1
|
Di Sciascio L, Orsatti A, Ambrosi F, Franchini E, Massari F, Mollica V, Bianchi FM, Colecchia M, De Leo A, Acosta AM, Lobo J, Fiorentino M, Ricci C. Characterization of testicular embryonic-type neuroectodermal tumor and embryonic-type neuroectodermal tissue admixed with mature neuro-glial tissue using a broad immunohistochemical panel. Virchows Arch 2024:10.1007/s00428-024-03911-8. [PMID: 39259293 DOI: 10.1007/s00428-024-03911-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/24/2024] [Accepted: 08/25/2024] [Indexed: 09/13/2024]
Abstract
Embryonic-type neuroectodermal tumor (ENT) is a somatic-type malignancy characterized by overgrowth of embryonic-type neuroectodermal tissue (EtNT). In germ cell tumors, EtNT is frequently intermingled with other components that may exhibit significant morphologic overlap [mature neuro-glial tissue (MNGT), nephroblastomatous tissues, and primitive endodermal-type glands]. Therefore, the quantification of EtNT (crucial for the diagnosis of ENT) can be challenging. In this study, we investigated the immunohistochemical profile of ENT, EtNT, and MNGT using a broad immunohistochemical panel. We found that SOX2 was the most sensitive marker for EtNT (100%), but it also stained MNGT (28.6%). GFAP and S100 were relatively sensitive (71.4%) and highly specific (GFAP 100%, S100 85.8%) for MNGT, whereas synaptophysin stained both. Combining our results with those of previous studies, we propose that a combination of SOX11, SOX2, GFAP, S100, AFP, villin, CDX2, PAX8, and nuclear WT1 may help to identify and quantify EtNT in germ cell tumors.
Collapse
Affiliation(s)
- Luisa Di Sciascio
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Agnese Orsatti
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Francesca Ambrosi
- DIAP-Dipartimento InterAziendale Di Anatomia Patologica Di Bologna, Pathology Unit, Maggiore Hospital-AUSL Bologna, Via Dell'Ospedale 2, Bologna, 40133, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Eugenia Franchini
- DIAP-Dipartimento InterAziendale Di Anatomia Patologica Di Bologna, Pathology Unit, Maggiore Hospital-AUSL Bologna, Via Dell'Ospedale 2, Bologna, 40133, Italy
| | - Francesco Massari
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | | | | | - Antonio De Leo
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Andres Martin Acosta
- Department of Pathology, Indiana University School of Medicine, Indianapolis, USA
| | - João Lobo
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), Porto, Portugal
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (GEBC CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (P.CCC), Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS-School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal
| | - Michelangelo Fiorentino
- DIAP-Dipartimento InterAziendale Di Anatomia Patologica Di Bologna, Pathology Unit, Maggiore Hospital-AUSL Bologna, Via Dell'Ospedale 2, Bologna, 40133, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Costantino Ricci
- DIAP-Dipartimento InterAziendale Di Anatomia Patologica Di Bologna, Pathology Unit, Maggiore Hospital-AUSL Bologna, Via Dell'Ospedale 2, Bologna, 40133, Italy.
| |
Collapse
|
2
|
Liu Y, Fan H, Kang X, Hao Y, Wang N, Zheng H, Li Y, Kang S. A rare germline BMP15 missense mutation causes hereditary ovarian immature teratoma in human. Proc Natl Acad Sci U S A 2024; 121:e2310409121. [PMID: 38427603 DOI: 10.1073/pnas.2310409121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 01/11/2024] [Indexed: 03/03/2024] Open
Abstract
Ovarian immature teratomas (OITs) are malignant tumors originating from the ovarian germ cells that mainly occur during the first 30 y of a female's life. Early age of onset strongly suggests the presence of susceptibility gene mutations for the disease yet to be discovered. Whole exon sequencing was used to screen pathogenic mutations from pedigrees with OITs. A rare missense germline mutation (C262T) in the first exon of the BMP15 gene was identified. In silico calculation suggested that the mutation could impair the formation of mature peptides. In vitro experiments on cell lines confirmed that the mutation caused an 84.7% reduction in the secretion of mature BMP15. Clinical samples from OIT patients also showed a similar pattern of decrease in the BMP15 expression. In the transgenic mouse model, the spontaneous parthenogenetic activation significantly increased in oocytes carrying the T allele. Remarkably, a mouse carrying the T allele developed the phenotype of OIT. Oocyte-specific RNA sequencing revealed that abnormal activation of the H-Ras/MAPK pathway might contribute to the development of OIT. BMP15 was identified as a pathogenic gene for OIT which improved our understanding of the etiology of OIT and provided a potential biomarker for genetic screening of this disorder.
Collapse
Affiliation(s)
- Yakun Liu
- Department of Gynecology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Hongwei Fan
- Department of Gynecology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Xi Kang
- Department of Surgery, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Yuntao Hao
- Department of Gynecology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Na Wang
- Department of Molecular Biology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Hui Zheng
- Nanjing Personal Oncology Biotechnology Co., Ltd., Nanjing, Jiangsu 211103, China
| | - Yan Li
- Department of Molecular Biology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Shan Kang
- Department of Gynecology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| |
Collapse
|
3
|
Travis LB, Feldman DR, Fung C, Poynter JN, Lockley M, Frazier AL. Adolescent and Young Adult Germ Cell Tumors: Epidemiology, Genomics, Treatment, and Survivorship. J Clin Oncol 2024; 42:696-706. [PMID: 37820296 PMCID: PMC11934052 DOI: 10.1200/jco.23.01099] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/05/2023] [Accepted: 08/11/2023] [Indexed: 10/13/2023] Open
Abstract
Innovations in the care of adolescent and young adult (AYA) germ cell tumors (GCTs) are needed for one of the most common AYA cancers for which treatment has not significantly changed for several decades. Testicular GCTs (TGCTs) are the most common cancers in 15- to 39-year-old men, and ovarian GCTs (OvGCTs) are the leading gynecologic malignancies in women younger than 25 years. Excellent outcomes, even in widely metastatic disease using cisplatin-based chemotherapy, can be achieved since Einhorn and Donohue's landmark 1977 study in TGCT. However, as the severity of accompanying late effects (ototoxicity, neurotoxicity, cardiovascular disease, second malignant neoplasms, nephrotoxicity, and others) has emerged, efforts to deintensity treatment and find alternatives to cisplatin have taken on new urgency. Current innovations include the collaborative design of clinical trials that accrue GCTs across all ages and both sexes, including adolescents (previously on pediatric trials), and OvGCT (previously on gynecologic-only trials). Joint trials accrue larger sample sizes at a faster rate and therefore evaluate new approaches more rapidly. These joint trials also allow for biospecimen collection to further probe GCT etiology and underlying mechanisms of tumor growth, thus providing new therapeutic options. This AYA approach has been fostered by The Malignant Germ Cell International Consortium, which includes over 115 GCT disease experts from pediatric, gynecologic, and genitourinary oncologies in 16 countries. Trials in development incorporate, to our knowledge, for the first time, molecular risk stratification and precision oncology approaches on the basis of specific GCT biology. This collaborative AYA approach pioneering successfully in GCT could serve as a model for impactful research for other AYA cancer types.
Collapse
Affiliation(s)
- Lois B. Travis
- Department of Medical Oncology, Indiana University, Indianapolis, IN
| | | | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| | - Jenny N. Poynter
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Michelle Lockley
- Centre for Cancer Genomics and Computational Biology; Barts Cancer Institute, Queen Mary University of London, UK
| | - A. Lindsay Frazier
- Dana Farber-Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| |
Collapse
|
4
|
Gatius S, Matias Guiu X, Davidson B. Molecular features for timely cancer diagnosis and treatment - tumors of the ovary, fallopian tube and endometrium. Virchows Arch 2024; 484:339-351. [PMID: 38099957 DOI: 10.1007/s00428-023-03710-7] [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: 07/07/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 03/19/2024]
Abstract
Gynecologic pathology has moved, within only a few years, from being a diagnostic area devoid of molecular testing into a diagnostic discipline in which such analyses are becoming routine. The direct relevance of molecular characterization to the choice of treatment of patients with carcinomas originating in both the uterus and adnexae makes it likely that such testing will only expand along with our understanding of the molecular make-up of these tumors. As a consequence, gynecologic pathologists have become an integral part of patient management, rather than lab personnel providing external services.In parallel, molecular testing is expanding as a tool for diagnosing rare tumors affecting these organs, including soft tissue tumors, sex cord-stromal tumors and germ cell tumors, as well as other rare entities. Increased knowledge in this area bears directly on the ability to diagnose these tumors in a reproducible manner, as well as recognize and consult on genetic diseases. Hopefully, despite the inherent difficulty in studying rare cancers, it will also translate into new therapeutic options for the malignant ones among these rare cancers.
Collapse
Affiliation(s)
- Sonia Gatius
- Department of Pathology, Hospital Universitari Arnau de Vilanova de Lleida, Universitat de Lleida, IRBLleida, CIBERONC, Lleida, Spain.
| | - Xavier Matias Guiu
- Department of Pathology, Hospital Universitari Arnau de Vilanova de Lleida, Universitat de Lleida, IRBLleida, CIBERONC, Lleida, Spain
- Department of Pathology, Hospital Universitari de Bellvitge, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Ben Davidson
- Department of Pathology, Oslo University Hospital Montebello, Norwegian Radium Hospital, N-0310, Oslo, Norway.
- University of Oslo, Faculty of Medicine, Institute of Clinical Medicine, N-0316, Oslo, Norway.
| |
Collapse
|
5
|
Christyani G, Carswell M, Qin S, Kim W. An Overview of Advances in Rare Cancer Diagnosis and Treatment. Int J Mol Sci 2024; 25:1201. [PMID: 38256274 PMCID: PMC10815984 DOI: 10.3390/ijms25021201] [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: 12/01/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer stands as the leading global cause of mortality, with rare cancer comprising 230 distinct subtypes characterized by infrequent incidence. Despite the inherent challenges in addressing the diagnosis and treatment of rare cancers due to their low occurrence rates, several biomedical breakthroughs have led to significant advancement in both areas. This review provides a comprehensive overview of state-of-the-art diagnostic techniques that encompass new-generation sequencing and multi-omics, coupled with the integration of artificial intelligence and machine learning, that have revolutionized rare cancer diagnosis. In addition, this review highlights the latest innovations in rare cancer therapeutic options, comprising immunotherapy, targeted therapy, transplantation, and drug combination therapy, that have undergone clinical trials and significantly contribute to the tumor remission and overall survival of rare cancer patients. In this review, we summarize recent breakthroughs and insights in the understanding of rare cancer pathophysiology, diagnosis, and therapeutic modalities, as well as the challenges faced in the development of rare cancer diagnosis data interpretation and drug development.
Collapse
Affiliation(s)
| | | | - Sisi Qin
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea; (G.C.); (M.C.)
| | - Wootae Kim
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea; (G.C.); (M.C.)
| |
Collapse
|
6
|
Alna'irat M, McCluggage WG, Al-Hussaini M. Ovarian Immature Teratoma With Nodal Gliomatosis: A Case Report and Literature Review. Int J Gynecol Pathol 2023; 42:627-631. [PMID: 36867505 DOI: 10.1097/pgp.0000000000000938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Gliomatosis involving lymph nodes (nodal gliomatosis) is rarely encountered in association with an ovarian teratoma, with 12 cases previously reported. We report this rare occurrence in a 23-yr-old female with an ovarian immature teratoma. The ovary contained a grade 3 immature teratoma, with immature neuroepithelium. A subcapsular liver mass contained metastatic immature teratoma with neuroepithelium. The omentum and peritoneum contained mature glial tissue, consistent with gliomatosis peritonei with no evidence of immature elements. One pelvic lymph node contained multiple nodules of mature glial tissue, diffusely positive for glial fibrillary acidic protein, in keeping with nodal gliomatosis. In reporting this case, we review prior reports of nodal gliomatosis.
Collapse
|
7
|
Laothamatas I, Fang E, Lee J, Abbas SMH, Ding J, Kagen A, Zakashansky K, Taouli B, Kamath A, King MJ. Benign and Malignant Ovarian Teratomas: Multimodality Imaging Findings With Histopathologic Correlation. J Comput Assist Tomogr 2023; 47:882-889. [PMID: 37948362 DOI: 10.1097/rct.0000000000001509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2023]
Abstract
ABSTRACT The purpose of this article is to provide a comprehensive review of the imaging findings along with histopathologic correlation of mature (benign) teratomas and malignant ovarian teratomas, which include both immature teratomas and malignant degeneration of mature teratomas. The radiologist's ability to provide an accurate diagnosis plays an essential role in guiding the interdisciplinary care of patients with malignant teratomas and improving their outcomes.
Collapse
Affiliation(s)
- Indira Laothamatas
- From the Department of Diagnostic, Molecular and Interventional Radiology
| | - Eric Fang
- From the Department of Diagnostic, Molecular and Interventional Radiology
| | - Justine Lee
- From the Department of Diagnostic, Molecular and Interventional Radiology
| | - Syed Muhammad Hashim Abbas
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Juan Ding
- Parkview Regional Medical Center, Fort Wayne, IN
| | - Alexander Kagen
- From the Department of Diagnostic, Molecular and Interventional Radiology
| | - Konstantin Zakashansky
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Amita Kamath
- From the Department of Diagnostic, Molecular and Interventional Radiology
| | - Michael J King
- From the Department of Diagnostic, Molecular and Interventional Radiology
| |
Collapse
|
8
|
Trozzi R, Rosati A, Panico C, Perisiano C, Santoro A, Fagotti A. Extra-cranial meningioma associated with relapse of immature ovarian teratoma. Int J Gynecol Cancer 2023; 33:1649-1654. [PMID: 37783480 DOI: 10.1136/ijgc-2023-004718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023] Open
Affiliation(s)
- Rita Trozzi
- Dipartimento di Scienze della salute della donna, del bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Universita Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Rosati
- Dipartimento di Scienze della salute della donna, del bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Universita Cattolica del Sacro Cuore, Rome, Italy
| | - Camilla Panico
- U.O.C. Radiologia Addomino-pelvica. Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo Perisiano
- UOC Ortopedia e Traumatologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Angela Santoro
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Anna Fagotti
- Dipartimento di Scienze della salute della donna, del bambino e di sanità pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Universita Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
9
|
Doe-Tetteh SA, Camp SY, Reales D, Crowdis J, Noronha AM, Wolff B, Alano T, Galle J, Duygu Selcuklu S, Viale A, Socci ND, Liu YL, Tew WP, Aghajanian C, Ladanyi M, He MX, AlDubayan SH, Mazor RD, Shpilberg O, Hershkovitz-Rokah O, Riancho JA, Hernandez JL, Gonzalez-Vela MC, Buthorn JJ, Wilson M, Webber AE, Yabe M, Petrova-Drus K, Rosenblum M, Durham BH, Abdel-Wahab O, Berger MF, Donoghue MT, Kung AL, Bender JG, Shukla NN, Funt SA, Dogan A, Soslow RA, Al-Ahmadie H, Feldman DR, Van Allen EM, Diamond EL, Solit DB. Overcoming Barriers to Tumor Genomic Profiling through Direct-to-Patient Outreach. Clin Cancer Res 2023; 29:2445-2455. [PMID: 36862133 PMCID: PMC10330105 DOI: 10.1158/1078-0432.ccr-22-3247] [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: 10/25/2022] [Revised: 01/05/2023] [Accepted: 02/28/2023] [Indexed: 03/03/2023]
Abstract
PURPOSE To overcome barriers to genomic testing for patients with rare cancers, we initiated a program to offer free clinical tumor genomic testing worldwide to patients with select rare cancer subtypes. EXPERIMENTAL DESIGN Patients were recruited through social media outreach and engagement with disease-specific advocacy groups, with a focus on patients with histiocytosis, germ cell tumors (GCT), and pediatric cancers. Tumors were analyzed using the MSK-IMPACT next-generation sequencing assay with the return of results to patients and their local physicians. Whole-exome recapture was performed for female patients with GCTs to define the genomic landscape of this rare cancer subtype. RESULTS A total of 333 patients were enrolled, and tumor tissue was received for 288 (86.4%), with 250 (86.8%) having tumor DNA of sufficient quality for MSK-IMPACT testing. Eighteen patients with histiocytosis have received genomically guided therapy to date, of whom 17 (94%) have had clinical benefit with a mean treatment duration of 21.7 months (range, 6-40+). Whole-exome sequencing of ovarian GCTs identified a subset with haploid genotypes, a phenotype rarely observed in other cancer types. Actionable genomic alterations were rare in ovarian GCT (28%); however, 2 patients with ovarian GCTs with squamous transformation had high tumor mutational burden, one of whom had a complete response to pembrolizumab. CONCLUSIONS Direct-to-patient outreach can facilitate the assembly of cohorts of rare cancers of sufficient size to define their genomic landscape. By profiling tumors in a clinical laboratory, results could be reported to patients and their local physicians to guide treatment. See related commentary by Desai and Subbiah, p. 2339.
Collapse
Affiliation(s)
- Seyram A. Doe-Tetteh
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Sabrina Y. Camp
- Department of Medical Oncology, Dana Farber Cancer Institute
- Cancer Program, Broad Institute of MIT and Harvard
| | - Dalicia Reales
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Jett Crowdis
- Department of Medical Oncology, Dana Farber Cancer Institute
- Cancer Program, Broad Institute of MIT and Harvard
| | - Anne Marie Noronha
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Bernadette Wolff
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Department of Nursing, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Tina Alano
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Department of Nursing, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Jesse Galle
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - S. Duygu Selcuklu
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Agnes Viale
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Nicholas D. Socci
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Bioinformatics Core, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Ying L. Liu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - William P. Tew
- Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Carol Aghajanian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Meng Xiao He
- Department of Medical Oncology, Dana Farber Cancer Institute
- Cancer Program, Broad Institute of MIT and Harvard
- Harvard Graduate Program in Biophysics, Boston, MA, 02115, USA
| | - Saud H. AlDubayan
- Department of Medical Oncology, Dana Farber Cancer Institute
- Cancer Program, Broad Institute of MIT and Harvard
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Roei David Mazor
- Clinic of Histiocytic Neoplasms, Institute of Hematology, Assuta Medical Center, Tel Aviv, Israel
| | - Ofer Shpilberg
- Clinic of Histiocytic Neoplasms, Institute of Hematology, Assuta Medical Center, Tel Aviv, Israel
- Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Oshrat Hershkovitz-Rokah
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel, Israel
- Translational Research Lab, Assuta Medical Center, Tel-Aviv, Israel
| | - Jose A. Riancho
- Department of Internal Medicine, Hospital U.M. Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Jose L. Hernandez
- Department of Internal Medicine, Hospital U.M. Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - M. Carmen Gonzalez-Vela
- Department of Pathology, Hospital U.M. Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Justin J. Buthorn
- Department of Neurology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Manda Wilson
- Bioinformatics Core, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Amy E. Webber
- Bioinformatics Core, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Mariko Yabe
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Kseniya Petrova-Drus
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Marc Rosenblum
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Benjamin H. Durham
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael F. Berger
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Mark T.A. Donoghue
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Andrew L. Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Julia Glade Bender
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Neerav N. Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Samuel A. Funt
- Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| | - Ahmet Dogan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Robert A. Soslow
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Hikmat Al-Ahmadie
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Darren R. Feldman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| | - Eliezer M. Van Allen
- Department of Medical Oncology, Dana Farber Cancer Institute
- Cancer Program, Broad Institute of MIT and Harvard
| | - Eli L. Diamond
- Department of Neurology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| | - David B. Solit
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
10
|
Pinto MT, Eiras Martins G, Vieira AGS, Galvão JMS, de Pádua Souza C, Macedo CRPD, Lopes LF. Molecular Biology of Pediatric and Adult Ovarian Germ Cell Tumors: A Review. Cancers (Basel) 2023; 15:cancers15112990. [PMID: 37296950 DOI: 10.3390/cancers15112990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 06/12/2023] Open
Abstract
Ovarian germ cell tumors (OGCTs) are rare in adults; indeed, they occur predominantly in children, adolescents, and young adults, and they account for approximately 11% of cancer diagnoses in these groups. Because OGCTs are rare tumors, our current understanding of them is sparse; this is because few studies have investigated the molecular basis of pediatric and adult cancers. Here, we review the etiopathogenesis of OGCTs in children and adults, and we address the molecular landscape of these tumors, including integrated genomic analysis, microRNAs, DNA methylation, the molecular implications of treatment resistance, and the development of in vitro and in vivo models. An elucidation of potential molecular alterations may provide a novel field for understanding the pathogenesis, tumorigenesis, diagnostic markers, and genetic peculiarity of the rarity and complexity of OGCTs.
Collapse
Affiliation(s)
| | - Gisele Eiras Martins
- Brazilian Childhood Germ Cell Tumor Study Group, The Brazilian Pediatric Oncology Society (SOBOPE), Barretos 14784400, Brazil
- Children's Cancer Hospital from Hospital de Amor, Barretos 14784400, Brazil
| | - Ana Glenda Santarosa Vieira
- Brazilian Childhood Germ Cell Tumor Study Group, The Brazilian Pediatric Oncology Society (SOBOPE), Barretos 14784400, Brazil
- Children's Cancer Hospital from Hospital de Amor, Barretos 14784400, Brazil
| | | | | | - Carla Renata Pacheco Donato Macedo
- Brazilian Childhood Germ Cell Tumor Study Group, The Brazilian Pediatric Oncology Society (SOBOPE), Barretos 14784400, Brazil
- Pediatric Oncology Department, IOP/GRAACC/Federal University of Sao Paulo, Sao Paulo 04038001, Brazil
| | - Luiz Fernando Lopes
- Brazilian Childhood Germ Cell Tumor Study Group, The Brazilian Pediatric Oncology Society (SOBOPE), Barretos 14784400, Brazil
- Children's Cancer Hospital from Hospital de Amor, Barretos 14784400, Brazil
| |
Collapse
|
11
|
Moraru L, Mitranovici MI, Chiorean DM, Coroș M, Moraru R, Oală IE, Turdean SG. Immature Teratoma: Diagnosis and Management-A Review of the Literature. Diagnostics (Basel) 2023; 13:diagnostics13091516. [PMID: 37174909 PMCID: PMC10177811 DOI: 10.3390/diagnostics13091516] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/05/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
An immature teratoma is a germinal malignant tumor composed of three germ cell layers, occurring more frequently in young women. It is the second most frequent among the malignant germinal tumors after dysgerminoma, and it is the only neoplasm with germ cells that are histologically graded. Even if we do not have a consensus regarding its therapeutical management, it has a good prognosis, with an excellent overall survival rate and good fertility preservation. More studies are needed regarding the necessity of adjuvant chemotherapy in pediatric oncology, and because of chemotherapy's long-term adverse effects, surveillance or a targeted treatment is preferred, but the main therapy is fertility-sparing surgery. Special attention should be given to the genetic mapping of the histological pieces for patient risk stratification due to its value in prognosis and future treatment.
Collapse
Affiliation(s)
- Liviu Moraru
- Department of Anatomy, "George Emil Palade" University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania
| | | | - Diana Maria Chiorean
- Department of Pathology, County Clinical Hospital of Targu Mures, 540072 Targu Mures, Romania
| | - Marius Coroș
- Department of Surgery, "George Emil Palade" University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania
| | - Raluca Moraru
- Faculty of Medicine, "George Emil Palade" University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania
| | - Ioan Emilian Oală
- Department of Obstetrics and Gynecology, Emergency County Hospital Hunedoara, 331057 Hunedoara, Romania
| | - Sabin Gligore Turdean
- Department of Pathology, County Clinical Hospital of Targu Mures, 540072 Targu Mures, Romania
- Department of Pathology, "George Emil Palade" University of Medicine, Pharmacy, Sciences and Technology, 540142 Targu Mures, Romania
| |
Collapse
|
12
|
Kato N, Kamataki A, Kurotaki H. Mature and immature ovarian teratomas share methylation profiles of imprinted genes: a MS-MLPA analysis. Virchows Arch 2023; 482:561-566. [PMID: 36637485 DOI: 10.1007/s00428-023-03491-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/25/2022] [Accepted: 01/09/2023] [Indexed: 01/14/2023]
Abstract
Immature teratomas are a subset of ovarian teratomas, and the pathogenic relationship between mature and immature ovarian teratomas is unclear. Mature ovarian teratomas are parthenogenetic tumors that arise from a single oocyte/ovum, whereas the origin of immature ovarian teratomas has not been extensively investigated. Since parthenogenetic tumors contain only maternal genomes, genome imprinting in these tumors usually follows a maternal pattern. DNA methylation is among the most important mechanisms of genome imprinting. Therefore, we analyzed the methylation profile of imprinted genes by performing methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA) of 25 imprinting control regions (ICRs) in 10 imprinted genes/gene clusters from formalin-fixed, paraffin-embedded samples obtained from 4 immature ovarian teratomas, 8 mature ovarian teratomas, and 4 ovarian yolk sac tumors (YSTs). Both the immature and mature components showed similar methylation levels in each ICR in immature teratomas. Overall, immature ovarian teratomas showed maternal methylation patterns of imprinted genes in concordance with their parthenogenetic origin. However, they also showed aberrant methylation levels in a few imprinted genes, suggesting that genome imprinting in immature teratomas may partially differ from that in mature teratomas. Microscopic foci of YST were seen in one immature teratoma; the YST component also showed a maternal methylation pattern, unlike the pure YSTs that showed irregular patterns. Thus, teratoma-associated YST and pure YST may have different pathogenic mechanisms.
Collapse
Affiliation(s)
- Noriko Kato
- Department of Anatomic Pathology, Hirosaki University School of Medicine and Hospital, 53 Honcho, Hirosaki, 036-8563, Japan.
| | - Akihisa Kamataki
- Department of Anatomic Pathology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Hidekachi Kurotaki
- Department of Pathology, Aomori Prefectural Central Hospital, Aomori, Japan
| |
Collapse
|
13
|
Cao M, Deng Y, Deng Y, Wu J, Yang C, Wang Z, Hou Q, Fu H, Ren Z, Xia X, Li Y, Wang W, Xu H, Liao X, Shu Y. Characterization of immature ovarian teratomas through single-cell transcriptome. Front Immunol 2023; 14:1131814. [PMID: 36936909 PMCID: PMC10020330 DOI: 10.3389/fimmu.2023.1131814] [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: 12/26/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Immature ovarian teratomas are a type of malignant germ cell tumor composed of complicated cell types and are characterized by pathological features of immature neuroectodermal tubules/rosettes. However, there is a lack of understanding of patient-derived immature ovarian teratomas (PDT) at the single cell level. Moreover, whether stem cell lines derived from immature teratomas (CDT) can be used as models for research on PDT remains to be elucidated. Methods Single-cell RNA sequencing (scRNA-seq) and subsequent bioinformatic analysis was performed on three patient-derived immature ovarian teratomas (PDT) samples to reveal the heterogeneity, evolution trajectory, and cell communication within the tumor microenvironment of PDT. Validations were conducted in additional seven samples through multiplex immunofluorescence. Result A total of qualified 22,153 cells were obtained and divided into 28 clusters, which can match to the scRNA-seq annotation of CDT as well as human fetal Cell Atlas, but with higher heterogeneity and more prolific cell-cell crosstalk. Radial glia cells (tagged by SOX2) and immature neuron (tagged by DCX) exhibited mutually exclusive expression and differentiated along distinct evolutionary trajectory from cycling neural progenitors. Proportions of these neuroectodermal cell subtypes may play important roles in PDT through contributing to the internal heterogeneity of PDTs. Moreover, the immune cells in PDTs were infiltrated rather than teratoma-derived, with more abundant macrophage in immature neuron than those in radial glia cells, and the infiltrated macrophage subtypes (i.e., M1 and M2) were significantly correlated to clinical grade. Overall, suppressed evolution process and transcriptome regulation in neuroectodermal cells, reduced cell-cell crosstalk, higher M1/M2 proportion ratio, and enhanced T cell effects in tumor microenvironment are enriched in patients with favorable prognosis. Discussion This study provides a comprehensive profile of PDT at the single cell level, shedding light on the heterogeneity and evolution of neuroectodermal cells within PDTs and the role of immune cells within the tumor microenvironment. Also, our findings highlight the potential usage of CDTs as a model for research on PDT.
Collapse
Affiliation(s)
- Minyuan Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yun Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yiqi Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chongyi Yang
- College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Zijun Wang
- College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Qianqian Hou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huancheng Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhixiang Ren
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuyang Xia
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yue Li
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Wang
- Department of Pathology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Heng Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yang Shu, ; Xin Liao, ; Heng Xu,
| | - Xin Liao
- Department of Pathology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yang Shu, ; Xin Liao, ; Heng Xu,
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yang Shu, ; Xin Liao, ; Heng Xu,
| |
Collapse
|
14
|
Guo F, Liu Y, Lu J, Wu Z, Zhu X. Human chorionic gonadotropin elevation in gliomatosis peritonei complicated with immature teratoma: A case report and review of the literature. Medicine (Baltimore) 2022; 101:e31305. [PMID: 36316907 PMCID: PMC9622604 DOI: 10.1097/md.0000000000031305] [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] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Gliomatosis peritonei (GP) refers to the implantation of glial tissue on the visceral and parietal peritoneal surface, often associated with immature teratoma. It is a rare condition and the pathogenesis is not fully understood. In addition, the indistinguishable radiological appearance of immature and mature teratomas, and limited pathology samples make an accurate diagnosis difficult in most cases. More importantly, patients are also at risk of recurrence after surgery. This report aims to describe the process of diagnosis and treatment of GP with immature teratoma. PATIENT CONCERNS The patient, a 38-year-old woman presented with GP complicated with immature teratoma after laparoscopic ovarian cyst excision. DIAGNOSES On physical examination, a 15 cm-pelvic mass, with poor mobility, was palpated. And tumor marker demonstrated a moderate increase in α-fetoprotein and carbohydrate antigen 125. We suspected malignancy according to the comprehensive preoperative evaluation, the postoperative pathology revealed an immature teratoma of the left ovary and complicated with gliomatosis peritonei. Three months after the second surgery, possible recurrence of immature teratoma was considered and the patient underwent the third laparotomy. But the postoperative pathology indicated mature teratoma and mature glial components in the pelvic lesions. INTERVENTIONS AND OUTCOME The patient underwent 2 more surgical resections after the initial resection and 3 cycles of bleomycin, etoposide, and cisplatin regimen chemotherapy. She was regularly followed up in the outpatient after surgery, and no recurrence has been reported in the pelvic cavity till date. LESSON The case illuminated that the primary diagnosis of GP complicated with immature teratoma is critical but highly challenging for both gynecologists and pathologists and more attention should be paid to "GP complicated with immature cystic teratoma" patients to avoid inappropriate treatment.
Collapse
Affiliation(s)
- Fei Guo
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yukai Liu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jiaqi Lu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Zhiyong Wu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiaoyong Zhu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- * Correspondence: Xiaoyong Zhu, Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China (e-mail: )
| |
Collapse
|
15
|
Takahashi N, Hatakeyama K, Nagashima T, Ohshima K, Urakami K, Yamaguchi K, Hirashima Y. Characterization of rare histological subtypes of ovarian cancer based on molecular profiling. Cancer Med 2022; 12:387-395. [PMID: 35676859 PMCID: PMC9844652 DOI: 10.1002/cam4.4927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/08/2022] [Accepted: 05/24/2022] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVE Pan-cancer analysis across The Cancer Genome Atlas has revealed the molecular profiles of major types of carcinomas. High-grade serous carcinomas (HGSCs) have been characterized; however, in ovarian cancer, the profile of carcinoma with minor histopathological changes remains unclear. This study aimed to perform the molecular profiling of rare malignant ovarian tumors, including non-epithelial tumors (NETs; germ cell tumors and sex cord tumors) and clear cell carcinoma (CCC), to determine how they differ from the major HGSCs. METHODS Sixty-nine malignant ovarian tumors surgically resected at the Shizuoka Cancer Center between January 2014 and March 2019 were classified based on their histopathological types. The germline and somatic mutations in these carcinomas, including NETs, were determined using next-generation sequencing. Gene expression analysis was performed to investigate the major pathways of drug resistance, which is a characteristic of CCC. RESULTS NETs harbored copy-neutral loss of heterozygosity, accompanied by a high homologous recombination deficiency score; germline mutations of PALB2 and BARD1 were identified in two patients with NET. In chemoresistant CCC, the epithelial-mesenchymal transition pathway was activated regardless of ABC transporter expression. CONCLUSION This study revealed some genomic characteristics of rare malignant ovarian tumors, including NETs and CCC.
Collapse
Affiliation(s)
| | - Keiichi Hatakeyama
- Medical Genetics DivisionShizuoka Cancer Center Research InstituteSunto‐gunShizuokaJapan
| | - Takeshi Nagashima
- Cancer Diagnostics Research DivisionShizuoka Cancer Center Research InstituteSunto‐gunShizuokaJapan,SRL Inc.Shinjuku‐kuTokyoJapan
| | - Keiichi Ohshima
- Medical Genetics DivisionShizuoka Cancer Center Research InstituteSunto‐gunShizuokaJapan
| | - Kenichi Urakami
- Cancer Diagnostics Research DivisionShizuoka Cancer Center Research InstituteSunto‐gunShizuokaJapan
| | | | | |
Collapse
|
16
|
Edwards MM, Zuccaro MV, Sagi I, Ding Q, Vershkov D, Benvenisty N, Egli D, Koren A. Delayed DNA replication in haploid human embryonic stem cells. Genome Res 2021; 31:2155-2169. [PMID: 34810218 PMCID: PMC8647822 DOI: 10.1101/gr.275953.121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/20/2021] [Indexed: 11/25/2022]
Abstract
Haploid human embryonic stem cells (ESCs) provide a powerful genetic system but diploidize at high rates. We hypothesized that diploidization results from aberrant DNA replication. To test this, we profiled DNA replication timing in isogenic haploid and diploid ESCs. The greatest difference was the earlier replication of the X Chromosome in haploids, consistent with the lack of X-Chromosome inactivation. We also identified 21 autosomal regions that had delayed replication in haploids, extending beyond the normal S phase and into G2/M. Haploid-delays comprised a unique set of quiescent genomic regions that are also underreplicated in polyploid placental cells. The same delays were observed in female ESCs with two active X Chromosomes, suggesting that increased X-Chromosome dosage may cause delayed autosomal replication. We propose that incomplete replication at the onset of mitosis could prevent cell division and result in re-entry into the cell cycle and whole genome duplication.
Collapse
Affiliation(s)
- Matthew M Edwards
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| | - Michael V Zuccaro
- Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, New York 10032, USA
- Columbia University Stem Cell Initiative, New York, New York 10032, USA
| | - Ido Sagi
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Qiliang Ding
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| | - Dan Vershkov
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Dieter Egli
- Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University, New York, New York 10032, USA
- Columbia University Stem Cell Initiative, New York, New York 10032, USA
| | - Amnon Koren
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
17
|
Jung SH, Park HC, Choi YJ, Song SY, Chung YJ, Lee SH. Molecular genetic evidence supporting diverse histogenic origins of germ cell tumors. J Pathol 2021; 256:38-49. [PMID: 34561860 DOI: 10.1002/path.5799] [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: 05/27/2021] [Revised: 08/24/2021] [Accepted: 09/20/2021] [Indexed: 11/10/2022]
Abstract
Germ cell tumors (GCTs) originate during the histogenesis of primordial germ cells to mature gametes. Previous studies identified five histogenic mechanisms in ovarian mature teratomas (type I: failure of meiosis I; type II: failure of meiosis II; type III: duplication of the genome of a mature gamete; type IV: no meiosis; and type V: fusion of two different ova), but those of other GCTs remain elusive. In this study, we analyzed 84 GCTs of various pathologic types to identify the histogenesis using single-nucleotide polymorphism array by analyzing copy-neutral loss of heterozygosity (CN-LOH) and copy number alterations (CNAs). We detected types I and II in ovarian teratomas, type III in ovarian teratomas and yolk sac tumors (YSTs), and type IV in all GCT types. The GCTs with multiple-type histogenesis (I-IV) (ovarian mature/immature teratomas and YST) show meiotic CN-LOH with scant CNAs. Type IV-only GCTs are either with mitotic CN-LOH and abundant CNAs (seminoma, dysgerminoma, testicular mixed GCTs) or with scant CNAs and no CN-LOH (pediatric testicular and mediastinal teratomas). The development sequences of CN-LOH and CNA are different between the multiple type (I-IV) GCTs and type IV-only GCTs. We analyzed two different histologic areas in eight GCTs (one mature teratoma with a mucin-secreting adenoma, two immature teratomas, and five mixed GCTs). We found that GCTs (mature teratoma, immature teratoma, and mixed GCT) showed different genomic alterations between histologic areas, suggesting that genomic differences within a GCT could accompany histologic differentiation. Of note, we found evidence for collision tumors in a mixed GCT. Our data indicate that GCTs may have various histogenesis and intratumoral genomic differences, which might provide important information for the identification of GCTs, especially for those with different histologic areas. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Seung-Hyun Jung
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Precision Medicine Research Center/IRCGP, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyeon-Chun Park
- Department of Precision Medicine Research Center/IRCGP, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Youn Jin Choi
- Department of Obstetrics/Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang Yong Song
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yeun-Jun Chung
- Department of Precision Medicine Research Center/IRCGP, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sug Hyung Lee
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
18
|
Ono R, Ueno H, Yoshida K, Takahashi S, Yoshihara H, Nozaki T, Suzuki K, Nakazawa A, Saiki R, Seki M, Takita J, Ogawa S, Manabe A, Hasegawa D. Clonal evidence for the development of neuroblastoma with extensive copy-neutral loss of heterozygosity arising in a mature teratoma. Cancer Sci 2021; 112:2921-2927. [PMID: 33934450 PMCID: PMC8253283 DOI: 10.1111/cas.14931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 11/27/2022] Open
Abstract
Mature teratomas are usually benign tumors that rarely undergo malignant transformation. We report an advanced neuroblastoma arising in a mature teratoma of the ovary. Whole-exome sequencing identified extensive copy-neutral loss of heterozygosity (LOH) in both neuroblastoma and teratoma elements, suggesting that the neuroblastoma evolved from the teratoma. In addition, several truncating germline heterozygous variants in tumor suppressor genes, including RBL2 and FBXW12, became homozygous as a result of LOH. Collectively, we speculate that extensive LOH in teratoma cells may force heterozygous germline variants to become homozygous, which, in turn, may contribute to the development of neuroblastoma with the acquisition of additional chromosomal changes.
Collapse
Affiliation(s)
- Rintaro Ono
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan
| | - Hiroo Ueno
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenichi Yoshida
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoko Takahashi
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan.,Department of Pediatrics, Japanese Red Cross Narita Hospital, Chiba, Japan
| | - Hiroki Yoshihara
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan
| | - Taiki Nozaki
- Department of Diagnostic Radiology, St. Luke's International Hospital, Tokyo, Japan
| | - Koyu Suzuki
- Department of Pathology, St. Luke's International Hospital, Tokyo, Japan
| | - Atsuko Nakazawa
- Department of Clinical Research, Saitama Children's Medical Center, Saitama, Japan
| | - Ryunosuke Saiki
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masafumi Seki
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junko Takita
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsushi Manabe
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan.,Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Daisuke Hasegawa
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan
| |
Collapse
|
19
|
Veena K, Jayasree M, Karthik B, Preethi N. Mature Teratoma with Gliomatosis Peritonei with Abdominal Tuberculosis: A Diagnostic Dilemma. J Gynecol Surg 2021. [DOI: 10.1089/gyn.2020.0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- K.S. Veena
- Department of Obstetrics and Gynecology, Sri Manakula Vinayagar Medical College and Hospital, Puducherry, India
| | - Manivasakan Jayasree
- Department of Obstetrics and Gynecology, Sri Manakula Vinayagar Medical College and Hospital, Puducherry, India
| | - Bhandary Karthik
- Department of Pediatric Surgery, Sri Manakula Vinayagar Medical College and Hospital, Puducherry, India
| | - Natarajan Preethi
- Department of Obstetrics and Gynecology, Sri Manakula Vinayagar Medical College and Hospital, Puducherry, India
| |
Collapse
|
20
|
Shao L, Heider A, Rabah R. Single nucleotide polymorphism array and cytogenetic analyses of ovarian teratomas in children. Genes Chromosomes Cancer 2021; 60:418-425. [PMID: 33377559 DOI: 10.1002/gcc.22934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 12/20/2022] Open
Abstract
Teratomas are the most common tumors in the ovary during childhood. Previous studies suggested that they may be derived from germ cells at any developmental stage from premeiotic oogonia through meiotic oocytes to post-meiotic ova. The majority of mature teratomas reveal normal karyotypes and immature teratomas show higher frequency of chromosomal abnormalities. We analyzed fresh tissue samples from 25 primary ovarian teratomas and three extraovarian deposits using whole genome single nucleotide polymorphism (SNP) array and karyotype. SNP array detected five patterns of copy neutral loss of heterozygosity (CN-LOH): failure of meiosis I (type I) in 12 tumors, failure of meiosis II (type II) in six tumors, endoreduplication of a haploid ovum (type III) in two tumors, premeiotic error (type IV) in four tumors, and both meiotic I and meiotic II errors in one tumor (type V). Three tumors with type I error had a single chromosome showing meiotic II error, and two tumors with type II error had a single chromosome showing premature sister-chromatid separation in meiosis I. Lack of recombination in multiple chromosomes in meiosis I were common, chromosomes 17, 7, 8, 21, and 22 were most commonly involved. Abnormal karyotypes were observed in four teratomas including +3, del(3q), +7, +8, +12, and i(18q). The extraovarian deposits revealed the same CN-LOH pattern as the primary teratoma. In summary, SNP array reveals the origin of ovarian teratoma and we propose a new mechanism that consecutive meiotic I and II errors occur frequently in ovarian teratomas.
Collapse
Affiliation(s)
- Lina Shao
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Amer Heider
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Raja Rabah
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
21
|
Kommoss FKF, Cheasley D, Wakefield MJ, Scott CL, Campbell IG, Gilks CB, Gorringe K. Primary mucinous ovarian neoplasms rarely show germ cell histogenesis. Histopathology 2020; 78:640-642. [PMID: 33151585 DOI: 10.1111/his.14297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Felix K F Kommoss
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Matthew J Wakefield
- Walter and Eliza Hall Institute, Parkville, Australia.,University of Melbourne, Parkville, Australia
| | - Clare L Scott
- Peter MacCallum Cancer Centre, Melbourne, Australia.,Walter and Eliza Hall Institute, Parkville, Australia.,University of Melbourne, Parkville, Australia
| | - Ian G Campbell
- Peter MacCallum Cancer Centre, Melbourne, Australia.,University of Melbourne, Parkville, Australia
| | - C Blake Gilks
- Department of Pathology, Vancouver General Hospital and University of British Columbia, Vancouver, BC, Canada
| | - Kylie Gorringe
- Peter MacCallum Cancer Centre, Melbourne, Australia.,University of Melbourne, Parkville, Australia
| |
Collapse
|
22
|
Molecular Pathways and Targeted Therapies for Malignant Ovarian Germ Cell Tumors and Sex Cord-Stromal Tumors: A Contemporary Review. Cancers (Basel) 2020; 12:cancers12061398. [PMID: 32485873 PMCID: PMC7353025 DOI: 10.3390/cancers12061398] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
Non-epithelial ovarian tumors are heterogeneous and account for approximately 10% of ovarian malignancies. The most common subtypes of non-epithelial ovarian tumors arise from germ cells or sex cord and stromal cells of the gonads. These tumors are usually detected at an early stage, and management includes surgical staging and debulking. When indicated for advanced disease, most respond to chemotherapy; however, options for patients with refractory disease are limited, and regimens can be associated with significant toxicities, including permanent organ dysfunction, secondary malignancies, and death. Targeted therapies that potentially decrease chemotherapy-related adverse effects and improve outcomes for patients with chemotherapy-refractory disease are needed. Here, we review the molecular landscape of non-epithelial ovarian tumors for the purpose of informing rational clinical trial design. Recent genomic discoveries have uncovered recurring somatic alterations and germline mutations in subtypes of non-epithelial ovarian tumors. Though there is a paucity of efficacy data on targeted therapies, such as kinase inhibitors, antibody–drug conjugates, immunotherapy, and hormonal therapy, exceptional responses to some compounds have been reported. The rarity and complexity of non-epithelial ovarian tumors warrant collaboration and efficient clinical trial design, including high-quality molecular characterization, to guide future efforts.
Collapse
|