaro J
      • et al.
      Detection of microsatellite instability in a panel of solid tumours with the Idylla MSI Test using extracted DNA.
      ].
      According to the literature, MSI occurs in approximately 13% of ovarian endometrioid carcinomas (EC) [

      Kim SR, Tone A, Kim RH, et al. Performance characteristics of screening strategies to identify Lynch syndrome in women with ovarian cancer. Cancer-Am Cancer Soc. 2020.

      ,
      • Tajima Y
      • Eguchi H
      • Chika N
      • et al.
      Prevalence and molecular characteristics of defective mismatch repair epithelial ovarian cancer in a Japanese hospital-based population.
      ,
      • Segev Y
      • Pal T
      • Rosen B
      • et al.
      Risk Factors for Ovarian Cancers With and Without Microsatellite Instability.
      ,
      • Simila-Maarala J
      • Soovares P
      • Pasanen A
      • et al.
      TCGA molecular classification in endometriosis-associated ovarian carcinomas: Novel data on clear cell carcinoma.
      ]. However, the published literature concerning MMR-D/MSI-H frequency in non-endometrioid ovarian tumors is equivocal [
      • Rambau PF
      • Duggan MA
      • Ghatage P
      • et al.
      Significant frequency of MSH2/MSH6 abnormality in ovarian endometrioid carcinoma supports histotype-specific Lynch syndrome screening in ovarian carcinomas.
      ,
      • Fraune C
      • Rosebrock J
      • Simon R
      • et al.
      High homogeneity of MMR deficiency in ovarian cancer.
      ,
      • Bennett JA
      • Morales-Oyarvide V
      • Campbell S
      • Longacre TA
      • Oliva E.
      Mismatch Repair Protein Expression in Clear Cell Carcinoma of the Ovary: Incidence and Morphologic Associations in 109 Cases.
      ,
      • Carter NJ
      • Marshall ML
      • Susswein LR
      • et al.
      Germline pathogenic variants identified in women with ovarian tumors.
      ,
      • Lu FI
      • Gilks CB
      • Mulligan AM
      • et al.
      Prevalence of Loss of Expression of DNA Mismatch Repair Proteins in Primary Epithelial Ovarian Tumors.
      ]. The aim of our study was to assess the frequency of MMR-D/MSI-H in a broad spectrum of non-endometrioid ovarian tumors, to compare different methodological approaches for MSI testing, and to assess the optimal approach for NGS MSI testing using three different sets of microsatellite markers. Our sample set consisted of 400 primary ovarian epithelial tumors with serous, mucinous and clear cell morphology. For all tumors, we evaluated IHC expression of four MMR proteins and assessed the stability of microsatellite markers by the PCR-based method of five mononucleotides. Moreover, for all tumors except high-grade serous carcinoma (HGSC), we correlated the findings of IHC and PCR with NGS-based MSI testing. In all tumors in which NGS was performed, the results were also compared with mutation status in MMR genes. For tumors showing MMR-D/MSI-H status, non-neoplastic tissue was tested by NGS parallel to tumor tissue to exclude the possibility of LS.

      MATERIALS AND METHODS

      Samples

      The archives of participating pathology departments were searched for cases diagnosed as low-grade serous carcinoma (LGSC), micropapillary subtype of serous borderline tumor (mSBT), clear cell carcinoma (CCC), mucinous carcinoma (MC) and mucinous borderline tumor (MBT). The sample set represents part of the sample set used in our previous studies [
      • Dundr P
      • Bartu M
      • Bosse T
      • et al.
      Primary Mucinous Tumors of the Ovary: An Interobserver Reproducibility and Detailed Molecular Study Reveals Significant Overlap Between Diagnostic Categories.
      ,
      • Dundr P
      • Bazalova B
      • Bartu M
      • et al.
      The cytokeratin 17 expression in primary ovarian tumors has diagnostic but not prognostic significance.
      ,
      • Nemejcova K
      • Bartu MK
      • Michalkova R
      • et al.
      A comprehensive immunohistochemical analysis of IMP2 and IMP3 in 542 cases of ovarian tumors.
      ]. 296 selected samples were eligible for IHC, PCR and NGS testing. These cases included 100 CCC, 75 LGSC, 29 mSBT and 92 mucinous tumors (29 MC, 49 MBT and 14 cases equivocal between MC and MBT as described in Dundr et al. 2023). Moreover, 104 tubo-ovarian high grade serous carcinomas (HGSC) were selected from the archives of the Department of Pathology, 1st Medical Faculty and General University Hospital in Prague. These cases were analyzed only by IHC and PCR methods. All LGSC, mSBT, HGSC and CCC cases were reviewed by at least two experienced pathologists and fulfilled the strict morphological and immunohistochemical criteria. The classification of mucinous tumors was based on the results of our previous study, which focused on the interobserver agreement and molecular analysis of ovarian mucinous tumors [
      • Dundr P
      • Bartu M
      • Bosse T
      • et al.
      Primary Mucinous Tumors of the Ovary: An Interobserver Reproducibility and Detailed Molecular Study Reveals Significant Overlap Between Diagnostic Categories.
      ]. Clinicopathological characteristics of the analyzed tumors are in Table 1.
      Table 1Patients and tumors characteristics
      OCCCLGSCmSBT*mucinous tumors*HGSC
      n. of samplesn= 100n= 75n= 29n= 92n= 104
      age mean (range)60.2 (34-82)52 (19-83)48 (25-85)52 (17-83)59.9 (36-81)
      FIGO
      I671111849
      II73117
      III164211564
      IV120122
      NA917612
      OCCC_ ovarian clear cell carcinoma, LGSC_ low-grade serous carcinoma, mSBT – micropapillary variant of serous borderline tumor, HGSC_ high-grade serous carcinoma, FIGO classification was done according to WHO classification of female genital tumors
      *mucinous tumors comprise of 29 mucinous carcinoma, 49 of mucinous borderline tumor and 14 of cases equivocal between mucinous carcinoma and mucinous borderline tumor
      *HGSC cohort was tested only by MMR IHC and PCR-based method
      The study has been approved by the Ethics Committee of General University Hospital in Prague in compliance with the Helsinki Declaration (No. 2140/19 S-IV). The Ethics Committee waived the requirement for informed consent, as according to the Czech Law (Act. no. 373/11, and its amendment Act no. 202/17) it is not necessary to obtain informed consent in fully anonymized studies.

      Immunohistochemical expression of mismatch repair proteins

      Immunohistochemistry was performed on tissue microarrays (TMAs) using 4μm thick sections of formalin-fixed and paraffin-embedded (FFPE) tissue. For construction of the TMAs, eligible areas of each tumor were identified and two tissue cores (each 2.0 mm in diameter) were taken from the donor block using the tissue microarray instrument TMA Master (3DHISTECH Ltd., Budapest, Hungary).
      Immunohistochemical analysis (IHC) was performed with antibodies against MSH2 (clone FE11, ready-to-use, Dako, USA), MSH6 (clone EP49, ready-to-use, Bio SB, USA), MLH1 (clone ES05, ready-to-use, Dako, USA), and PMS2 (clone EP51, ready-tu-use, Dako, USA) using Dako Omnis (Agilent Technologies, California, USA) with the EnVision FLEX system.
      Loss of MMR protein expression was defined as a loss of nuclear staining in all tumor cells for any of the four MMR proteins with preserved positive internal control (nuclear expression of endothelial cells, lymphocytes and/or stromal cells). Weak nuclear staining (weaker in comparison to the internal control) in less than 5% of tumor cells was regarded as lost. Weak nuclear staining (weaker than the internal control) in more than 5% of tumor cells or negativity of both tumor cells and internal control on the initial TMA sections were regarded as equivocal. All cases with an equivocal result on TMA, with loss of expression of any MMR protein or with an MSI-H status detected by PCR and/or NGS were immunohistochemically reanalyzed on whole tissue sections. All immunohistochemical stains were scored by at least two experienced pathologists (PD, MB, KN).

      Microsatellite instability (MSI) testing by PCR and fragment analysis (PCR-FA)

      A pentaplex PCR reaction was performed with fluorescent labeled primers for the set of five quasi monomorphic mononucleotide microsatellite markers BAT-26, BAT-25, NR-21, NR-22, NR-24, followed by fragmentation analysis on ABI 3500 (ThermoFisher). Size of PCR products was evaluated in GeneMapper™ Software (ThermoFisher). MSI-high (MSI-H) phenotype was defined as the presence of two or more unstable microsatellite markers. Cases with one unstable marker (MSI-low) were included in a group of MSS tumors.

      Capture DNA NGS

      For the purpose of this study, samples used as a part of a large project that focused on rare epithelial ovarian tumors were included. For all samples (100 CCC, 75 LGSC, 29 mSBT and 92 mucinous tumors) the NGS sequencing was part of our previous studies, the methodology is detailed there [
      • Dundr P
      • Bartu M
      • Bosse T
      • et al.
      Primary Mucinous Tumors of the Ovary: An Interobserver Reproducibility and Detailed Molecular Study Reveals Significant Overlap Between Diagnostic Categories.
      ]. DNA was extracted also from the adjacent non-neoplastic tissue (Magcore Genomic DNA FFPE One step kit; RBC Bioscience) for sequencing analysis to rule out a potential germline origin of MMR gene mutation in cases with detected MMR mutation in the tumor. The NGS capture panel used in the current and previous studies included 727 genes or gene parts (2097 kbp; NimbleGen, Roche). In this study, we focused only on detected mutations in mismatch repair genes MLH1, PMS2, MSH2, MSH6, and on evaluation of microsatellite markers as described below.

      MSI testing by NGS approach

      The set of NGS 17 loci included standard recommended Bethesda markers BAT-26, BAT-25, NR-21, NR-22, NR-24, D5S346 [
      • Umar A
      • Boland CR
      • Terdiman JP
      • et al.
      Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability.
      ], and microsatellite loci in cancer-related genes TGFBR2, BAX, IGF-II. These are frequently targeted by microsatellite instability and their inactivation may contribute to tumor progression [
      • Shima K
      • Morikawa T
      • Yamauchi M
      • et al.
      TGFBR2 and BAX Mononucleotide Tract Mutations, Microsatellite Instability, and Prognosis in 1072 Colorectal Cancers.
      ]. In addition, MONO-27, Penta-C, ACVR2A, BTBD7, DIDO1, MRE11A, RYR3, SEC31A were included. These are used in commercial kits for testing microsatellite instability in different types of tumors, in particular, colorectal and endometrial cancer (Promega, IdyllaTM) [
      • Zhao H
      • De Craene B
      • Sagaert X
      • et al.
      Association of a novel set of 7 homopolymer indels for detection of MSI with tumor mutation burden and total indel load in endometrial and colorectal cancers.
      ]. The NGS raw data were processed (including trim reads, mapping reads to GrCH38 reference genome) by QIAGEN CLC Genomics Workbench software (Qiagen). Loci track of our 17 microsatellite markers were imported to the module “Detect MSI status”. Fifteen samples with optimal DNA quality and with known MMR (IHC) and MSS (PCR-FA) status were used for creating a MSI-baseline. Default setting of this module was used. The “Detect MSI Status” module measures the statistical variation of the length distribution of each microsatellite locus and determines the stability of each locus by comparing the statistical variation of the tested sample with the normal baseline samples.
      To increase the specificity of MSI detection in ovarian tumors, we identified in our targeted custom panel (2097 kbp) all microsatellite markers (1–5 bp in length and comprising 5 repeats or more) by comparison with a set of genome-wide microsatellite markers described by Hause et al [
      • Hanse RJ
      • Pritchard CC
      • Shendure J
      • Salipante SJ.
      Classification and characterization of microsatellite instability across 18 cancer types.
      ]. The algorithm for selection of ovarian-specific microsatellite loci is shown in Figure 1. Out of 10,249 microsatellite loci identified (with sufficient read depth) 36 ovarian-tumors specific loci were selected. They were unstable in at least 4 out of 6 MSI-H samples in our sample set (previously evaluated by PCR-FA), while stable in all MSS and proficient mismatch repair (MMR-P) samples. The most unstable microsatellite markers identified in >5 MSI-H tumors are summarized in Table 2.
      Figure 1
      Figure 1Examples of immunohistochemical staining for the MMR proteins.
      Table 2Spectrum of the most unstable markers in MSI rare EOCs.
      Locus coordinates (GRCh37)Nucleotide repetitive motifgene (exon or adjacent intron region)
      chr.15:41803530-41803544(C)5LTK (exonic)
      chr.17:30293145-30293168(T)14SUZ12 (intronic)
      chr.10:27333117-27333138(A)12ANKRD26 (intronic)
      chr.11:108141951-108141975(T)15ATM (intronic)
      chr.11:112832387-112832410(T)14NCAM1 (intronic)
      chr.13:48954278-48954298(T)11RB1 (intronic)
      chr.15:41991032-41991056(T)15MGA (intronic)
      chr.2:48032736-48032758(T)13MSH6 (intronic)
      chr.3:138664706-138664720(G)5FOXL2 (exonic)
      chr.8:30954240-30954284(T)13c(T)6cttttgtttg(T)5WRN (intronic)
      chr.9:8341276-8341304(A)12gg(A)5PTPRD (intronic)
      There are shown selected unstable loci, which was detected in at least in 5 MSI-high OCCCs

      Statistical analyses

      All statistical tests were carried out using the program R (version 4.0.2, https://www.r-project.org/) and /or Statistica (TIBCO). Association between age (continuous variable) and MSI/MSS status (dichotomous variable) was evaluated using the Mann-Whitney U test. To analyze the agreement among three methodical approaches (IHC, PCR-FA and NGS), the methods of inter-rater reliability was implemented using the package “irr” in the R program (available at http://CRAN.R-project.org/package=irr). The level of agreement among three methods was described using Fleiss Kappa coefficient (κ). Consistent with prior literature on the level of agreement (Landis and Koch 1977), kappa coefficients were interpreted as poor (0.01-0.20), fair (0.21-0.40), moderate (0.41-0.60), substantial (0.61-0.80) and almost perfect (0.81-1.00). The package ‘cutpointr’ implemented in R software (https://github.com/thie1e/cutpointr) was used for the evaluation of an optimal cut-point value for sensitivity and specificity. All tests were two-sided and a p-value of less than 0.05 was considered as significant.

      RESULTS

      Among the whole cohort of 400 tumors, 7 MMR-D cases were found (Figure 1); 6 of these were MSI-H and one MSS on PCR analysis. All these cases were CCC. In all these tumors, mutation of an MMR gene was found. In two of these cases, the mutation was germline (MLH1 and MSH6, respectively) and in 5 cases, the mutation was somatic (in 2 cases each MSH2 and MSH6, in one case MLH1). In 5/7 cases, in which TMB was evaluable, high TMB (range 2 - 86) was found. The case with the highest TMB (TMB=86) contained a concurrent POLE mutation. In addition, there were 5 cases identified with mutation in MMR genes but with a MSS status. Two of these cases showed a germline mutation of MSH6. Two MMR proficient cases with a somatic mutation of an MMR gene (one case with MLH1 and concurrent PMS2 mutation, second case with MSH2 mutation) showed high TMB, but both showed concurrent POLE mutation. The results are summarized in Table 3.
      Table 3List of cases with detected pathogenic mutation in MMR genes, including results of MMR IHC, PCR based method and NGS approach for evaluation of MMR-D/MSI status.
      IHCPCRNGSMutation analysis
      dg.age at dg.MLH1PMS2MSH2MSH6MMR-D/

      MMR-P
      MSI/

      MSS
      53 lociMMR genes

      G_Germline

      S_Somatic
      POLE geneTMB (mut/Mb)
      % of positively stained nucleus
      CCC44989500MMR-DMSI55%S MSH2:c.2375_2378del,p.(N792fs)noNA
      CCC44139540MMR-DMSI45%G MLH1:c.116+1G>A,p.?no2
      CCC3910070045MMR-DMSI66%S MSH2:c.1386+1G>A,p.?no14
      CCC451110090MMR-DMSI79%S MLH1:c.1459C>T,p.(R487*)no26**
      CCC4555855510MMR-DMSI55%S MSH6:c.1610_1613del,p.(K537fs)no25
      CCC59100100905MMR-DMSI55%G MSH6:c.1238G>C,p.(W413S)no18**
      CCC6610085905MMR-DMSS23%S MSH6:c.1630G>T,p.(E544*)c.1376C>T,p.(S459F)86
      CCC55100607045MMR-PMSS0%S MLH1:c.1896+1G>A,p.? and

      S PMS2:c.1882C>T,p.(R628*)
      c.1366G>C,p.(A456P)66**
      CCC6890952555MMR-PMSS8%S MSH6:c.1805C>G,p.(S602*)no7
      CCC4810010010095MMR-PMSS6%S MSH2:c.1447G>T,p.(E483*)c.1231G>C,p.(V411L)51
      MBT4190809020MMR-PMSS4%G MSH6:c.2351_2352del,p.(N784fs)no1**
      LGSC4680209035MMR-PMSS8%G MSH6:c.3226C>T,p.(R1076C)no1
      dg._histological diagnosis, IHC_immunohistochemistry, MMR_miss match repair, CCC_ clear cell carcinoma, LGSC_low grade serous carcinoma, MBT_mucinous borderline tumor, PCR-FA_Polymerase chain reaction and fragment analysis, NGS_Next generation sequencing
      MMR-P_proficent mismatch repair protein staining, MMR-D_deficient/loss of mismatch repair protein staining, MSI_microsatellite instable, MSS_microsatellite stable
      TMB_tumor mutation burden, mut/Mb_ mutation per megabase, ** TMB in this samples could be higher because of the purity of tumors cell of the input DNA under 40%,
      NA_data are not available
      reference sequences for MSH2: NM_000251.2; MLH1: NM_000249.3; MSH6: NM_000179.2; PMS2: NM_000535.5; NM_006231.2, POLE: NM_006231.2
      Thirteen cases (8 CCC, 3 LGSC, 2 mucinous tumors) were MSI-low by PCR. These tumors were classified as MSS by using the NGS approach and immunohistochemistry showed proficient expression for all MMR proteins. The most frequently detected unstable microsatellite in these cases was NR-21 (in 8 of 13).
      Pathogenic mutation or likely pathogenic mutation in at least one MMR gene was found in 12/296 (4 %) cases (10 OCC, 1 MBT, 1 LGSC; Table 3). Out of those, seven cases were MMR-D, six cases were MSI-H and 6 cases were MSS. Three MSS samples with mutation in the MMR gene had a concurrent mutation in the POLE gene and had also the highest TMB (range 51-86 mut/Mb; mean 67.7). TMB in 6 MSI-H tumors ranged between 2-26 mut/Mb (mean 17 mut/Mb).
      The sequence capture NGS-based approach for MSI testing showed higher sensitivity and specificity using 53 selected microsatellite loci, based on the analysis of our MMR-D and MSS cases compared to using 17 microsatellite loci typical for colorectal and endometrial carcinoma and to analyzing all microsatellite loci present in our NGS DNA panel (Figure 2). Using the 17 microsatellite loci panel, the MMR-D cases showed between 18-65% of unstable microsatellites (mean 30%). The cut-off value for MSI-H status by this panel was 18% of unstable loci (sensitivity=1, specificity= 0.97, AUC = 0.996). Using the 53 microsatellite loci panel the MMR-D cases showed 23-79% of unstable microsatellites (mean 51%). The optimal cut-off value for MSI-H status by this panel was 23% of unstable loci (sensitivity=1, specificity=1, AUC= 1). Analysis of all microsatellite loci (10,249) showed a cut-off value of 5% for MSI-H (sensitivity = 1, specificity = 0.65, AUC = 0.807).
      Figure 2
      Figure 2Design of NGS-based approach for the testing of microsatellite instability in ovarian tumors.
      PCR-FA_polymerase chain reaction and fragment analysis (5 mononucleotide markers); IHC MMR_ immunohistochemical analysis of 4 mismatch repair protein MLH1, MSH2, MSH6, and PMS2; NGS – next generation sequencing (capture DNA NGS, panel of 727 genes including MLH1, MSH2, MSH6 and PMS2); MSI_microsatellite instability; MMR-D_deficient mismatch repair; MMR-P_proficient mismatch repair; MSS-baseline was calculated from randomly chosen set of MSS samples with sufficient read depth for automated analysis in CLC Genomic Workbench software (Qiagen).
      As the best result of both specificity and sensitivity was reached by the 53-loci panel, this panel was selected for further analysis of methods agreement.
      The agreement between evaluation of MMR/MSI by different methods (IHC/PCR-FA/NGS – 53 loci panel) was almost perfect (Kappa = 0.949, Z = 27.9, p < 0.001). Only one case showed discrepancy in the PCR approach (MSS) versus NGS and IHC (MSI in both methods).

      Clinicopathological correlations

      The average age of MMR-D CCC patient was significantly lower compared to MSS CCC patients (mean/median age 48.9/45 years versus 61.5/62 years, p < 0.05). All seven MMR-D cases were FIGO stage I.
      There was no significant difference for the onset of the disease between patients with germline (n = 4) and somatic (n = 8) MMR gene mutations (age 47.5 and 51.2, respectively, p = 0.610).

      DISCUSSION

      It has been shown that 15-25% of all ovarian carcinomas are related to hereditary factors, of which 10-15% are associated with LS [
      • Chui MH
      • Ryan P
      • Radigan J
      • et al.
      The histomorphology of Lynch syndrome-associated ovarian carcinomas: toward a subtype-specific screening strategy.
      ,
      • Shah S
      • Cheung A
      • Kutka M
      • Sheriff M
      • Boussios S.
      Epithelial Ovarian Cancer: Providing Evidence of Predisposition Genes.
      ]. For patients with LS, the estimated lifetime risk of developing ovarian carcinoma is between 6-12% and up to 38% (10-38%) for those with germline MSH2 mutation [

      Kim SR, Tone A, Kim RH, et al. Performance characteristics of screening strategies to identify Lynch syndrome in women with ovarian cancer. Cancer-Am Cancer Soc. 2020.

      ,
      • Bonadona V
      • Bonaiti B
      • Olschwang S
      • et al.
      Cancer risks associated with germline mutations in MLH1, MSH2, and MSH6 genes in Lynch syndrome.
      ,
      • Lu KH
      • Daniels M.
      Endometrial and ovarian cancer in women with Lynch syndrome: update in screening and prevention.
      ]. Several studies have shown that the ovarian tumor types associated with LS are mostly endometrioid carcinoma followed by CCC, the latter accounting for about 12-14% of ovarian carcinomas associated with LS [

      Kim SR, Tone A, Kim RH, et al. Performance characteristics of screening strategies to identify Lynch syndrome in women with ovarian cancer. Cancer-Am Cancer Soc. 2020.

      ,
      • Chui MH
      • Ryan P
      • Radigan J
      • et al.
      The histomorphology of Lynch syndrome-associated ovarian carcinomas: toward a subtype-specific screening strategy.
      ,
      • Helder-Woolderink JM
      • Blok EA
      • Vasen HF
      • et al.
      Ovarian cancer in Lynch syndrome; a systematic review.
      ]. In HGSC, germline pathogenic variants have been described in approximately 25% of cases, mainly in genes involved in the DNA damage response pathway such as BRCA1, BRCA2, BRIP1, PALB2, RAD51, ATM, and CHEK2 [
      • Shah S
      • Cheung A
      • Kutka M
      • Sheriff M
      • Boussios S.
      Epithelial Ovarian Cancer: Providing Evidence of Predisposition Genes.
      ,
      • Lhotova K
      • Stolarova L
      • Zemankova P
      • et al.
      Multigene Panel Germline Testing of 1333 Czech Patients with Ovarian Cancer.
      ].
      According to the literature, the frequency of MMR-D/MSI in ovarian endometrioid carcinomas is lower compared to endometrioid endometrial carcinomas and occurs in about 12% of cases (range 0-22.9%) [

      Kim SR, Tone A, Kim RH, et al. Performance characteristics of screening strategies to identify Lynch syndrome in women with ovarian cancer. Cancer-Am Cancer Soc. 2020.

      ,
      • Rambau PF
      • Duggan MA
      • Ghatage P
      • et al.
      Significant frequency of MSH2/MSH6 abnormality in ovarian endometrioid carcinoma supports histotype-specific Lynch syndrome screening in ovarian carcinomas.
      ,
      • Segev Y
      • Pal T
      • Rosen B
      • et al.
      Risk Factors for Ovarian Cancers With and Without Microsatellite Instability.
      ,
      • Simila-Maarala J
      • Soovares P
      • Pasanen A
      • et al.
      TCGA molecular classification in endometriosis-associated ovarian carcinomas: Novel data on clear cell carcinoma.
      ,
      • Tanaka T
      • Takehara K
      • Yamashita N
      • et al.
      Frequency and clinical features of deficient mismatch repair in ovarian clear cell and endometrioid carcinoma.
      ,
      • Vierkoetter KR
      • Ayabe AR
      • VanDrunen M
      • et al.
      Lynch Syndrome in patients with clear cell and endometrioid cancers of the ovary.
      ,
      • Bennett JA
      • Pesci A
      • Morales-Oyarvide V
      • et al.
      Incidence of Mismatch Repair Protein Deficiency and Associated Clinicopathologic Features in a Cohort of 104 Ovarian Endometrioid Carcinomas.
      ,
      • Leskela S
      • Romero I
      • Cristobal E
      • et al.
      Mismatch Repair Deficiency in Ovarian Carcinoma Frequency, Causes, and Consequences.
      ]. The published literature concerning the frequency of MSI in non-endometrioid ovarian tumors is equivocal. In low-grade and high-grade serous carcinomas, the frequency of MSI is absent to very low with a range of 0-4.2% (average 0.3%) [
      • Rambau PF
      • Duggan MA
      • Ghatage P
      • et al.
      Significant frequency of MSH2/MSH6 abnormality in ovarian endometrioid carcinoma supports histotype-specific Lynch syndrome screening in ovarian carcinomas.
      ,
      • Lu FI
      • Gilks CB
      • Mulligan AM
      • et al.
      Prevalence of Loss of Expression of DNA Mismatch Repair Proteins in Primary Epithelial Ovarian Tumors.
      ,
      • Schmoeckel E
      • Hofmann S
      • Fromberger D
      • et al.
      Comprehensive analysis of PD-L1 expression, HER2 amplification, ALK/EML4 fusion, and mismatch repair deficiency as putative predictive and prognostic factors in ovarian carcinoma.
      ,
      • Malander S
      • Rambech E
      • Kristoffersson U
      • et al.
      The contribution of the hereditary nonpolyposis colorectal cancer syndrome to the development of ovarian cancer.
      ,
      • Xiao X
      • Dong DD
      • He WJ
      • et al.
      Mismatch repair deficiency is associated with MSI phenotype, increased tumor-infiltrating lymphocytes and PD-L1 expression in immune cells in ovarian cancer.
      ,
      • Yamashita H
      • Nakayama K
      • Ishikawa M
      • et al.
      Relationship between Microsatellite Instability, Immune Cells Infiltration, and Expression of Immune Checkpoint Molecules in Ovarian Carcinoma: Immunotherapeutic Strategies for the Future.
      ,
      • Crosbie EJ
      • Ryan NAJ
      • McVey RJ
      • et al.
      Assessment of mismatch repair deficiency in ovarian cancer.
      ,
      • Jensen KC
      • Mariappan MR
      • Putcha GV
      • et al.
      Microsatellite instability and mismatch repair protein defects in ovarian epithelial neoplasms in patients 50 years of age and younger.
      ]. The only exception is a study by Segev et al. on 504 serous carcinomas in which 13.7% of cases were found to be MSI-H using a PCR method [
      • Segev Y
      • Pal T
      • Rosen B
      • et al.
      Risk Factors for Ovarian Cancers With and Without Microsatellite Instability.
      ]. However, the results could be potentially influenced by using a PCR based method with 2 mononucleotides and 3 dinucleotide microsatellites. We think that using dinucleotide microsatellites could overestimate MSI in ovarian tumors, as it has been shown that a panel of five mononucleotide repeats is more suitable [
      • Suraweera N
      • Duval A
      • Reperant M
      • et al.
      Evaluation of tumor microsatellite instability using five quasimonomorphic mononucleotide repeats and pentaplex PCR.
      ]. Our results support the low frequency of MSI in ovarian serous tumors, low-grade and high-grade, as in our cohort of 208 tumors all were MSS. Only two other studies analyzed SBT (together 70 cases), all of them showing MSS, which is in concordance with the results of our study [
      • Lu FI
      • Gilks CB
      • Mulligan AM
      • et al.
      Prevalence of Loss of Expression of DNA Mismatch Repair Proteins in Primary Epithelial Ovarian Tumors.
      ,
      • Jensen KC
      • Mariappan MR
      • Putcha GV
      • et al.
      Microsatellite instability and mismatch repair protein defects in ovarian epithelial neoplasms in patients 50 years of age and younger.
      ]. Concerning MC, MSI has been shown in the range of 0-27.3% (average 6.7%) [
      • Rambau PF
      • Duggan MA
      • Ghatage P
      • et al.
      Significant frequency of MSH2/MSH6 abnormality in ovarian endometrioid carcinoma supports histotype-specific Lynch syndrome screening in ovarian carcinomas.
      ,
      • Tajima Y
      • Eguchi H
      • Chika N
      • et al.
      Prevalence and molecular characteristics of defective mismatch repair epithelial ovarian cancer in a Japanese hospital-based population.
      ,
      • Fraune C
      • Rosebrock J
      • Simon R
      • et al.
      High homogeneity of MMR deficiency in ovarian cancer.
      ,
      • Lu FI
      • Gilks CB
      • Mulligan AM
      • et al.
      Prevalence of Loss of Expression of DNA Mismatch Repair Proteins in Primary Epithelial Ovarian Tumors.
      ,
      • Schmoeckel E
      • Hofmann S
      • Fromberger D
      • et al.
      Comprehensive analysis of PD-L1 expression, HER2 amplification, ALK/EML4 fusion, and mismatch repair deficiency as putative predictive and prognostic factors in ovarian carcinoma.
      ,
      • Xiao X
      • Dong DD
      • He WJ
      • et al.
      Mismatch repair deficiency is associated with MSI phenotype, increased tumor-infiltrating lymphocytes and PD-L1 expression in immune cells in ovarian cancer.
      ,
      • Yamashita H
      • Nakayama K
      • Ishikawa M
      • et al.
      Relationship between Microsatellite Instability, Immune Cells Infiltration, and Expression of Immune Checkpoint Molecules in Ovarian Carcinoma: Immunotherapeutic Strategies for the Future.
      ,
      • Crosbie EJ
      • Ryan NAJ
      • McVey RJ
      • et al.
      Assessment of mismatch repair deficiency in ovarian cancer.
      ,
      • Dellas A
      • Puhl A
      • Schraml P
      • et al.
      Molecular and clinicopathological analysis of ovarian carcinomas with and without microsatellite instability.
      ]. In most studies, however, the frequency was 0%, with the highest frequency (27.3%) in the above-mentioned study by Segev et al. [
      • Segev Y
      • Pal T
      • Rosen B
      • et al.
      Risk Factors for Ovarian Cancers With and Without Microsatellite Instability.
      ]. One might speculate that in the Segev study also seromucinous tumors were included which are related to endometrioid tumors and were not studied by us. Our results support the finding that MSI is rare in ovarian MC, as all our mucinous tumors were MSS. For CCC, the published literature shows an MSI frequency of 0-25% (average 5.4%), which concurs with our finding of 7% [

      Kim SR, Tone A, Kim RH, et al. Performance characteristics of screening strategies to identify Lynch syndrome in women with ovarian cancer. Cancer-Am Cancer Soc. 2020.

      ,
      • Rambau PF
      • Duggan MA
      • Ghatage P
      • et al.
      Significant frequency of MSH2/MSH6 abnormality in ovarian endometrioid carcinoma supports histotype-specific Lynch syndrome screening in ovarian carcinomas.
      ,
      • Simila-Maarala J
      • Soovares P
      • Pasanen A
      • et al.
      TCGA molecular classification in endometriosis-associated ovarian carcinomas: Novel data on clear cell carcinoma.
      ,
      • Bennett JA
      • Morales-Oyarvide V
      • Campbell S
      • Longacre TA
      • Oliva E.
      Mismatch Repair Protein Expression in Clear Cell Carcinoma of the Ovary: Incidence and Morphologic Associations in 109 Cases.
      ,
      • Carter NJ
      • Marshall ML
      • Susswein LR
      • et al.
      Germline pathogenic variants identified in women with ovarian tumors.
      ,
      • Tanaka T
      • Takehara K
      • Yamashita N
      • et al.
      Frequency and clinical features of deficient mismatch repair in ovarian clear cell and endometrioid carcinoma.
      ,
      • Leskela S
      • Romero I
      • Cristobal E
      • et al.
      Mismatch Repair Deficiency in Ovarian Carcinoma Frequency, Causes, and Consequences.
      ,
      • Ge HJ
      • Xiao YX
      • Qin GQ
      • et al.
      Mismatch repair deficiency is associated with specific morphologic features and frequent loss of ARID1A expression in ovarian clear cell carcinoma.
      ,
      • Zhu J
      • Ke GH
      • Bi R
      • Wu XH.
      Clinicopathological and survival characteristic of mismatch repair status in ovarian clear cell carcinoma.
      ,
      • Lin SY
      • Hang JF
      • Lin YY
      • et al.
      Diffuse Intratumoral Stromal Inflammation in Ovarian Clear Cell Carcinoma is Associated With Loss of Mismatch Repair Protein and High PD-L1 Expression.
      ,
      • Parra-Herran C
      • Bassiouny D
      • Lerner-Ellis J
      • et al.
      Mismatch Repair Protein, and POLE Abnormalities in Ovarian Clear Cell Carcinoma: An Outcome-based Clinicopathologic Analysis.
      ,
      • Cai KQ
      • Albarracin C
      • Rosen D
      • et al.
      Microsatellite instability and alteration of the expression of hMLH1 and hMSH2 in ovarian clear cell carcinoma.
      ].
      Altogether, we detected 12 patients with class 4/5 mutation of any MMR genes, including 10 patients with CCC, one with LGSC and one with MBT. Mutations were somatic in 8 and germline in 4 patients, including two CCC, one LGSC and one MBT. In three patients with somatic MMR gene mutation (two MSS, one MMR-D) a concurrent POLE mutation was found. All these mutations in MMR genes were classified as pathogenic or likely pathogenic (class 4/5) with the exception of one case with the germline MSH6 variant NM_000179.2:c.1238G>C, p.(Trp413Ser). This variant is assessed as a variant of uncertain significance, according to the ClinVar database. However, our case with this variant was MMR-D/MSI-H, and along with the findings of one previous study, which detected this variant in one colorectal and one endometrial carcinoma associated with LS, this variant should be considered likely pathogenic (class 4) [
      • Kiyozumi Y
      • Matsubayashi H
      • Higashigawa S
      • et al.
      Role of Tumor Mutation Burden Analysis in Detecting Lynch Syndrome in Precision Medicine: Analysis of 2,501 Japanese Cancer Patients.
      ].
      Our study showed a 100% specificity of MSI testing for the detection of MMR gene mutation. The sensitivity was rather low, as we found MMR-D/MSI-H status in only 7/12 (58%) cases. The 5 discordant cases included a somatic mutation of MLH1 and concurrent PMS2 in one case and of MSH2 in the second case. Both occurred in POLE mutant tumors. All remaining discordant cases were MSH6 mutated, two of them (LGSC and MBT) with germline mutation. Our results are similar to others showing that MSI testing can be false negative in up to half of MSH6-mutated cases and could lead to false-negative screening tests for some LS patients with a MSH6 mutation [
      • Hampel H
      • Frankel W
      • Panescu J
      • et al.
      Screening for Lynch syndrome (hereditary nonpolyposis colorectal cancer) among endometrial cancer patients.
      ,
      • Latham A
      • Srinivasan P
      • Kemel Y
      • et al.
      Microsatellite Instability Is Associated With the Presence of Lynch Syndrome Pan-Cancer.
      ]. The only discrepant case between IHC and PCR testing in our study was a case with a somatic MSH6 mutation showing MMR-D, but a false negative result on PCR based testing.
      It has been demonstrated that an NGS approach for MSI testing can be used as an alternative to IHC or PCR based methods, but one should be aware that the spectrum of microsatellite loci can differ among tumor types and different loci can be preferentially mutated in different tumors [
      • Hanse RJ
      • Pritchard CC
      • Shendure J
      • Salipante SJ.
      Classification and characterization of microsatellite instability across 18 cancer types.
      ,
      • Cortes-Ciriano I
      • Lee S
      • Park WY
      • Kim TM
      • Park PJ.
      A molecular portrait of microsatellite instability across multiple cancers.
      ,
      • Long DR
      • Waalkes A
      • Panicker VP
      • Hause RJ
      • Salipante SJ.
      Identifying Optimal Loci for the Molecular Diagnosis of Microsatellite Instability.
      ]. All previous studies focusing on unstable microsatellite loci in different tumor types used data from The Cancer Genome Atlas (TCGA). Some of these studies focused on the description of unstable loci occurring in stomach, colorectal and endometrial carcinomas, but ovarian carcinomas were not included [
      • Hanse RJ
      • Pritchard CC
      • Shendure J
      • Salipante SJ.
      Classification and characterization of microsatellite instability across 18 cancer types.
      ,
      • Cortes-Ciriano I
      • Lee S
      • Park WY
      • Kim TM
      • Park PJ.
      A molecular portrait of microsatellite instability across multiple cancers.
      ]. One study analyzed 32 different tumor types. Although the results were promising, they failed to identify a set of unstable microsatellite loci with sufficient sensitivity and specificity, which could be used in ovarian and breast carcinoma [
      • Long DR
      • Waalkes A
      • Panicker VP
      • Hause RJ
      • Salipante SJ.
      Identifying Optimal Loci for the Molecular Diagnosis of Microsatellite Instability.
      ]. These results further suggested that microsatellite loci are tumor specific and tissue-specific loci should be used for optimal NGS MSI testing, which was confirmed by our study. Comparing three sets of microsatellites, our study showed that 17 microsatellites typical for colorectal carcinoma and microsatellites selected based on the results of our MSI testing by IHC and PCR) have high sensitivity and specificity and could be used in routine practice. However, we are aware of the limitations of our study. The definition of an optimal microsatellite set for ovarian carcinomas would require validation on a confirmatory sample set, but that was not the goal of this study.
      In conclusion, the results of our study show that MSI occurs in 7% of ovarian CCC. Absence of MSI in other tumor types suggests that occurrence of MSI in these tumors is unusual, which is concordant with the majority of the published literature. In our unselected population of CCC, 2% of patients had LS. Our results confirm that in addition to ovarian EC, also cases of ovarian CCC should be routinely tested for MSI as a part of LS screening and to identify candidate patients for immunotherapy. IHC testing of MMR protein expression seems to be superior to PCR based testing. NGS-MSI could be used in practice, but validation of microsatellite sets optimal for this testing in ovarian tumors is needed. Finally, our results suggest that germline mutation of MMR genes and possibly LS can rarely occur in ovarian tumors other than CCC and endometrioid carcinoma. However, some of these cases, particularly, with MSH6 mutation would be missed by all testing methods, including IHC, PCR, and NGS-MSI.

      DATA AVAILABILITY

      The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

      AUTHORS CONTRIBUTION

      PD performed study concept and design. NH collected and interpreted data and prepared manuscript, IS prepared the manuscript. All authors participated on material preparation, data collection and / or data analyses. RM provided statistical analysis. All authors read and approved the final paper.

      Brief commentary

      Background

      The significance of microsatellite testing in ovarian tumours is increasing. Testing is not only used as a triage for possible Lynch syndrome, but also to predict immunotherapy treatment response. But the optimal screening strategy to identify ovarian carcinomas patients associated with Lynch syndrome or microsatellite instable ovarian tumours has not been determined, yet.

      Translational Significance

      Precision assessing of the frequency of mismatch repair deficiency and/or microsatellite instability in non-endometrioid ovarian tumours. Comparison of different methodological approaches of testing and assessing of the optimal approach for next generation sequencing (NGS).

      Declaration of Competing Interest

      The authors have no conflicts of interest that are relevant to the content of this article to declare. All authors have read the journal's authorship agreement.

      ACKNOWLWDGEMENTS

      This work was supported by the Ministry of Health, Czech Republic (MH CZ DRO-VFN 64165 and AZV NV19-03-00007), by Charles University (Project UNCE204065), and by the European Regional Development Fund (EF16_013/0001674 and BBMRI.cz reg. no. LM2023033).
      The authors wish to thank Mgr. Zachary Harold Kane Kendall, B.A. (Institute for History of Medicine and Foreign Languages, First Faculty of Medicine, Charles University) for the English proofreading.

      References

        • Hampel H
        • Frankel W
        • Panescu J
        • et al.
        Screening for Lynch syndrome (hereditary nonpolyposis colorectal cancer) among endometrial cancer patients.
        Cancer Res. 2006; 66: 7810-7817
      1. Kim SR, Tone A, Kim RH, et al. Performance characteristics of screening strategies to identify Lynch syndrome in women with ovarian cancer. Cancer-Am Cancer Soc. 2020.

        • Lynch HT
        • Lynch PM
        • Lanspa SJ
        • et al.
        Review of the Lynch syndrome: history, molecular genetics, screening, differential diagnosis, and medicolegal ramifications.
        Clin Genet. 2009; 76: 1-18
        • Rambau PF
        • Duggan MA
        • Ghatage P
        • et al.
        Significant frequency of MSH2/MSH6 abnormality in ovarian endometrioid carcinoma supports histotype-specific Lynch syndrome screening in ovarian carcinomas.
        Histopathology. 2016; 69: 288-297
        • Ryan N
        • Wall J
        • Crosbie EJ
        • et al.
        Lynch syndrome screening in gynaecological cancers: results of an international survey with recommendations for uniform reporting terminology for mismatch repair immunohistochemistry results.
        Histopathology. 2019; 75: 813-824
        • Concin N
        • Matias-Guiu X
        • Vergote I
        • et al.
        ESGO/ESTRO/ESP guidelines for the management of patients with endometrial carcinoma.
        Int J Gynecol Cancer. 2021; 31: 12-39
        • Le DT
        • Uram JN
        • Wang H
        • et al.
        PD-1 Blockade in Tumors with Mismatch-Repair Deficiency.
        N Engl J Med. 2015; 372: 2509-2520
        • Bartley AN
        • Mills AM
        • Konnick E
        • et al.
        Mismatch Repair and Microsatellite Instability Testing for Immune Checkpoint Inhibitor Therapy: Guideline From the College of American Pathologists in Collaboration With the Association for Molecular Pathology and Fight Colorectal Cancer.
        Arch Pathol Lab Med. 2022; 146: 1194-1210
        • Latham A
        • Srinivasan P
        • Kemel Y
        • et al.
        Microsatellite Instability Is Associated With the Presence of Lynch Syndrome Pan-Cancer.
        J Clin Oncol. 2019; 37 (286-+)
        • Hechtman JF
        • Rana S
        • Middha S
        • et al.
        Retained mismatch repair protein expression occurs in approximately 6% of microsatellite instability-high cancers and is associated with missense mutations in mismatch repair genes.
        Mod Pathol. 2020; 33: 871-879
        • Adeleke S
        • Haslam A
        • Choy A
        • et al.
        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
        • Luchini C
        • Bibeau F
        • Ligtenberg MJL
        • et al.
        ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: a systematic review-based approach.
        Ann Oncol. 2019; 30: 1232-1243
        • Aiyer KTS
        • Doeleman T
        • Ryan NA
        • et al.
        Validity of a two-antibody testing algorithm for mismatch repair deficiency testing in cancer; a systematic literature review and meta-analysis.
        Mod Pathol. 2022; 35: 1775-1783
        • Gatius S
        • Velasco A
        • Varela M
        • et al.
        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
        • Velasco A
        • Tokat F
        • Bonde J
        • et al.
        Multi-center real-world comparison of the fully automated Idylla microsatellite instability assay with routine molecular methods and immunohistochemistry on formalin-fixed paraffin-embedded tissue of colorectal cancer.
        Virchows Arch. 2021; 478: 851-863
        • Farmkiss L
        • Hopkins I
        • Jones M.
        Idylla microsatellite instability assay versus mismatch repair immunohistochemistry: a retrospective comparison in gastric adenocarcinoma.
        J Clin Pathol. 2021; 74: 604-607
        • Samaison L
        • Uguen A.
        Idylla MSI test combined with immunohistochemistry is a valuable and cost effective strategy to search for microsatellite instable tumors of noncolorectal origin.
        Pathol Int. 2022; 72: 234-241
        • Pecriaux A
        • Favre L
        • Calderaro J
        • et al.
        Detection of microsatellite instability in a panel of solid tumours with the Idylla MSI Test using extracted DNA.
        J Clin Pathol. 2021; 74: 36-42
        • Tajima Y
        • Eguchi H
        • Chika N
        • et al.
        Prevalence and molecular characteristics of defective mismatch repair epithelial ovarian cancer in a Japanese hospital-based population.
        Jpn J Clin Oncol. 2018; 48: 728-735
        • Segev Y
        • Pal T
        • Rosen B
        • et al.
        Risk Factors for Ovarian Cancers With and Without Microsatellite Instability.
        International Journal of Gynecological Cancer. 2014; 24: 664-669
        • Simila-Maarala J
        • Soovares P
        • Pasanen A
        • et al.
        TCGA molecular classification in endometriosis-associated ovarian carcinomas: Novel data on clear cell carcinoma.
        Gynecol Oncol. 2022;
        • Fraune C
        • Rosebrock J
        • Simon R
        • et al.
        High homogeneity of MMR deficiency in ovarian cancer.
        Gynecol Oncol. 2020; 156: 669-675
        • Bennett JA
        • Morales-Oyarvide V
        • Campbell S
        • Longacre TA
        • Oliva E.
        Mismatch Repair Protein Expression in Clear Cell Carcinoma of the Ovary: Incidence and Morphologic Associations in 109 Cases.
        Am J Surg Pathol. 2016; 40: 656-663
        • Carter NJ
        • Marshall ML
        • Susswein LR
        • et al.
        Germline pathogenic variants identified in women with ovarian tumors.
        Gynecol Oncol. 2018; 151: 481-488
        • Lu FI
        • Gilks CB
        • Mulligan AM
        • et al.
        Prevalence of Loss of Expression of DNA Mismatch Repair Proteins in Primary Epithelial Ovarian Tumors.
        Int J Gynecol Pathol. 2012; 31: 524-531
        • Dundr P
        • Bartu M
        • Bosse T
        • et al.
        Primary Mucinous Tumors of the Ovary: An Interobserver Reproducibility and Detailed Molecular Study Reveals Significant Overlap Between Diagnostic Categories.
        Mod Pathol. 2023; 36100040
        • Dundr P
        • Bazalova B
        • Bartu M
        • et al.
        The cytokeratin 17 expression in primary ovarian tumors has diagnostic but not prognostic significance.
        Virchows Arch. 2022; 481: 201-212
        • Nemejcova K
        • Bartu MK
        • Michalkova R
        • et al.
        A comprehensive immunohistochemical analysis of IMP2 and IMP3 in 542 cases of ovarian tumors.
        Diagn Pathol. 2023; 18: 15
        • Umar A
        • Boland CR
        • Terdiman JP
        • et al.
        Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability.
        J Natl Cancer Inst. 2004; 96: 261-268
        • Shima K
        • Morikawa T
        • Yamauchi M
        • et al.
        TGFBR2 and BAX Mononucleotide Tract Mutations, Microsatellite Instability, and Prognosis in 1072 Colorectal Cancers.
        Plos One. 2011; 6
        • Zhao H
        • De Craene B
        • Sagaert X
        • et al.
        Association of a novel set of 7 homopolymer indels for detection of MSI with tumor mutation burden and total indel load in endometrial and colorectal cancers.
        J Clin Oncol. 2018; : 36
        • Hanse RJ
        • Pritchard CC
        • Shendure J
        • Salipante SJ.
        Classification and characterization of microsatellite instability across 18 cancer types.
        Nat Med. 2016; 22: 1342-1350
        • Chui MH
        • Ryan P
        • Radigan J
        • et al.
        The histomorphology of Lynch syndrome-associated ovarian carcinomas: toward a subtype-specific screening strategy.
        Am J Surg Pathol. 2014; 38: 1173-1181
        • Shah S
        • Cheung A
        • Kutka M
        • Sheriff M
        • Boussios S.
        Epithelial Ovarian Cancer: Providing Evidence of Predisposition Genes.
        Int J Environ Res Public Health. 2022; : 19
        • Bonadona V
        • Bonaiti B
        • Olschwang S
        • et al.
        Cancer risks associated with germline mutations in MLH1, MSH2, and MSH6 genes in Lynch syndrome.
        JAMA. 2011; 305: 2304-2310
        • Lu KH
        • Daniels M.
        Endometrial and ovarian cancer in women with Lynch syndrome: update in screening and prevention.
        Fam Cancer. 2013; 12: 273-277
        • Helder-Woolderink JM
        • Blok EA
        • Vasen HF
        • et al.
        Ovarian cancer in Lynch syndrome; a systematic review.
        Eur J Cancer. 2016; 55: 65-73
        • Lhotova K
        • Stolarova L
        • Zemankova P
        • et al.
        Multigene Panel Germline Testing of 1333 Czech Patients with Ovarian Cancer.
        Cancers (Basel). 2020; : 12
        • Tanaka T
        • Takehara K
        • Yamashita N
        • et al.
        Frequency and clinical features of deficient mismatch repair in ovarian clear cell and endometrioid carcinoma.
        J Gynecol Oncol. 2022; 33: e67
        • Vierkoetter KR
        • Ayabe AR
        • VanDrunen M
        • et al.
        Lynch Syndrome in patients with clear cell and endometrioid cancers of the ovary.
        Gynecol Oncol. 2014; 135: 81-84
        • Bennett JA
        • Pesci A
        • Morales-Oyarvide V
        • et al.
        Incidence of Mismatch Repair Protein Deficiency and Associated Clinicopathologic Features in a Cohort of 104 Ovarian Endometrioid Carcinomas.
        Am J Surg Pathol. 2019; 43: 235-243
        • Leskela S
        • Romero I
        • Cristobal E
        • et al.
        Mismatch Repair Deficiency in Ovarian Carcinoma Frequency, Causes, and Consequences.
        Am J Surg Pathol. 2020; 44: 649-656
        • Schmoeckel E
        • Hofmann S
        • Fromberger D
        • et al.
        Comprehensive analysis of PD-L1 expression, HER2 amplification, ALK/EML4 fusion, and mismatch repair deficiency as putative predictive and prognostic factors in ovarian carcinoma.
        Virchows Arch. 2019; 474: 599-608
        • Malander S
        • Rambech E
        • Kristoffersson U
        • et al.
        The contribution of the hereditary nonpolyposis colorectal cancer syndrome to the development of ovarian cancer.
        Gynecol Oncol. 2006; 101: 238-243
        • Xiao X
        • Dong DD
        • He WJ
        • et al.
        Mismatch repair deficiency is associated with MSI phenotype, increased tumor-infiltrating lymphocytes and PD-L1 expression in immune cells in ovarian cancer.
        Gynecol Oncol. 2018; 149: 146-154
        • Yamashita H
        • Nakayama K
        • Ishikawa M
        • et al.
        Relationship between Microsatellite Instability, Immune Cells Infiltration, and Expression of Immune Checkpoint Molecules in Ovarian Carcinoma: Immunotherapeutic Strategies for the Future.
        Int J Mol Sci. 2019; : 20
        • Crosbie EJ
        • Ryan NAJ
        • McVey RJ
        • et al.
        Assessment of mismatch repair deficiency in ovarian cancer.
        J Med Genet. 2021; 58: 687-691
        • Jensen KC
        • Mariappan MR
        • Putcha GV
        • et al.
        Microsatellite instability and mismatch repair protein defects in ovarian epithelial neoplasms in patients 50 years of age and younger.
        Am J Surg Pathol. 2008; 32: 1029-1037
        • Suraweera N
        • Duval A
        • Reperant M
        • et al.
        Evaluation of tumor microsatellite instability using five quasimonomorphic mononucleotide repeats and pentaplex PCR.
        Gastroenterology. 2002; 123: 1804-1811
        • Dellas A
        • Puhl A
        • Schraml P
        • et al.
        Molecular and clinicopathological analysis of ovarian carcinomas with and without microsatellite instability.
        Anticancer Res. 2004; 24: 361-369
        • Ge HJ
        • Xiao YX
        • Qin GQ
        • et al.
        Mismatch repair deficiency is associated with specific morphologic features and frequent loss of ARID1A expression in ovarian clear cell carcinoma.
        Diagnostic Pathology. 2021; : 16
        • Zhu J
        • Ke GH
        • Bi R
        • Wu XH.
        Clinicopathological and survival characteristic of mismatch repair status in ovarian clear cell carcinoma.
        J Surg Oncol. 2020; 122: 538-546
        • Lin SY
        • Hang JF
        • Lin YY
        • et al.
        Diffuse Intratumoral Stromal Inflammation in Ovarian Clear Cell Carcinoma is Associated With Loss of Mismatch Repair Protein and High PD-L1 Expression.
        Int J Gynecol Pathol. 2021; 40: 148-155
        • Parra-Herran C
        • Bassiouny D
        • Lerner-Ellis J
        • et al.
        Mismatch Repair Protein, and POLE Abnormalities in Ovarian Clear Cell Carcinoma: An Outcome-based Clinicopathologic Analysis.
        Am J Surg Pathol. 2019; 43: 1591-1599
        • Cai KQ
        • Albarracin C
        • Rosen D
        • et al.
        Microsatellite instability and alteration of the expression of hMLH1 and hMSH2 in ovarian clear cell carcinoma.
        Hum Pathol. 2004; 35: 552-559
        • Kiyozumi Y
        • Matsubayashi H
        • Higashigawa S
        • et al.
        Role of Tumor Mutation Burden Analysis in Detecting Lynch Syndrome in Precision Medicine: Analysis of 2,501 Japanese Cancer Patients.
        Cancer Epidemiol Biomarkers Prev. 2021; 30: 166-174
        • Cortes-Ciriano I
        • Lee S
        • Park WY
        • Kim TM
        • Park PJ.
        A molecular portrait of microsatellite instability across multiple cancers.
        Nat Commun. 2017; 8
        • Long DR
        • Waalkes A
        • Panicker VP
        • Hause RJ
        • Salipante SJ.
        Identifying Optimal Loci for the Molecular Diagnosis of Microsatellite Instability.
        Clin Chem. 2020; 66: 1310-1318