12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Clinical and Molecular Assessment of Patients with Lynch Syndrome and Sarcomas Underpinning the Association with MSH2 Germline Pathogenic Variants

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Lynch syndrome (LS) is a hereditary cancer-predisposing syndrome associated most frequently with epithelial tumors, particularly colorectal (CRC) and endometrial carcinomas (EC). The aim of this study was to investigate the relationship between sarcomas and LS by performing clinical and molecular characterization of patients presenting co-occurrence of sarcomas and tumors from the LS spectrum. We identified 27 patients diagnosed with CRC, EC, and other LS-associated tumors who had sarcomas in the same individuals or families. Germline genetic testing, mismatch repair (MMR) protein immunohistochemistry, microsatellite instability (MSI), and other molecular analyses were performed. Five LS patients presenting personal or family history of sarcomas were identified (3 MSH2 carriers and 2 MLH1), with 2 having Muir–Torre phenotypes. For two MSH2 carriers we confirmed the etiology of the sarcomas (one liposarcoma and two osteosarcomas) as LS-related, since the tumors were MSH2/MSH6-deficient, MSI-high, or presented a truncated MSH2 transcript. Additionally, we reviewed 43 previous reports of sarcomas in patients with LS, which revealed a high frequency (58%) of MSH2 alterations. In summary, sarcomas represent a rare clinical manifestation in patients with LS, especially in MSH2 carriers, and the analysis of tumor biological characteristics can be useful for definition of tumor etiology and novel therapeutic options.

          Related collections

          Most cited references44

          • Record: found
          • Abstract: found
          • Article: not found

          A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer.

          In December 1997, the National Cancer Institute sponsored "The International Workshop on Microsatellite Instability and RER Phenotypes in Cancer Detection and Familial Predisposition," to review and unify the field. The following recommendations were endorsed at the workshop. (a) The form of genomic instability associated with defective DNA mismatch repair in tumors is to be called microsatellite instability (MSI). (b) A panel of five microsatellites has been validated and is recommended as a reference panel for future research in the field. Tumors may be characterized on the basis of: high-frequency MSI (MSI-H), if two or more of the five markers show instability (i.e., have insertion/deletion mutations), and low-frequency MSI (MSI-L), if only one of the five markers shows instability. The distinction between microsatellite stable (MSS) and low frequency MSI (MSI-L) can only be accomplished if a greater number of markers is utilized. (c) A unique clinical and pathological phenotype is identified for the MSI-H tumors, which comprise approximately 15% of colorectal cancers, whereas MSI-L and MSS tumors appear to be phenotypically similar. MSI-H colorectal tumors are found predominantly in the proximal colon, have unique histopathological features, and are associated with a less aggressive clinical course than are stage-matched MSI-L or MSS tumors. Preclinical models suggest the possibility that these tumors may be resistant to the cytotoxicity induced by certain chemotherapeutic agents. The implications for MSI-L are not yet clear. (d) MSI can be measured in fresh or fixed tumor specimens equally well; microdissection of pathological specimens is recommended to enrich for neoplastic tissue; and normal tissue is required to document the presence of MSI. (e) The "Bethesda guidelines," which were developed in 1996 to assist in the selection of tumors for microsatellite analysis, are endorsed. (f) The spectrum of microsatellite alterations in noncolonic tumors was reviewed, and it was concluded that the above recommendations apply only to colorectal neoplasms. (g) A research agenda was recommended.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Pathogenic Germline Variants in 10,389 Adult Cancers

            We conducted the largest investigation of predisposition variants in cancer to date, discovering 853 pathogenic or likely pathogenic variants in 8% of 10,389 cases from 33 cancer types. Twenty-one genes showed single or cross-cancer associations, including novel associations of SDHA in melanoma and PALB2 in stomach adenocarcinoma. The 659 predisposition variants and 18 additional large deletions in tumor suppressors, including ATM, BRCA1, and NF1, showed low gene expression and frequent (43%) loss of heterozygosity or biallelic two-hit events. We also discovered 33 such variants in oncogenes, including missenses in MET, RET, and PTPN11 associated with high gene expression. We nominated 47 additional predisposition variants from prioritized VUSs supported by multiple evidences involving case-control frequency, loss of heterozygosity, expression effect, and co-localization with mutations and modified residues. Our integrative approach links rare predisposition variants to functional consequences, informing future guidelines of variant classification and germline genetic testing in cancer.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Cancer risk and survival in path_MMR carriers by gene and gender up to 75 years of age: a report from the Prospective Lynch Syndrome Database

              Background Most patients with path_MMR gene variants (Lynch syndrome (LS)) now survive both their first and subsequent cancers, resulting in a growing number of older patients with LS for whom limited information exists with respect to cancer risk and survival. Objective and design This observational, international, multicentre study aimed to determine prospectively observed incidences of cancers and survival in path_MMR carriers up to 75 years of age. Results 3119 patients were followed for a total of 24 475 years. Cumulative incidences at 75 years (risks) for colorectal cancer were 46%, 43% and 15% in path_MLH1, path_MSH2 and path_MSH6 carriers; for endometrial cancer 43%, 57% and 46%; for ovarian cancer 10%, 17% and 13%; for upper gastrointestinal (gastric, duodenal, bile duct or pancreatic) cancers 21%, 10% and 7%; for urinary tract cancers 8%, 25% and 11%; for prostate cancer 17%, 32% and 18%; and for brain tumours 1%, 5% and 1%, respectively. Ovarian cancer occurred mainly premenopausally. By contrast, upper gastrointestinal, urinary tract and prostate cancers occurred predominantly at older ages. Overall 5-year survival for prostate cancer was 100%, urinary bladder 93%, ureter 85%, duodenum 67%, stomach 61%, bile duct 29%, brain 22% and pancreas 0%. Path_PMS2 carriers had lower risk for cancer. Conclusion Carriers of different path_MMR variants exhibit distinct patterns of cancer risk and survival as they age. Risk estimates for counselling and planning of surveillance and treatment should be tailored to each patient’s age, gender and path_MMR variant. We have updated our open-access website www.lscarisk.org to facilitate this.
                Bookmark

                Author and article information

                Journal
                Cancers (Basel)
                Cancers (Basel)
                cancers
                Cancers
                MDPI
                2072-6694
                09 July 2020
                July 2020
                : 12
                : 7
                : 1848
                Affiliations
                [1 ]Genomics and Molecular Biology Group, International Research Center/CIPE, A.C.Camargo Cancer Center, São Paulo 01508-010, Brazil; nathalia.carvalho-aluno@ 123456accamargo.org.br (N.d.A.d.C.); bianca.niitsuma@ 123456accamargo.org.br (B.N.N.); dirce.carraro@ 123456accamargo.org.br (D.M.C.)
                [2 ]Oncogenetics Service, Hospital Erasto Gaertner, Curitiba 81520-060, Brazil; vanessakozak@ 123456hotmail.com (V.N.K.); casali.rocha@ 123456accamargo.org.br (J.C.C.-d.-R.)
                [3 ]Postgraduate Program in Genetics, Federal University of Parana, Curitiba 81530-000, Brazil
                [4 ]Anatomic Pathology Department, A.C.Camargo Cancer Center, São Paulo 01509-900, Brazil; felipe.costa@ 123456accamargo.org.br (F.D.C.); maripetaccia@ 123456gmail.com (M.P.d.M.)
                [5 ]Colorectal Cancer Department, A.C.Camargo Cancer Center, São Paulo 01509-900, Brazil; bruna.catin@ 123456accamargo.org.br (B.E.C.K.); samuel.aguiar.jr@ 123456gmail.com (S.A.J.)
                [6 ]Radiotherapy Department, A.C.Camargo Cancer Center, São Paulo 01509-900, Brazil; mlgobo@ 123456yahoo.com.br
                [7 ]Oncogenetics Department, A.C.Camargo Cancer Center, São Paulo 01509-900, Brazil; nirvana.formiga@ 123456accamargo.org.br
                [8 ]Faculdades Pequeno Principe, Curitiba 80230-020, Brazil; sahlua@ 123456hotmail.com (S.M.V.); edenirip@ 123456yahoo.com.br (E.I.P.)
                [9 ]Oncogenetics Department, Barretos Cancer Hospital, Barretos 14784-400, Brazil
                [10 ]Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-384, Brazil
                [11 ]Instituto de Pesquisa Pelé Pequeno Principe, Curitiba 80250-060, Brazil
                [12 ]National Institute of Science and Technology in Oncogenomics (INCITO), São Paulo 01508-010, Brazil
                Author notes
                [* ]Correspondence: giovana.torrezan@ 123456accamargo.org.br ; Tel.: +55-(11)-2189-5000 (ext. 2953)
                [†]

                These authors contributed equally.

                Author information
                https://orcid.org/0000-0003-3733-965X
                https://orcid.org/0000-0002-3293-5420
                https://orcid.org/0000-0003-3476-727X
                https://orcid.org/0000-0003-1904-2158
                https://orcid.org/0000-0002-1838-2153
                https://orcid.org/0000-0002-8659-5329
                Article
                cancers-12-01848
                10.3390/cancers12071848
                7408879
                32659967
                af14d8d1-caa1-4f4c-92d1-bd28ac04cdcf
                © 2020 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 24 May 2020
                : 07 June 2020
                Categories
                Article

                lynch syndrome,sarcoma,msh2 germline variant,hereditary cancer,mismatch repair deficiency

                Comments

                Comment on this article