1
|
Batatinha JAP, Nishi MY, Batista RL, Faria Júnior JAD, Sircili MHP, Denes FT, Mesia MJC, Salvanini LDS, Costa EMF, Carvalho FM, Mendonca BB, Domenice S. Estrogen-secreting testicular tumors in 46,XY female patients with 17α-hydroxylase/17,20-lyase deficiency: two unusual case reports and a review of the literature. Front Genet 2025; 16:1508792. [PMID: 40313596 PMCID: PMC12043694 DOI: 10.3389/fgene.2025.1508792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/18/2025] [Indexed: 05/03/2025] Open
Abstract
Context 17α-hydroxylase/17,20-lyase deficiency (17OHD) is a rare autosomal recessive condition. Women who have the complete form of 17OHD typically have a female phenotype, with an absence of secondary sexual characteristics, primary amenorrhea, and hypertension, which is usually detected in adolescence. Generally, 46,XY patients with a partial form of 17OHD have atypical genitalia and intra-abdominal or inguinal testes. The risk of developing malignant testicular tumors or testicular adrenal rest tumors in 21-hydroxylase deficiency congenital adrenal hyperplasia is reported in 46,XY patients. In contrast, these conditions are rarely described in patients with 17OHD. Objective This study aims to investigate patients with 17OHD who exhibit testicular tumors and spontaneous pubertal development. Patients and Results Two unrelated women with 46,XY karyotype with 17OHD who presented with unexpected spontaneous development of secondary sexual characteristics and testicular tumors were described. Pathogenic allelic variants in CYP17A1 were identified in the compound heterozygous state in both patients. The variants p.Trp406Arg and p.Pro428Leu were identified in the patient with Leydig cell neoplasia plus germ cell neoplasia in situ, and the p.Arg358Gln and p.Trp406Arg variants were identified in the patient with intratubular seminoma associated with invasive classic seminoma. Conclusion Our findings reinforce the risk of testicular tumor development among 46,XY patients with 17OHD and add data to the discussion of the risk/benefit ratio of prophylactic gonadectomy in the treatment patients with 46, XY differences in sex development (DSD).
Collapse
Affiliation(s)
- Julio Americo Pereira Batatinha
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Mirian Yumie Nishi
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Rafael Loch Batista
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - José Antônio Diniz Faria Júnior
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Maria Helena Palma Sircili
- Departamento de Urologia do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Francisco Tibor Denes
- Departamento de Urologia do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Maria Jimena Chafloque Mesia
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Laura da Silva Salvanini
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Elaine Maria Frade Costa
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Filomena Marino Carvalho
- Departamento de Patologia da Faculdade de Medicina da Universidade de São Paulo,São Paulo, Brazil
| | - Berenice Bilharinho Mendonca
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sorahia Domenice
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Bode PK, Blasco-Santana L, Colmenero I, Reyes-Múgica M. Germ cell tumors in children. Virchows Arch 2025; 486:65-79. [PMID: 39779499 PMCID: PMC11782398 DOI: 10.1007/s00428-025-04023-7] [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: 12/08/2024] [Revised: 12/27/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Pediatric germ cell tumors represent a rare but biologically diverse group of neoplasms arising from pluripotent primordial germ cells. The 2022 edition of the WHO Classification of Pediatric Tumors introduced the first organ independent classification of germ cell tumors, reflecting advances in molecular biology, histopathology, and clinical practice. This review highlights the key changes, including the refined distinctions between the different subtypes. These updates enhance diagnostic accuracy and provide a framework for understanding age-dependent differences in tumor biology and behavior. Emphasis is placed on integrating the new classification into multidisciplinary care, particularly in addressing diagnostic challenges in pre- and post-pubertal-type germ cell tumors. By bridging the gap between histopathology and oncology, the updated classification represents a pivotal step forward in improving outcomes for children with germ cell tumors.
Collapse
Affiliation(s)
- Peter Karl Bode
- Department of Pathology, Kantonsspital Winterthur, Winterthur, Switzerland.
| | - Luis Blasco-Santana
- Department of Histopathology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Isabel Colmenero
- Department of Histopathology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Miguel Reyes-Múgica
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
3
|
Bîcă O, Ciongradi CI, Benchia D, Sârbu I, Alecsa M, Cristofor AE, Bîcă DE, Lozneanu L. Assessment of Molecular Markers in Pediatric Ovarian Tumors: Romanian Single-Center Experience. Int J Mol Sci 2024; 25:6752. [PMID: 38928458 PMCID: PMC11204196 DOI: 10.3390/ijms25126752] [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: 05/19/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Pediatric ovarian tumors exhibit unique diagnostic and therapeutic challenges. This study evaluates the expression of SALL4 and OCT3/4 biomarkers in pediatric ovarian tumors and their associations with tumor subtype, stage, and clinical outcome. A retrospective analysis was conducted on 64 patients under 18 years old, examining demographic data, tumor characteristics, immunohistochemical staining, and clinical outcomes. Our results show that SALL4 was significantly expressed in adenocarcinoma, dysgerminoma (DSG), mixed germ cell tumors (GCTs), and immature teratoma, while OCT3/4 was highly expressed in DSG and mixed GCTs. Both markers are associated with a higher tumor grade and stage, indicating a more aggressive disease. The SALL4 positivity expression was correlated with high alpha fetoprotein (AFP) and lactate dehydrogenase (LDH) levels, while OCT3/4 positivity significantly predicted the risk of subsequent metastasis. The mean progression-free survival (PFS) was notably shorter in patients with positive markers. These findings underscore the diagnostic and prognostic value of SALL4 and OCT3/4 in pediatric ovarian tumors, aligning with previous research and supporting their use in clinical practice for better disease management and patient outcomes.
Collapse
Affiliation(s)
- Ovidiu Bîcă
- 2nd Department of Surgery—Pediatric Surgery and Orthopedics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Carmen Iulia Ciongradi
- 2nd Department of Surgery—Pediatric Surgery and Orthopedics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Diana Benchia
- 2nd Department of Surgery—Pediatric Surgery and Orthopedics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ioan Sârbu
- 2nd Department of Surgery—Pediatric Surgery and Orthopedics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Mirabela Alecsa
- Department of Mother and Child, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Alexandra Elena Cristofor
- Department of Obstetrics and Gynecology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Delia Elena Bîcă
- Department of Clinical Pharmacology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ludmila Lozneanu
- Department of Morpho-Functional Sciences I—Histology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| |
Collapse
|
4
|
Wessman S, Nistér M, Kokaraki G, Pal N, Tettamanti G, Petta TB, Carlson JW. A comprehensive population-based study of malignant ovarian tumors, including histologic and immunohistochemical review, in children and adolescents 0-19 years old in Sweden between 1970 and 2014. Gynecol Oncol 2024; 184:206-213. [PMID: 38340646 DOI: 10.1016/j.ygyno.2024.01.035] [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: 04/24/2023] [Revised: 07/21/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024]
Abstract
OBJECTIVES Ovarian tumors in the pediatric population are rare. The incidence and frequency of subtypes differ between children and adults. Although not all tumors are aggressive, they may still lead to morbidity. The goal of this study was a comprehensive review of malignant ovarian tumors in children and adolescents diagnosed and registered in Sweden. METHODS Individuals were identified through a search in the National Cancer Register, limited for ages 0-19, years 1970-2014. Stored tumor diagnostic material from regional biobanks was retrieved and reviewed. RESULTS The study includes 345 individuals with ovarian tumors and 70.7% of them were between 15 and 19 years at time of diagnosis. No differences in incidence over time or geographic location were identified. The average follow-up time was 21.2 years and 5-year survival was 88.4%. Survival was similar in the different time periods, except for 1970-1979. Review was possible for 260 cases, resulting in 85 epithelial tumors, 121 GCTs, 47 SCSTs and 7 others. For age 0-4 years SCSTs dominated (85.7%), for 5-9- and 10-14-years GCTs dominated (70,8% and 75.0% respectively), and for age 15-19 years epithelial tumors dominated (43.8%). There was a strong agreement between review diagnosis and original diagnosis (Cohen's κ 0.944). Differentiating between entities within the sex cord-stromal group posed the biggest diagnostic challenge. CONCLUSIONS Ovarian tumors in children and adolescents are rare and distinct from their adult counterparts regarding incidence and frequency. There was a strong concurrence between original and review diagnoses. The greatest diagnostic difficulty was subtyping of epithelial tumors and differentiating between tumors within the SCST group.
Collapse
Affiliation(s)
- Sandra Wessman
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Pathology and Cancer diagnostics, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Monica Nistér
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Pathology and Cancer diagnostics, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Georgia Kokaraki
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, 90033 Los Angeles, CA, USA
| | - Niklas Pal
- Department of Women's and Children's Health, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Pediatric Oncology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Giorgio Tettamanti
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Institute of Environmental Medicine, Unit of Epidemiology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Tirzah Braz Petta
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, 90033 Los Angeles, CA, USA; Department of Cellular Biology and Genetics, Federal University of Rio Grande de Norte, Natal, RN 59078-970, Brazil
| | - Joseph W Carlson
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, 90033 Los Angeles, CA, USA.
| |
Collapse
|
5
|
Choi JH, Thung SN. Recent Advances in Pathology of Intrahepatic Cholangiocarcinoma. Cancers (Basel) 2024; 16:1537. [PMID: 38672619 PMCID: PMC11048541 DOI: 10.3390/cancers16081537] [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: 02/29/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Intrahepatic cholangiocarcinoma (ICCA) is a malignant epithelial neoplasm characterized by biliary differentiation within the liver. ICCA is molecularly heterogeneous and exhibits a broad spectrum of histopathological features. It is a highly aggressive carcinoma with high mortality and poor survival rates. ICCAs are classified into two main subtypes: the small-duct type and large-duct types. These two tumor types have different cell origins and clinicopathological features. ICCAs are characterized by numerous molecular alterations, including mutations in KRAS, TP53, IDH1/2, ARID1A, BAP1, BRAF, SAMD4, and EGFR, and FGFR2 fusion. Two main molecular subtypes-inflammation and proliferation-have been proposed. Recent advances in high-throughput assays using next-generation sequencing have improved our understanding of ICCA pathogenesis and molecular genetics. The diagnosis of ICCA poses a significant challenge for pathologists because of its varied morphologies and phenotypes. Accurate diagnosis of ICCA is essential for effective patient management and prognostic determination. This article provides an updated overview of ICCA pathology, focusing particularly on molecular features, histological subtypes, and diagnostic approaches.
Collapse
Affiliation(s)
- Joon Hyuk Choi
- Department of Pathology, Yeungnam University College of Medicine, Daegu 42415, Republic of Korea
| | - Swan N. Thung
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY 10029, USA;
| |
Collapse
|
6
|
Wood GE, Bunting CP, Veli M, Arora R, Berney DM, Alifrangis C, MacDonald ND, Miller RE, Shamash J, Stoneham S, Lockley M. Seminoma and dysgerminoma: evidence for alignment of clinical trials and de-escalation of systemic chemotherapy. Front Oncol 2023; 13:1271647. [PMID: 37954076 PMCID: PMC10634240 DOI: 10.3389/fonc.2023.1271647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/18/2023] [Indexed: 11/14/2023] Open
Abstract
Malignant germ cell tumours are a group of rare cancers whose incidence peaks in late adolescence and early adulthood. Dysgerminomas of the ovary and seminomas of the testis are analogous diseases, but seminomas have a 10-fold higher incidence. The two tumours are morphologically identical and are only differentiated by surrounding organ-specific tissue or testicular germ cell neoplasia in situ. They share genetic features including KIT and RAS mutations, amplification of chromosome 12p, and expression of pluripotency markers (NANOG (Nanog homeobox), OCT3/4 (Octamer-binding transcription factor 3/4), and SAL4 (Spalt-like trascription factor 4)). Both histologies are exquisitely sensitive to platinum chemotherapy, and the combination of bleomycin, etoposide, and cisplatin (BEP) yields survival rates greater than 90%. However, BEP causes significant, lifelong toxicity (cardiovascular, renal, respiratory, and neurological) in these young patients with an expectation of cure. Here, we comprehensively review the biological features of dysgerminoma and seminoma to demonstrate that they are biologically analogous diseases. We present available clinical trial data supporting de-escalation of chemotherapy treatment. Finally, we propose that future trials should enrol men, women, and children to benefit all patients regardless of age or sex.
Collapse
Affiliation(s)
- Georgina E. Wood
- Medical Oncology, University College London Hospital, London, United Kingdom
| | - Christopher P. Bunting
- Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mesel Veli
- Medical Oncology, University College London Hospital, London, United Kingdom
| | - Rupali Arora
- Histopathology, University College London Hospital, London, United Kingdom
| | | | | | - Nicola D. MacDonald
- Department of Gynaecology, University College London Hospital, London, United Kingdom
| | - Rowan E. Miller
- Medical Oncology, University College London Hospital, London, United Kingdom
- Medical Oncology, Barts Health NHS Trust, London, United Kingdom
| | - Jonathan Shamash
- Medical Oncology, Barts Health NHS Trust, London, United Kingdom
| | - Sara Stoneham
- Paediatric Oncology, University College London Hospital, London, United Kingdom
| | - Michelle Lockley
- Medical Oncology, University College London Hospital, London, United Kingdom
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
7
|
Nagata K, Shimada T, Eishi C, Nishi M, Murakami T, Ohashi K, Kajimura I, Miura K. Dysgerminoma of the Left Ovary in a Patient with Balanced Translocation 46X, t(X:1) (q22;q21): A Case Report. Int Med Case Rep J 2023; 16:117-122. [PMID: 36915667 PMCID: PMC10008024 DOI: 10.2147/imcrj.s395511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/28/2023] [Indexed: 03/09/2023] Open
Abstract
We report a case of dysgerminoma in a 22-year-old woman diagnosed with chromosomal abnormality, balanced translocation 46X,t(X:1)(q22;q21). She had received hormone replacement therapy for 7 years for primary amenorrhea. She visited a primary care physician because of lower abdominal distension, and a large tumor in the pelvis was discovered. She was admitted to our hospital for further examination of the pelvic tumor. She underwent laparotomy and was diagnosed with stage IIIA1 dysgerminoma (pT3apN0pM0) of the left ovary. Young female patients without the Y chromosome who are treated for primary amenorrhea may also develop malignant germ cell tumors; therefore, gynecologists should provide hormone replacement therapy and periodic pelvic evaluation.
Collapse
Affiliation(s)
- Koh Nagata
- Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki, Japan
| | - Takako Shimada
- Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki, Japan
| | - Chiaki Eishi
- Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki, Japan
| | - Masaki Nishi
- Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki, Japan
| | - Toru Murakami
- Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki, Japan
| | - Kazuaki Ohashi
- Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki, Japan
| | - Itsuki Kajimura
- Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki, Japan
| | - Kiyonori Miura
- Obstetrics and Gynecology, Nagasaki University Hospital, Nagasaki, Japan
| |
Collapse
|
8
|
Fang F, Iaquinta PJ, Xia N, Liu L, Diao L, Reijo Pera RA. Transcriptional control of human gametogenesis. Hum Reprod Update 2022; 28:313-345. [PMID: 35297982 PMCID: PMC9071081 DOI: 10.1093/humupd/dmac002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 11/22/2021] [Indexed: 11/14/2022] Open
Abstract
The pathways of gametogenesis encompass elaborate cellular specialization accompanied by precise partitioning of the genome content in order to produce fully matured spermatozoa and oocytes. Transcription factors are an important class of molecules that function in gametogenesis to regulate intrinsic gene expression programs, play essential roles in specifying (or determining) germ cell fate and assist in guiding full maturation of germ cells and maintenance of their populations. Moreover, in order to reinforce or redirect cell fate in vitro, it is transcription factors that are most frequently induced, over-expressed or activated. Many reviews have focused on the molecular development and genetics of gametogenesis, in vivo and in vitro, in model organisms and in humans, including several recent comprehensive reviews: here, we focus specifically on the role of transcription factors. Recent advances in stem cell biology and multi-omic studies have enabled deeper investigation into the unique transcriptional mechanisms of human reproductive development. Moreover, as methods continually improve, in vitro differentiation of germ cells can provide the platform for robust gain- and loss-of-function genetic analyses. These analyses are delineating unique and shared human germ cell transcriptional network components that, together with somatic lineage specifiers and pluripotency transcription factors, function in transitions from pluripotent stem cells to gametes. This grand theme review offers additional insight into human infertility and reproductive disorders that are linked predominantly to defects in the transcription factor networks and thus may potentially contribute to the development of novel treatments for infertility.
Collapse
Affiliation(s)
- Fang Fang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Phillip J Iaquinta
- Division of Research, Economic Development, and Graduate Education, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Ninuo Xia
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Lei Liu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Lei Diao
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Renee A Reijo Pera
- Division of Research, Economic Development, and Graduate Education, California Polytechnic State University, San Luis Obispo, CA, USA
- McLaughlin Research Institute, Great Falls, MT, USA
| |
Collapse
|
9
|
Warnnissorn M, Watkins JC, Young RH. Dysgerminoma of the Ovary: An Analysis of 140 Cases Emphasizing Unusual Microscopic Findings and Resultant Diagnostic Problems. Am J Surg Pathol 2021; 45:1009-1027. [PMID: 33577182 DOI: 10.1097/pas.0000000000001687] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
One-hundred fourty pure dysgerminomas were evaluated with particular focus on the microscopic features as seen in 125 cases with available slides. The patients ranged from 8 to 59 years of age (mean, 24.1 y). The tumors, bilateral in 4% of the cases and with a mean tumor diameter of 13 cm, were typically soft, lobulated, homogeneous, and creamy white to tan to yellow but necrosis was found in 13%, hemorrhage in 20%, and focal cystic change in 15%. On microscopic examination, the patterns and other notable features encountered, including their frequency, were as follows: an alveolar pattern resulting from delicate fibrovascular septa (51%), diffuse (33%), macronodular (14%), insular (26%), cords (28%), solid tubular (17%), microspaces (sometimes simulating glands) (12%), follicle-like spaces (5%), prominent fibrous bands (65%), stromal edema (56%), stromal luteinization (9%), granulomatous infiltrate (46%), lymphocytic infiltrate (100%), Langhans cell type giant cells (35%), syncytiotrophoblast giant cells (6%), prominent population of cells with pale to clear cytoplasm (73%), cells with amphophilic to eosinophilic cytoplasm (53%) and vacuolated occasionally signet ring-like cells (7%). Various constellations of the above findings often resulted in an appearance different from that usually portrayed in the literature and certain tumors of very different nature being in the differential such as undifferentiated carcinoma not otherwise specified, small cell carcinoma of hypercalcemic type, and malignant lymphoma. The correct diagnosis can be arrived at by considering the usual relative youth of the patient, often rather characteristic gross features, and most crucially careful attention to the microscopic features and awareness of variant morphologic findings. Those that are particularly problematic based on this study are diffuse growth with inconspicuous fibrovascular septa, macronodules, cords, solid tubular formations, spaces ranging from small to large, and mimicking glands or follicles, prominent fibrous to edematous stroma, and cells with amphophilic to eosinophilic cytoplasm. According to the degree of difficulty and confidence of the interpreter, well-known immunohistochemical features of dysgerminoma, which largely differ from those of other neoplasms in the differential, will aid if felt indicated.
Collapse
Affiliation(s)
- Malee Warnnissorn
- James Homer Wright Pathology Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | | |
Collapse
|
10
|
Chen J, Li Y, Wu J, Liu Y, Kang S. Whole-exome sequencing reveals potential germline and somatic mutations in 60 malignant ovarian germ cell tumors†. Biol Reprod 2021; 105:164-178. [PMID: 33739378 DOI: 10.1093/biolre/ioab052] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/05/2021] [Accepted: 03/16/2021] [Indexed: 11/13/2022] Open
Abstract
Malignant ovarian germ cell tumors (MOGCTs) are rare and heterogeneous ovary tumors. We aimed to identify potential germline mutations and somatic mutations in MOGCTs by whole-exome sequencing. The peripheral blood and tumor samples from these patients were used to identify germline mutations and somatic mutations, respectively. For those genes with copy number alterations (deletion and duplication region), functional annotation was performed. Immunohistochemistry was performed to evaluate the expression of mutated genes corresponding to CNA deletion region and duplication region. In peripheral blood, copy number loss and gain were mostly found in yolk sac tumors (YSTs). Moreover, POU5F1 was the most significant mutated gene with mutation frequency >10% in both CNA deletion and duplication region. In addition, strong cytoplasm staining of POU5F1 (corresponding to CNA deletion region and duplication region) was found in two YST and nuclear staining in two dysgerminomas tumor samples. Genes corresponding to CNA deletion region were significantly enriched in the signaling pathway of regulating pluripotency of stem cells. In addition, genes corresponding to CNA duplication region were significantly enriched in the signaling pathways of RIG-I (DExD/H-box helicase 58)-like receptor, Toll-like receptor and nuclear factor (NF)-kappa. Keratin 4 (KRT4), ribosomal protein L14 (RPL14), proprotein convertase subtilisin/kexin type 6 (PCSK6), poly(A)-binding protein cytoplasmic 3 (PABPC3), and sterile alpha and TIR motif containing 1 (SARM1) mutations were detected in both peripheral blood and tumor samples. Identification of potential germline mutations and somatic mutations in MOGCTs may provide a new field in understanding the genetic feature of the rare biological tumor type in the ovary.
Collapse
Affiliation(s)
- Juan Chen
- Department of Obstetrics and Gynaecology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Hebei, China
| | - Yan Li
- Department of Molecular Biology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Hebei, China
| | - Jianlei Wu
- Department of Obstetrics and Gynaecology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Hebei, China
| | - Yakun Liu
- Department of Obstetrics and Gynaecology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Hebei, China
| | - Shan Kang
- Department of Obstetrics and Gynaecology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Hebei, China
| |
Collapse
|
11
|
Francis F, Zhou M. Clinical Applications of Immunohistochemistry in Germ Cell Tumors in Men. Methods Mol Biol 2021; 2195:13-29. [PMID: 32852754 DOI: 10.1007/978-1-0716-0860-9_2] [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] [Indexed: 12/24/2022]
Abstract
Germ cell tumors (GCT) in men comprise of tumor subtypes with distinct histomorphologies, genetic and genomic alterations, and clinical behavior. Immunohistochemical (IHC) markers, including many newly described nuclear transcription factors, are often applied in challenging cases to arrive at a correct diagnosis and classification, and to establish germ cell origin for metastatic tumors. However, there is no established role for IHC markers in prognosis and therapy response prediction in GCTs. This chapter briefly reviews the clinical utility of IHC in diagnosis and classification of GCTs, including technical aspects of performing IHC and clinical applications of commonly used IHC markers in the workup of common and clinically relevant diagnostic scenarios.
Collapse
Affiliation(s)
- Franto Francis
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ming Zhou
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Tufts Medical Center and Tufts School of Medicine, Boston, MA, USA.
| |
Collapse
|
12
|
Litzka C, Leebmann H, Wiesinger H, Kirchner T, Seelbach-Göbel B, Häusler SFM. [Management of intra-abdominal dysgerminoma During pregnancy - A Case Report and Literature Review]. Z Geburtshilfe Neonatol 2020; 224:306-314. [PMID: 32242331 DOI: 10.1055/a-1112-0506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Dysgerminomas are rare malignant germ cell tumors. They usually arise from the ovary, but case reports describing extraovarian dysgerminomas do exist. When treated adequately the disease has a good prognosis. Dysgerminomas diagnosed during pregnancy are very rare. METHODOLOGY Report of extraovarian intra-abdominal dysgerminoma during pregnancy. Systematic literature review. CASE REPORT A 35-year-old second gravida was diagnosed with a suspected intra-abdominal mass at 20 gestational weeks. During an exploratory laparotomy, a tumor infiltrating the transverse colon and histologically identified as a dysgerminoma was resected. Ovaries were clinically unremarkable. The induction of chemotherapy was postponed until after delivery. At 34 gestational weeks the patient underwent cesarean section and tumor debulking. Four cycles of bleomycin, etoposide and cisplatin were administered. After 12 months, cystic ovaries were found. Hysterectomy with bilateral adnexectomy was performed but no malignancy found. After 16 months, the patient was still in complete remission. CONCLUSION We describe the first-ever published dysgerminoma in gravida primarily evolving intraabdominally and not affecting the ovaries. The decision for cytoreductive surgery, prolongation of pregnancy and postponing chemotherapy until after delivery combined the best benefit for the baby with a good maternal prognosis. Due to limited data regarding dysgerminomas in pregnancy, individual interdisciplinary concepts are mandatory.
Collapse
Affiliation(s)
- Christian Litzka
- Schwerpunkt Geburtshilfe, Krankenhaus Barmherzige Brüder Regensburg, Frauenklinik Sankt Hedwig, Regensburg
| | - Hubert Leebmann
- Klinik für Allgemein- und Viszeralchirurgie, Krankenhaus Barmherzige Brüder Regensburg, Regensburg
| | | | - Thomas Kirchner
- Institut für Pathologie, Ludwig-Maximilians-Universität München, München
| | - Birgit Seelbach-Göbel
- Schwerpunkt Geburtshilfe, Krankenhaus Barmherzige Brüder Regensburg, Frauenklinik Sankt Hedwig, Regensburg
| | - Sebastian Franz Martin Häusler
- Schwerpunkt Geburtshilfe, Krankenhaus Barmherzige Brüder Regensburg, Frauenklinik Sankt Hedwig, Regensburg.,Universitätsfrauenklinik, Universitätsklinikum Würzburg Frauenklinik und Poliklinik, Würzburg
| |
Collapse
|
13
|
Kei S, Adeyi OA. Practical Application of Lineage-Specific Immunohistochemistry Markers: Transcription Factors (Sometimes) Behaving Badly. Arch Pathol Lab Med 2019; 144:626-643. [PMID: 31385722 DOI: 10.5858/arpa.2019-0226-ra] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Transcription factors (TFs) are proteins that regulate gene expression and control RNA transcription from DNA. Lineage-specific TFs have increasingly been used by pathologists to determine tumor lineage, especially in the setting of metastatic tumors of unknown primary, among other uses. With experience gathered from its daily application and increasing pitfalls reported from immunohistochemical studies, these often-touted highly specific TFs are not as reliable as once thought. OBJECTIVES.— To summarize the established roles of many of the commonly used TFs in clinical practice and to discuss known and potential sources for error (eg, false-positivity from cross-reactivity, aberrant, and overlap "lineage-specific" expression) in their application and interpretation. DATA SOURCES.— Literature review and the authors' personal practice experience were used. Several examples selected from the University Health Network (Toronto, Ontario, Canada) are illustrated. CONCLUSIONS.— The application of TF diagnostic immunohistochemistry has enabled pathologists to better assess the lineage/origin of primary and metastatic tumors. However, the awareness of potential pitfalls is essential to avoid misdiagnosis.
Collapse
Affiliation(s)
- Si Kei
- From the Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (Dr Lou); and the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Adeyi)
| | - Oyedele A Adeyi
- From the Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (Dr Lou); and the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Adeyi)
| |
Collapse
|
14
|
|
15
|
Monferrer E, Burgos-Panadero R, Blanquer-Maceiras M, Cañete A, Navarro S, Noguera R. High Oct4 expression: implications in the pathogenesis of neuroblastic tumours. BMC Cancer 2019; 19:1. [PMID: 30606139 PMCID: PMC6318846 DOI: 10.1186/s12885-018-5219-3] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/13/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Neuroblastic tumours (NBTs) are paediatric solid tumours derived from embryonic neural crest cells which harbour their own cancer stem cells (CSC). There is evidence indicating that CSC may be responsible for tumour progression, chemotherapy resistance and recurrence in NBTs. Oct4 is a transcription factor which plays a key role in mammal embryonic development and stem cell fate regulation. The aim of the study is to elucidate the clinical significance of Oct4 in NBTs. METHODS We studied Oct4 expression in 563 primary NBTs using digital image quantification. Chi-square test was applied to analyse the correlation between histopathology and the Oct4+ cell percentage. Survival analysis was carried out with Kaplan-Meier curves and log-rank test. Additionally, a multivariate Cox regression analysis with the stepwise backwards (Wald) method was undertaken to calculate the impact of Oct4 expression level on survival. RESULTS We found that tumours with a high proportion of cells expressing Oct4 correlated statistically with undifferentiated and poorly differentiated neuroblastoma / nodular ganglioneuroblastoma, and that Oct4 expression was not present in ganglioneuroma (p < 0.05). Statistical analysis also indicated a relationship between high Oct4 expression levels, high-risk patients according to the International Neuroblastoma Risk Group pre-treatment classification parameters, larger blood vessels and low survival rates. CONCLUSIONS These results suggest that the Oct4 gene may regulate NBT pathogenic differentiation pathways, and should thus be considered as a target for knockdown when developing novel therapies for high-risk NBT patients.
Collapse
Affiliation(s)
- Ezequiel Monferrer
- Pathology Department, Medical School, University of Valencia-INCLIVA, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Rebeca Burgos-Panadero
- Pathology Department, Medical School, University of Valencia-INCLIVA, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Maite Blanquer-Maceiras
- Pathology Department, Medical School, University of Valencia-INCLIVA, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
- CIBERONC, Madrid, Spain
| | - Adela Cañete
- Pediatric Oncology Unit, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Samuel Navarro
- Pathology Department, Medical School, University of Valencia-INCLIVA, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
- CIBERONC, Madrid, Spain
| | - Rosa Noguera
- Pathology Department, Medical School, University of Valencia-INCLIVA, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
- CIBERONC, Madrid, Spain
| |
Collapse
|
16
|
Expression of OCT4 and SALL4 in Diffuse Large B-cell Lymphoma: An Analysis of 145 Consecutive Cases and Testicular Lymphomas. Am J Surg Pathol 2017; 40:950-7. [PMID: 27035612 DOI: 10.1097/pas.0000000000000648] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OCT4 and SALL4 are transcription factors within a complex network that functions to maintain pluripotency in primitive stem cells and germ cells. Nuclear expression of OCT4 is widely cited as sensitive and specific for primary and metastatic germ cell tumors and is commonly used in the diagnosis of central nervous system (CNS) germinomas. Studies have failed to systematically examine the expression of OCT4 or SALL4 in diffuse large B-cell lymphoma (DLBCL), although this entity enters the morphologic differential diagnosis of some germ cell tumors. A retrospective review was conducted on 145 consecutive cases of DLBCL and testicular lymphoma to evaluate the prevalence of OCT4 and SALL4 expression. Nuclear OCT4 expression was present in 2/11 (18%) testicular DLBCLs and 6/134 (4.5%) nontesticular DLBCLs. Most OCT4 cases demonstrated moderate to strong expression in >50% of neoplastic cells. Rare, weak nuclear SALL4 expression was detected in only 3 nontesticular DLBCLs. Within the extratesticular DLBCL group, 2/6 (33%) primary CNS DLBCLs expressed nuclear OCT4. In addition, OCT4 DLBCL showed an overall predilection toward non-germinal center B-cell phenotype (7/8; 88%) and had a higher than expected rate of CD5 coexpression (4/8, 50%). These results are cautionary against using OCT4 as a sole marker of germ cell differentiation in testicular and extratesticular sites, especially in the CNS. The apparent associations of OCT4 expression with primary CNS DLBCL, non-germinal center B-cell phenotype, and CD5 coexpression raise the question of whether OCT4 expression in DLBCL may reflect more aggressive biology.
Collapse
|
17
|
Perspectives on testicular germ cell neoplasms. Hum Pathol 2017; 59:10-25. [DOI: 10.1016/j.humpath.2016.08.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 11/16/2022]
|
18
|
Yadav P, Khaladkar S, Gujrati A. Imaging Findings in Dysgerminoma in a Case of 46 XY, Complete Gonadal Dysgenesis (Swyer syndrome). J Clin Diagn Res 2016; 10:TD10-TD12. [PMID: 27790550 DOI: 10.7860/jcdr/2016/19488.8493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 07/05/2016] [Indexed: 11/24/2022]
Abstract
A 46 XY pure gonadal dysgenesis also known as Swyer syndrome. These patients are phenotypic females with normal female external genitalia and absent testicular tissue. The patients with swyer syndrome have streak gonads and increased risk of dysgerminoma and gonadoblastoma. We present a case of dysgerminoma in dysgenetic gonads of swyer syndrome. A 23-year-old female had come with complaints of primary amenorrhea, pelvic mass and abdominal pain. Clinical findings, pathology investigation and imaging findings revealed swyer syndrome. On MRI it showed a large lobulated mass in the pelvis. Mass was excised and dysgerminoma was given on the histopathology.
Collapse
Affiliation(s)
- Pratiksha Yadav
- Associate Professor, Department of Radio-diagnosis, Dr. D. Y. Patil Medical College, Hospital and Research Centre , Pimpri, Pune, Maharashtra, India
| | - Sanjay Khaladkar
- Professor, Department of Radio-diagnosis, Dr. D. Y. Patil Medical College, Hospital and Research Centre , Pimpri, Pune, Maharashtra, India
| | - Aditi Gujrati
- Resident, Department of Radio-diagnosis, Dr. D. Y. Patil Medical College, Hospital and Research Centre , Pimpri, Pune, Maharashtra, India
| |
Collapse
|
19
|
Webster JD, Yuzbasiyan-Gurkan V, Trosko JE, Chang CC, Kiupel M. Expression of the Embryonic Transcription Factor Oct4 in Canine Neoplasms: A Potential Marker for Stem Cell Subpopulations in Neoplasia. Vet Pathol 2016; 44:893-900. [DOI: 10.1354/vp.44-6-893] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neoplastic cells and stem cells share several phenotypic characteristics. Recently, numerous studies have identified adult stem cells that have been hypothesized to be the cellular origin for cancer in several tissues. Oct4 has been consistently associated with pluripotent or stemlike cells, and it is hypothesized that Oct4 is necessary for the maintenance of pluripotency. We hypothesize that Oct4-positive cells are present in all canine neoplasms and that these subpopulations of neoplastic cells might represent “cancer stem” cells. To test this hypothesis, 83 canine neoplasms representing 21 neoplastic diseases were evaluated for Oct4 expression using immunohistochemistry. The results of this study showed that all tumors included in this study contained a subpopulation of Oct4-positive cells, although the proportion of Oct4-positive cells and the intensity of immunoreactivity varied both within and between tumor types. Subpopulations of Oct4-positive cells identified in these tumors are likely to represent “cancer stem” cells and therefore might be responsible for the maintenance and propagation of the tumors. If these cells represent cancer stem cells, and are therefore responsible for the maintenance and growth of the neoplastic cellular population, then these cells should serve as relevant therapeutic targets and offer the greatest potential for curative treatment.
Collapse
Affiliation(s)
- J. D. Webster
- Comparative Medicine and Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, MI
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI
| | - V. Yuzbasiyan-Gurkan
- Comparative Medicine and Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, MI
- Department of Microbiology and Molecular Genetics, College of Veterinary Medicine, Michigan State University, East Lansing, MI
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI
| | - J. E. Trosko
- Comparative Medicine and Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, MI
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI
| | - C.-C. Chang
- Comparative Medicine and Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, MI
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI
| | - M. Kiupel
- Comparative Medicine and Integrative Biology Program, College of Veterinary Medicine, Michigan State University, East Lansing, MI
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI
| |
Collapse
|
20
|
Zhao Q, Ren H, Feng S, Chi Y, He Y, Yang D, Ma F, Li J, Lu S, Chen F, Xu J, Yang S, Han Z. Aberrant expression and significance of OCT-4A transcription factor in leukemia cells. Blood Cells Mol Dis 2015; 54:90-6. [DOI: 10.1016/j.bcmd.2014.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 07/24/2014] [Indexed: 12/13/2022]
|
21
|
Kaspar HG, Crum CP. The Utility of Immunohistochemistry in the Differential Diagnosis of Gynecologic Disorders. Arch Pathol Lab Med 2015; 139:39-54. [DOI: 10.5858/arpa.2014-0057-ra] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context
Immunohistochemistry has assumed an increasing role in the identification and characterization of gynecologic disorders including lesions with deceptively bland morphology, uncommon and underdiagnosed neoplasms, and neoplasms with specific genetic alterations associated with overexpression or loss of expression of specific proteins. The diagnostic accuracy has been significantly improved owing to the discovery and increasing experience with the tumor-associated biomarkers, and the increasing demand for precise tumor classification to assess suitability for the expanding therapeutic modalities including clinical trials.
Objective
To differentiate lesions of the gynecologic tract through the use of effective immunohistochemical panels.
Data Sources
Literature review and authors' personal practice experience.
Conclusions
The application of diagnostic and prognostic immunohistochemical panels has enabled pathologists to better guide therapeutic decisions and to better predict the clinical outcome. It is now well established that the use of ancillary testing, including immunohistochemistry, has a significant power in the identification, differentiation, and classification of reactive, premalignant, and malignant gynecologic disorders. This article discusses the utilities and pitfalls of the commonly used immunohistochemical markers in the context of overlapping morphologic features encountered in the uterus, ovaries, and fallopian tubes.
Collapse
Affiliation(s)
- Hanna G. Kaspar
- From the Department of Laboratory Medicine, Geisinger Health System, Wilkes-Barre, Pennsylvania (Dr Kaspar)
| | - Christopher P. Crum
- and the Department of Pathology, Division of Women's and Perinatal Pathology, Brigham and Women's Hospital, Boston, Massachusetts (Dr Crum)
| |
Collapse
|
22
|
Zheng YW, Nie YZ, Taniguchi H. Cellular reprogramming and hepatocellular carcinoma development. World J Gastroenterol 2013; 19:8850-8860. [PMID: 24379607 PMCID: PMC3870535 DOI: 10.3748/wjg.v19.i47.8850] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Revised: 11/13/2013] [Accepted: 11/28/2013] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers, and is also the leading cause of death worldwide. Studies have shown that cellular reprogramming contributes to chemotherapy and/or radiotherapy resistance and the recurrence of cancers. In this article, we summarize and discuss the latest findings in the area of cellular reprogramming in HCC. The aberrant expression of transcription factors OCT4, KLF4, SOX2, c-MYC, NANOG, and LIN28 have been also observed, and the expression of these transcription factors is associated with unfavorable clinical outcomes in HCC. Studies indicate that cellular reprogramming may play a critical role in the occurrence and recurrence of HCC. Recent reports have shown that DNA methylation, miRNAs, tumor microenvironment, and signaling pathways can induce the expression of stemness transcription factors, which leads to cellular reprogramming in HCC. Furthermore, studies indicate that therapies based on cellular reprogramming could revolutionize HCC treatment. Finally, a novel therapeutic concept is discussed: reprogramming control therapy. A potential reprogramming control therapy method could be developed based on the reprogramming demonstrated in HCC studies and applied at two opposing levels: differentiation and reprogramming. Our increasing understanding and control of cellular programming should facilitate the exploitation of this novel therapeutic concept and its application in clinical HCC treatment, which may represent a promising strategy in the future that is not restricted to liver cancer.
Collapse
|
23
|
OCT4 promotes tumorigenesis and inhibits apoptosis of cervical cancer cells by miR-125b/BAK1 pathway. Cell Death Dis 2013; 4:e760. [PMID: 23928699 PMCID: PMC3763434 DOI: 10.1038/cddis.2013.272] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 06/15/2013] [Accepted: 06/19/2013] [Indexed: 01/04/2023]
Abstract
Octamer-binding transcription factor 4 (OCT4) is a key regulatory gene that maintains the pluripotency and self-renewal properties of embryonic stem cells. Although there is emerging evidence that it can function as oncogene in several cancers, the role in mediating cervical cancer remains unexplored. Here we found that OCT4 protein expression showed a pattern of gradual increase from normal cervix to cervical carcinoma in situ and then to invasive cervical cancer. Overexpression of OCT4 in two types of cervical cancer cells promotes the carcinogenesis, and inhibits cancer cell apoptosis. OCT4 induces upregulation of miR-125b through directly binding to the promoter of miR-125b-1 confirmed by chromatin immunoprecipitation analysis. MiRNA-125b overexpression suppressed apoptosis and expression of BAK1 protein. In contrast, miR-125b sponge impaired the anti-apoptotic effect of OCT4, along with the upregulated expression of BAK1. Significantly, Luciferase assay showed that the activity of the wild-type BAK1 3'-untranslated region reporter was suppressed and this suppression was diminished when the miR-125b response element was mutated or deleted. In addition, we observed negative correlation between levels of BAK1 and OCT4, and positive between OCT4 and miR-125b in primary cervical cancers. These findings suggest an undescribed regulatory pathway in cervical cancer, by which OCT4 directly induces expression of miR-125b, which inhibits its direct target BAK1, leading to suppression of cervical cancer cell apoptosis.
Collapse
|
24
|
Kraggerud SM, Hoei-Hansen CE, Alagaratnam S, Skotheim RI, Abeler VM, Rajpert-De Meyts E, Lothe RA. Molecular characteristics of malignant ovarian germ cell tumors and comparison with testicular counterparts: implications for pathogenesis. Endocr Rev 2013; 34:339-76. [PMID: 23575763 PMCID: PMC3787935 DOI: 10.1210/er.2012-1045] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This review focuses on the molecular characteristics and development of rare malignant ovarian germ cell tumors (mOGCTs). We provide an overview of the genomic aberrations assessed by ploidy, cytogenetic banding, and comparative genomic hybridization. We summarize and discuss the transcriptome profiles of mRNA and microRNA (miRNA), and biomarkers (DNA methylation, gene mutation, individual protein expression) for each mOGCT histological subtype. Parallels between the origin of mOGCT and their male counterpart testicular GCT (TGCT) are discussed from the perspective of germ cell development, endocrinological influences, and pathogenesis, as is the GCT origin in patients with disorders of sex development. Integrated molecular profiles of the 3 main histological subtypes, dysgerminoma (DG), yolk sac tumor (YST), and immature teratoma (IT), are presented. DGs show genomic aberrations comparable to TGCT. In contrast, the genome profiles of YST and IT are different both from each other and from DG/TGCT. Differences between DG and YST are underlined by their miRNA/mRNA expression patterns, suggesting preferential involvement of the WNT/β-catenin and TGF-β/bone morphogenetic protein signaling pathways among YSTs. Characteristic protein expression patterns are observed in DG, YST and IT. We propose that mOGCT develop through different developmental pathways, including one that is likely shared with TGCT and involves insufficient sexual differentiation of the germ cell niche. The molecular features of the mOGCTs underline their similarity to pluripotent precursor cells (primordial germ cells, PGCs) and other stem cells. This similarity combined with the process of ovary development, explain why mOGCTs present so early in life, and with greater histological complexity, than most somatic solid tumors.
Collapse
Affiliation(s)
- Sigrid Marie Kraggerud
- Department of Cancer Prevention, Institute for Cancer Research, Oslo University Hospital, N-0310 Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
25
|
Rabban JT, Zaloudek CJ. A practical approach to immunohistochemical diagnosis of ovarian germ cell tumours and sex cord-stromal tumours. Histopathology 2012; 62:71-88. [DOI: 10.1111/his.12052] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 10/20/2012] [Indexed: 12/14/2022]
Affiliation(s)
- Joseph T Rabban
- Pathology Department; University of California San Francisco; San Francisco; CA; USA
| | - Charles J Zaloudek
- Pathology Department; University of California San Francisco; San Francisco; CA; USA
| |
Collapse
|
26
|
Samardzija C, Quinn M, Findlay JK, Ahmed N. Attributes of Oct4 in stem cell biology: perspectives on cancer stem cells of the ovary. J Ovarian Res 2012; 5:37. [PMID: 23171809 PMCID: PMC3536609 DOI: 10.1186/1757-2215-5-37] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 10/30/2012] [Indexed: 01/05/2023] Open
Abstract
Epithelial ovarian cancer (EOC) remains the most lethal of all the gynaecological malignancies with drug resistance and recurrence remaining the major therapeutic barrier in the management of the disease. Although several studies have been undertaken to understand the mechanisms responsible for chemoresistance and subsequent recurrence in EOC, the exact mechanisms associated with chemoresistance/recurrence continue to remain elusive. Recent studies have shown that the parallel characteristics commonly seen between embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSC) are also shared by a relatively rare population of cells within tumors that display stem cell-like features. These cells, termed 'cancer initiating cells' or 'cancer stem cells (CSCs)' have been shown not only to display increased self renewal and pluripotent abilities as seen in ESCs and iPSCs, but are also highly tumorigenic in in vivo mouse models. Additionally, these CSCs have been implicated in tumor recurrence and chemoresistance, and when isolated have consistently shown to express the master pluripotency and embryonic stem cell regulating gene Oct4. This article reviews the involvement of Oct4 in cancer progression and chemoresistance, with emphasis on ovarian cancer. Overall, we highlight why ovarian cancer patients, who initially respond to conventional chemotherapy subsequently relapse with recurrent chemoresistant disease that is essentially incurable.
Collapse
Affiliation(s)
- Chantel Samardzija
- Women's Cancer Research Centre, Royal Women's Hospital, 20 Flemington Road, Parkville, VIC, 3052, Australia.
| | | | | | | |
Collapse
|
27
|
Clear cell carcinoma of the female genital tract (not everything is as clear as it seems). Adv Anat Pathol 2012; 19:296-312. [PMID: 22885379 DOI: 10.1097/pap.0b013e31826663b1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Clear cell carcinoma has a storied history in the female genital tract. From the initial designation of ovarian clear cell adenocarcinoma as "mesonephroma" to the linkage between vaginal clear cell carcinoma and diethylstilbestrol exposure in utero, gynecologic tract clear cell tumors have puzzled investigators, posed therapeutic dilemmas for oncologists, and otherwise presented major differential diagnostic challenges for pathologists. One of the most common errors in gynecologic pathology is misdiagnosis of clear cell carcinoma, on both frozen section and permanent section. Given the poor response to platinum-based chemotherapy for advanced-stage disease and increased risk of thromboembolism, accurate diagnosis of clear cell carcinoma is important in the female genital tract. This review (1) presents the clinical and pathologic features of female genital tract clear cell carcinomas; (2) highlights recent molecular developments; (3) identifies areas of potential diagnostic confusion; and (4) presents solutions for these diagnostic problems where they exist.
Collapse
|
28
|
Prevalence of c-KIT mutations in gonadoblastoma and dysgerminomas of patients with disorders of sex development (DSD) and ovarian dysgerminomas. PLoS One 2012; 7:e43952. [PMID: 22937135 PMCID: PMC3429439 DOI: 10.1371/journal.pone.0043952] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 07/27/2012] [Indexed: 11/19/2022] Open
Abstract
Activating c-KIT mutations (exons 11 and 17) are found in 10–40% of testicular seminomas, the majority being missense point mutations (codon 816). Malignant ovarian dysgerminomas represent ∼3% of all ovarian cancers in Western countries, resembling testicular seminomas, regarding chromosomal aberrations and c-KIT mutations. DSD patients with specific Y-sequences have an increased risk for Type II Germ Cell Tumor/Cancer, with gonadoblastoma as precursor progressing to dysgerminoma. Here we present analysis of c-KIT exon 8, 9, 11, 13 and 17, and PDGFRA exon 12, 14 and 18 by conventional sequencing together with mutational analysis of c-KIT codon 816 by a sensitive and specific LightCycler melting curve analysis, confirmed by sequencing. The results are combined with data on TSPY and OCT3/4 expression in a series of 16 DSD patients presenting with gonadoblastoma and dysgerminoma and 15 patients presenting pure ovarian dysgerminomas without DSD. c-KIT codon 816 mutations were detected in five out of the total of 31 cases (all found in pure ovarian dysgerminomas). A synonymous SNP (rs 5578615) was detected in two patients, one DSD patient (with bilateral disease) and one patient with dysgerminoma. Next to these, three codon N822K mutations were detected in the group of 15 pure ovarian dysgerminomas. In total activating c-KIT mutations were found in 53% of ovarian dysgerminomas without DSD. In the group of 16 DSD cases a N505I and D820E mutation was found in a single tumor of a patient with gonadoblastoma and dysgerminoma. No PDGFRA mutations were found. Positive OCT3/4 staining was present in all gonadoblastomas and dysgerminomas investigated, TSPY expression was only seen in the gonadoblastoma/dysgerminoma lesions of the 16 DSD patients. This data supports the existence of two distinct but parallel pathways in the development of dysgerminoma, in which mutational status of c-KIT might parallel the presence of TSPY.
Collapse
|
29
|
Abstract
Ovarian yolk sac tumour (OYST) is a very rare malignancy arising most often in young women. Preoperative clinical, biological (alpha-foetoprotein) and radiological findings should help to establish the diagnosis of OYST, in order to propose adequate surgical treatment. The aim of surgery is to remove the primary tumour, to obtain an accurate histological diagnosis and to assess the disease extent. In young women, fertility-sparing surgery should be performed, in order to preserve the possibility of pregnancy later on. Chemotherapy has substantially modified the prognosis of these tumours, and practically all patients will be cured. The overall 5-year survival rate is 94% when patients are treated with BEP chemotherapy. Depending on the clinical situation, two to four cycles of the BEP regimen should be administered after surgery. Identification of prognostic factors may help to propose risk-adapted treatment in order to increase the cure rate in patients with a poor prognosis and to decrease toxicity in patients with a low risk of relapse. Fertility preservation represents a major objective in women treated for OYSTs.
Collapse
|
30
|
Yokouchi Y, Imaoka M, Sayama A, Sanbuissho A. Mixed germ cell tumor with embryonal carcinoma, choriocarcinoma, and epithelioid trophoblastic tumor in the ovary of a cynomolgus monkey. Toxicol Pathol 2011; 39:553-8. [PMID: 21519041 DOI: 10.1177/0192623311399787] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A seven-year-old female cynomolgus monkey had a mass in the left ovary with metastasis to the lung and the right ovary. The mass of these organs showed three different characteristics, and its immunohistochemical profiles were consistent with embryonal carcinoma (EC), choriocarcinoma (CC), and epithelioid trophoblastic tumor (ETT). The EC was characterized with sheets and glandlike structures with large pleomorphic, single-nucleated epithelial cells that were immunohistochemically positive for α-fetoprotein, octamer-4, and CD30, and with multinucleated giant cells resembling syncytiotrophoblasts. The CC also represented biphasic proliferation of the cytotrophoblast positive for cytokeratin 7 (CK7), which showed negative immunoreactivity for all three of the above antibodies, and it was syncytiotrophoblast positive for human chorionic gonadotropin. The ETT showed numerous floating cells in an abundant eosinophilic extracellular matrix with vacuolated or eosinophilic cytoplasm and was immunohistochemically positive for CK7, p63, and α-inhibin, which features nodule or cordlike structures. Collectively, this neoplasm was identified as a mixed germ cell tumor with EC, CC, and ETT. To our knowledge, this is the first report of EC in nonhuman primates as a component of mixed germ cell tumor.
Collapse
Affiliation(s)
- Y Yokouchi
- Daiichi-Sankyo Co., Ltd., Medicinal Safety Research Laboratories, Tokyo, Japan.
| | | | | | | |
Collapse
|
31
|
O’Connor WG, Bianchi GD, Willis MS, McCudden CR. Testicular Mass in a 40-Year-Old Man. Lab Med 2011. [DOI: 10.1309/lm0qfesymx42nlxb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
32
|
Zhou X, Huang GR, Hu P. Over-expression of Oct4 in human esophageal squamous cell carcinoma. Mol Cells 2011; 32:39-45. [PMID: 21547540 PMCID: PMC3887652 DOI: 10.1007/s10059-011-2315-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 04/17/2011] [Accepted: 04/21/2011] [Indexed: 12/11/2022] Open
Abstract
The Octamer 4 gene (Oct4) is a master pluripotency controller that has been detected in several types of tumors. Here, we examine the expression of Oct4 in human esophageal squamous cell carcinoma (ESCC). We found that punctate Oct4 protein was expressed in most (93.7%) ESCC samples but it was not observed in esophageal mucosa. Some ESCC cells had the capacity to form tumorospheres; those with an Oct4(+)-rich cell phenotype had increased proliferation and Oct4 mRNA levels compared to those of differentiated cells in culture or xenograft tumors. The over-expression of Oct4 in ESCCs suggests that it is a potential target for ESCC therapy. Oct4 could be a useful tumor marker in an immunohistochemical panel designed to differentiate between ESCC and esophageal mucosa. Expression of Oct4 in tumorospheres might indicate the presence of a population of ECSCs and its expression in xenograft tumors suggests that Oct4 is also associated with tumor metastasis.
Collapse
Affiliation(s)
- Xi Zhou
- Renmin Hospital, Hubei Medical University, Shiyan, China.
| | | | | |
Collapse
|
33
|
Gillis AJM, Stoop H, Biermann K, van Gurp RJHLM, Swartzman E, Cribbes S, Ferlinz A, Shannon M, Oosterhuis JW, Looijenga LHJ. Expression and interdependencies of pluripotency factors LIN28, OCT3/4, NANOG and SOX2 in human testicular germ cells and tumours of the testis. ACTA ACUST UNITED AC 2011; 34:e160-74. [PMID: 21631526 DOI: 10.1111/j.1365-2605.2011.01148.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OCT3/4, NANOG, SOX2 and, most recently, LIN28 have been identified as key regulators of pluripotency in mammalian embryonic and induced stem cells, and are proven to be crucial for generation of the mouse germ-cell lineage. These factors are a hallmark of certain histological types of germ-cell tumours (GCTs). Here, we report novel information on the temporal and spatial expression pattern of LIN28 during normal human male germ-cell development as well as various types of GCTs. To investigate LIN28 expression, immunohistochemical analyses and quantitative proximity ligation assay-based TaqMan protein assays were applied on snap-frozen and formalin-fixed, paraffin-embedded samples as well as representative cell lines. LIN28 was found in primordial germ cells, gonocytes and pre-spermatogonia, in contrast to OCT3/4 and NANOG, which were found only in the first two stages. LIN28 was also found in all precursor lesions (carcinoma in situ and gonadoblastoma) of type II GCTs, as well as the invasive components seminoma and the non-seminomatous elements embryonal carcinoma and yolk sac tumour. Choriocarcinoma showed a heterogeneous pattern, while teratomas and spermatocytic seminomas (type III GCTs) were negative. This expression pattern suggests that LIN28 is associated with malignant behaviour of type II GCTs. Cell line experiments involving siRNA knockdown of LIN28, OCT3/4 and SOX2 showed that LIN28 plays a role in the maintenance of the undifferentiated state of both seminoma and embryonal carcinoma, closely linked to, and likely upstream of OCT3/4 and NANOG. In conclusion, LIN28 regulates the differentiation status of seminoma and embryonal carcinoma and is likely to play a related role in normal human germ-cell development.
Collapse
Affiliation(s)
- A J M Gillis
- Department of Pathology, Erasmus MC-University Medical Center Rotterdam, Josephine Nefkens Institute, Daniel den Hoed Cancer Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Byskov AG, Høyer PE, Yding Andersen C, Kristensen SG, Jespersen A, Møllgård K. No evidence for the presence of oogonia in the human ovary after their final clearance during the first two years of life. Hum Reprod 2011; 26:2129-39. [PMID: 21572085 DOI: 10.1093/humrep/der145] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Conflicting results of studies on mouse and human have either verified or refuted the presence of oogonia/primordial germ cells in the post-natal ovary. The aim of this study was to trace whether oogonia recognized by immunohistochemical methods in the first trimester human ovary were present also in peri- and post-natal ovaries. METHODS For this study, 82 human ovaries were collected: 25 from embryos from 5 to 10 weeks post conception (wpc), 2 at 18 wpc, 32 from 32 wpc to 2 years and 23 from 2 to 32 years. Of these, 80 ovaries were fixed and paraffin-embedded and 2 (8 year-old) ovaries were processed for plastic sections. Serial sections were prepared for immunohistochemical detection of markers for oogonia: tyrosine kinase receptor for stem cell factor (SCF)(C-KIT), stage-specific embryonic antigen-4 (SSEA4), homeobox gene transcription factor (NANOG), octamer binding transcription factor 4 (OCT4) and melanoma antigen-4 (Mage-A4), while noting that C-KIT also stains diplotene oocytes. RESULTS Almost all oogonia exclusively stained for SSEA4, NANOG, OCT4 and C-KIT, whereas MAGE-A4 only stained a small fraction. At birth only a few oogonia were stained. These disappeared before 2 years, leaving only diplotene oocytes stained for C-KIT. From 18 wpc to 2 years, the medulla contained conglomerates of healthy and degenerating oogonia and small follicles, waste baskets (WBs) and oogonia enclosed in growing follicles (FWB). Medulla of older ovaries contained groups of primordial, healthy follicles. CONCLUSIONS We found no evidence for the presence of oogonia in the human ovary after their final clearing during the first 2 years. We suggest that perinatal medullary WB and FWB give rise to the groups of small, healthy follicles in the medulla.
Collapse
Affiliation(s)
- A G Byskov
- Laboratory of Reproductive Biology, Section 5712, University Hospital of Copenhagen, Rigshospitalet, Section 5712, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
35
|
Strunk A, Imai DM, Osofsky A, Tell LA. Dysgerminoma in an eastern rosella (Platycercus eximius eximius). Avian Dis 2011; 55:133-8. [PMID: 21500650 DOI: 10.1637/9502-081510-case.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This report describes a case of dysgerminoma in a 21-year-old eastern rosella (Platycercus eximius eximius) that presented with dyspnea and a severely distended coelom. The bird was euthanatized, and a large, left-sided coelomic mass was identified. Microscopically, the mass was composed of sheets and nests of round to polygonal neoplastic cells with lacy cytoplasm. The neoplastic cells were weakly positive for vimentin and c-kit but negative for pancytokeratin, AE1, and inhibin. On the basis of the histomorphology and immunoreactivity, the neoplasm was determined to be a dysgerminoma. The variability of histologic appearance and immunohistochemical staining of dysgerminomas in humans compared with veterinary species is discussed.
Collapse
Affiliation(s)
- Anneliese Strunk
- Research Boulevard Pet and Bird Hospital, 11679 Research Boulevard, Austin, TX 78757, USA.
| | | | | | | |
Collapse
|
36
|
Cheng L, Roth LM, Zhang S, Wang M, Morton MJ, Zheng W, Abdul Karim FW, Montironi R, Lopez-Beltran A. KIT gene mutation and amplification in dysgerminoma of the ovary. Cancer 2010; 117:2096-103. [DOI: 10.1002/cncr.25794] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 10/09/2010] [Accepted: 10/15/2010] [Indexed: 01/04/2023]
|
37
|
Emerson RE, Ulbright TM. Intratubular germ cell neoplasia of the testis and its associated cancers: the use of novel biomarkers. Pathology 2010; 42:344-55. [PMID: 20438407 DOI: 10.3109/00313021003767355] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Recent advances in the understanding of the molecular pathology of testicular tumours have led to the identification of several new immunohistochemical markers for invasive and in situ germ cell neoplasms. OCT3/4 and NANOG are nuclear stains that have high sensitivity and specificity for the identification of intratubular germ cell neoplasia as well as seminoma and embryonal carcinoma. A potential pitfall in their application to the detection of intratubular germ cell neoplasia, as in other markers that represent oncofetal antigens, is their expression in non-neoplastic germ cells with 'delayed maturation'. SALL4, another nuclear stain, is positive for most germ cell tumours as a group and may be especially helpful in the distinction of these tumours from somatic carcinomas in non-testicular sites. Glypican 3 is a more sensitive marker for yolk sac tumour than alpha-fetoprotein. SOX2 and SOX17 may be useful for differentiating seminoma and embryonal carcinoma, especially following chemotherapy as embryonal carcinoma may lose CD30 expression in this setting. This article reviews the application of these immunohistochemical markers and others to the diagnosis of germ cell neoplasia with reference to older immunohistochemical stains when appropriate. Suggested immunohistochemical panels are described for individual tumour types.
Collapse
Affiliation(s)
- Robert E Emerson
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| | | |
Collapse
|
38
|
Liu A, Cheng L, Du J, Peng Y, Allan RW, Wei L, Li J, Cao D. Diagnostic utility of novel stem cell markers SALL4, OCT4, NANOG, SOX2, UTF1, and TCL1 in primary mediastinal germ cell tumors. Am J Surg Pathol 2010; 34:697-706. [PMID: 20410807 DOI: 10.1097/pas.0b013e3181db84aa] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Primary mediastinal germ cell tumors (GCTs) are rare and sometimes they pose diagnostic difficulty without immunohistochemical studies. Here, we investigated the diagnostic utility of 6 stem cell markers (SCMs) SALL4, OCT4, NANOG, SOX2, UTF1, and TCL1 in 16 primary mediastinal seminomas, 3 embryonal carcinomas (ECs), 10 yolk sac tumors (YSTs), 7 teratomas (4 mature, 3 immature), and 1 choriocarcinoma. The percentage of tumor cells stained was scored as: 0 (no tumor cell staining), 1+ (< or =30%), 2+ (31% to 60%), 3+ (61% to 90%), and 4+ (>90%). The staining intensity of SCMs was scored as weak, moderate, or strong. We also compared them with currently used GCT markers placental-like alkaline phosphatase (PLAP), alpha-fetoprotein (AFP), c-KIT, CD30, and glypican-3. All 16 seminomas showed staining for SALL4 (4+ in 15, 2+ in 1) (15 strong, 1 moderate), OCT4 (4+ in 11, 3+ in 4, 2+ in 1) (13 strong, 3 moderate), and UTF1 (4+ in 13, 3+ in 2, 2+ in 1) (7 strong, 5 moderate, 4 weak). Positive staining was shown by 9/9 seminomas tested for NANOG (4+ in 7, 2+ in 2) (8 strong, 1 weak), TCL1 (4+ strong in all), c-KIT (4+ in all), and PLAP (4+ in 5, 3+ in 1, 2+ in 2, 1+ in 1), but SOX2 staining was negative in all these tumors. All 3 ECs showed 4+ strong staining for SALL4, OCT4, and UTF1 but negative for TCL1. SOX2 staining was seen in 3/3 ECs (4+ strong in 1, 3+ weak to moderate in 2) whereas NANOG staining was seen in 2/3 ECs (2+ weak, 1+ moderate). CD30 staining was seen in 3/3 ECs (1+, 2+, 4+). Strong SALL4 staining was seen in 10/10 YSTs (4+ in 9, 2+ in 1). All 10 YSTs showed AFP (1+ in 7, 2+ in 1, 3+ in 2) and glypican-3 (1+ in 3, 2+ in 1, 3+ in 5, 4+ in 1) staining but only 4/10 YSTs showed PLAP staining (1+ in all 4). The mean percentage of YST cells stained with SALL4 was 92%, whereas it was 23% for AFP, 50% for glypican-3, and 4% for PLAP (P<0.01). Focal (1+) SALL4 (weak) and SOX2 (weak to moderate) staining was seen in 2/7 and 4/7 teratomas, respectively. The choriocarcinoma was negative for all 6 SCMs. Eleven thymomas and 6 thymic carcinomas were negative for 6 SCMs. No staining of NANOG and SOX2 was seen in 20 lymphomas (5 Hodgkin, 5 large B cell, 5 lymphoblastic, 5 anaplastic large cell) (other 4 SCMs in lymphomas earlier studied). Our study indicates that SALL4, OCT4, NANOG, SOX2, UTF1, and TLC1 are novel sensitive diagnostic markers for primary mediastinal GCTs, with high specificity. Of these 6 SCMs, SALL4 is the only 1 expressed in YST. These novel SCMs are more sensitive than the currently used markers for mediastinal GCTs.
Collapse
Affiliation(s)
- Aijun Liu
- Department of Pathology, The Chinese PLA General Hospital, Beijing Hospital, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Cheng L, Zhang S, Talerman A, Roth LM. Morphologic, immunohistochemical, and fluorescence in situ hybridization study of ovarian embryonal carcinoma with comparison to solid variant of yolk sac tumor and immature teratoma. Hum Pathol 2010; 41:716-23. [PMID: 20096442 DOI: 10.1016/j.humpath.2009.10.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 10/21/2009] [Accepted: 10/23/2009] [Indexed: 02/01/2023]
Abstract
The prognosis and therapy for malignant ovarian germ cell tumors including embryonal carcinoma differ from those of other categories of ovarian tumors, making accurate diagnosis imperative for patient care. Because of its rarity, the protein markers and genomic alterations typifying primary ovarian embryonal carcinoma have not been fully characterized. The present study aims to establish a set of sensitive and specific protein markers useful for the diagnosis and delineation of ovarian embryonal carcinoma. Chromosomal 12p anomalies were analyzed by dual-color fluorescence in situ hybridization. In a series of 6 ovarian mixed germ cell tumors with a component of embryonal carcinomas, OCT4, CD30, SOX2, and glypican 3 expressions were analyzed immunohistochemically on formalin-fixed paraffin-embedded tissue specimens. The results were compared to 4 cases of mixed germ cell tumor that were originally mistaken for embryonal carcinoma. OCT4 marked the nuclei of 6 cases, among which 5 cases also showed glypican 3 expression indicative of an admixed yolk sac tumor component. SOX2 was positive in only 3 cases of embryonal carcinoma. In 1 case of mixed germ cell tumor containing embryonal carcinoma, embryoid bodies from a component of polyembryoma were demonstrated to be both OCT4 and CD30 positive. Two cases originally classified as embryonal carcinoma were OCT4 and CD30 negative and showed glypican 3 positivity. They were reclassified as solid variant of yolk sac tumor. Two other cases originally classified as embryonal carcinoma were OCT4 positive and CD30 negative and were classified as immature teratoma with neuroectodermal differentiation based on the immunohistochemical findings as well as morphologic features and were diagnosed as immature teratoma. Chromosome 12p alterations were identified in 5 of 6 cases of embryonal carcinomas. In summary, a panel of immunostains is more useful than a single biomarker in the differential diagnosis of ovarian germ cell tumors. Chromosome 12p fluorescence in situ hybridization combined with OCT4, CD30, and glypican 3 immunostains is useful in confirming the diagnosis of ovarian embryonal carcinoma.
Collapse
Affiliation(s)
- Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | |
Collapse
|
40
|
Cao D, Guo S, Allan RW, Molberg KH, Peng Y. SALL4 is a novel sensitive and specific marker of ovarian primitive germ cell tumors and is particularly useful in distinguishing yolk sac tumor from clear cell carcinoma. Am J Surg Pathol 2009; 33:894-904. [PMID: 19295406 DOI: 10.1097/pas.0b013e318198177d] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ovarian primitive germ cell tumors (GCTs) are uncommon tumors and sometimes pose diagnostic challenges. Among them, yolk sac tumor (YST) poses the greatest diagnostic difficulty and can be mistaken for clear cell carcinoma (CCC). Current immunohistochemical markers such as alpha-fetoprotein (AFP), glypican-3, cytokeratin (CK) 7, and epithelial membrane antigen (EMA) used to distinguish YST from CCC lack adequate sensitivity and specificity. Here by immunohistochemistry, we investigated a novel marker SALL4 in 98 GCTs (29 YSTs, 18 dysgerminomas, 6 gonadoblastomas, 6 embryonal carcinomas, 15 immature and 12 mature teratomas, 7 carcinoid tumors, 3 strumal carcinoids, and 2 struma ovarii) with particular interest of exploring SALL4 to distinguish YST from CCC. One hundred sixty-three non-GCTs including 45 CCCs were also stained. We found that SALL4 is strongly positive in more than 90% tumor cells in all YSTs, dysgerminomas, gonadoblastomas, and embryonal carcinomas. Variable SALL4 staining is seen in 11 of 15 immature teratomas. All other GCTs included in this study are negative for SALL4. Except 3 CCCs with focal SALL4 staining (<15% tumor cells), SALL4 is negative in the remaining 160 non-GCTs. We also compared SALL4 with AFP, glypican-3, CK7, and EMA in all YSTs and CCCs. AFP and glypican-3 are positive in 24 (83%) and 20 (69%) YSTs, respectively, whereas 16 (35%) and 13(28%) CCCs show positive AFP and glypican-3 staining, respectively. Three (10%) and 4 (14%) YSTs show focal (<2% tumor cells) CK7 and EMA staining, respectively. CK7 and EMA are positive in all 45 CCCs but 3 (7%) and 1 (2%) cases show staining in less than 30% tumor cells, respectively. Our findings indicate that SALL4 is a novel sensitive and specific marker for ovarian primitive GCTs. SALL4 is particularly useful in distinguishing YST from CCC and better than AFP, glypican-3, CK7, and EMA.
Collapse
Affiliation(s)
- Dengfeng Cao
- The Lauren V. Ackerman Laboratory of Surgical Pathology, Division of Anatomic and Molecular Pathology, Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
41
|
Chen Z, Xu WR, Qian H, Zhu W, Bu XF, Wang S, Yan YM, Mao F, Gu HB, Cao HL, Xu XJ. Oct4, a novel marker for human gastric cancer. J Surg Oncol 2009; 99:414-9. [PMID: 19347886 DOI: 10.1002/jso.21270] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND OBJECTIVE Octamer-4 (Oct4), a transcription factor involved in regulating human embryonic stem cells (ESCs), may play a role in tumorigenesis. Since little is known about the efficacy of Oct4 as a potential biomarker for gastric cancer (GC), we investigated its expression in GC tissues and its relationship to various clinicopathological parameters. METHODS Primary tumor tissues and matching, adjacent non-cancerous tissues were obtained from 62 GC patients, and Oct4 expression was examined by reverse transcription-PCR (RT-PCR) and real-time PCR. Twenty biopsy specimens of atrophic gastritis and gastric ulcer individually were collected as control. To detect Oct4 expression in the paired GC and non-cancerous tissues at the protein level, Western blotting and immunohistochemistry (IHC) were employed. Correlation analyses were conducted to assess the relationship between Oct4 expression and clinicopathological parameters. RESULTS Oct4 expression levels were higher in GC tissues compared to matching, adjacent non-cancerous tissues, atrophic gastritis and gastric ulcer tissues. Additionally, Oct4 expression in GC tumors correlated with their differentiation status, but not with patient age or gender, tumor size, TNM stage, depth of invasion, or the presence of lymph node metastasis. CONCLUSIONS Oct4 may be a potential biomarker for the initiation, progression, and differentiation of human GC.
Collapse
Affiliation(s)
- Zhong Chen
- School of Medical Science and Laboratory Medicine, Key Institute of Clinical Laboratory Science, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Yu CH, Hwang DN, Yhee JY, Kim JH, Im KS, Nho WG, Lyoo YS, Sur JH. Comparative immunohistochemical characterization of canine seminomas and Sertoli cell tumors. J Vet Sci 2009; 10:1-7. [PMID: 19255517 PMCID: PMC2801099 DOI: 10.4142/jvs.2009.10.1.1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Primary testicular tumors are the most common causes of cancer in male dogs. Overall, the majority of canine patients should be cured by testicular surgery. However, tumor markers are not well-known in veterinary medicine. We sought to determine using immunohistochemistry whether the combined human testicular tumor markers (placental alkaline phosphatase, OCT3/4, CD30, alpha-fetoprotein, inhibin-alpha, vimentin, c-KIT, and desmin) are expressed in canine seminomas and Sertoli cell tumors (SCTs). We examined 35 canine testicular tumors, 20 seminomas and 15 SCTs. c-KIT was expressed markedly in canine seminomas. Both inhibin-alpha and vimentin were expressed significantly in canine SCTs. The results of this study demonstrate differences and similarities between tumor marker expression of testicular tumors in dogs and humans. All the main markers in current routine use are discussed as well as potential useful markers for benign and malignant tumors, and tumor progression.
Collapse
Affiliation(s)
- Chi-Ho Yu
- Department of Veterinary Pathobiology, Small Animal Tumor Diagnostic Center, College of Veterinary Medicine, Konkuk University, Seoul 143-701, Korea
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Sturgeon CM, Duffy MJ, Stenman UH, Lilja H, Brünner N, Chan DW, Babaian R, Bast RC, Dowell B, Esteva FJ, Haglund C, Harbeck N, Hayes DF, Holten-Andersen M, Klee GG, Lamerz R, Looijenga LH, Molina R, Nielsen HJ, Rittenhouse H, Semjonow A, Shih IM, Sibley P, Sölétormos G, Stephan C, Sokoll L, Hoffman BR, Diamandis EP. National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines for Use of Tumor Markers in Testicular, Prostate, Colorectal, Breast, and Ovarian Cancers. Clin Chem 2008; 54:e11-79. [DOI: 10.1373/clinchem.2008.105601] [Citation(s) in RCA: 458] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Abstract
Background: Updated National Academy of Clinical Biochemistry (NACB) Laboratory Medicine Practice Guidelines for the use of tumor markers in the clinic have been developed.
Methods: Published reports relevant to use of tumor markers for 5 cancer sites—testicular, prostate, colorectal, breast, and ovarian—were critically reviewed.
Results: For testicular cancer, α-fetoprotein, human chorionic gonadotropin, and lactate dehydrogenase are recommended for diagnosis/case finding, staging, prognosis determination, recurrence detection, and therapy monitoring. α-Fetoprotein is also recommended for differential diagnosis of nonseminomatous and seminomatous germ cell tumors. Prostate-specific antigen (PSA) is not recommended for prostate cancer screening, but may be used for detecting disease recurrence and monitoring therapy. Free PSA measurement data are useful for distinguishing malignant from benign prostatic disease when total PSA is <10 μg/L. In colorectal cancer, carcinoembryonic antigen is recommended (with some caveats) for prognosis determination, postoperative surveillance, and therapy monitoring in advanced disease. Fecal occult blood testing may be used for screening asymptomatic adults 50 years or older. For breast cancer, estrogen and progesterone receptors are mandatory for predicting response to hormone therapy, human epidermal growth factor receptor-2 measurement is mandatory for predicting response to trastuzumab, and urokinase plasminogen activator/plasminogen activator inhibitor 1 may be used for determining prognosis in lymph node–negative patients. CA15-3/BR27–29 or carcinoembryonic antigen may be used for therapy monitoring in advanced disease. CA125 is recommended (with transvaginal ultrasound) for early detection of ovarian cancer in women at high risk for this disease. CA125 is also recommended for differential diagnosis of suspicious pelvic masses in postmenopausal women, as well as for detection of recurrence, monitoring of therapy, and determination of prognosis in women with ovarian cancer.
Conclusions: Implementation of these recommendations should encourage optimal use of tumor markers.
Collapse
Affiliation(s)
- Catharine M Sturgeon
- Department of Clinical Biochemistry, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Michael J Duffy
- Department of Pathology and Laboratory Medicine, St Vincent’s University Hospital and UCD School of Medicine and Medical Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Ulf-Håkan Stenman
- Department of Clinical Chemistry, Helsinki University Central Hospital, Helsinki, Finland
| | - Hans Lilja
- Departments of Clinical Laboratories, Urology, and Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Nils Brünner
- Section of Biomedicine, Department of Veterinary Pathobiology, Faculty of Life Sciences, University of Copenhagen, Denmark
| | - Daniel W Chan
- Departments of Pathology and Oncology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Richard Babaian
- Department of Urology, The University of Texas Anderson Cancer Center, Houston, TX
| | - Robert C Bast
- Department of Experimental Therapeutics, University of Texas Anderson Cancer Center, Houston, Texas, USA
| | | | - Francisco J Esteva
- Departments of Breast Medical Oncology, Molecular and Cellular Oncology, University of Texas M.D. Anderson Cancer Center, Houston TX
| | - Caj Haglund
- Department of Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Nadia Harbeck
- Frauenklinik der Technischen Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Daniel F Hayes
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Mads Holten-Andersen
- Section of Biomedicine, Department of Veterinary Pathobiology, Faculty of Life Sciences, University of Copenhagen, Denmark
| | - George G Klee
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN
| | - Rolf Lamerz
- Department of Medicine, Klinikum of the University of Munich, Grosshadern, Germany
| | - Leendert H Looijenga
- Laboratory of Experimental Patho-Oncology, Erasmus MC-University Medical Center Rotterdam, and Daniel den Hoed Cancer Center, Rotterdam, the Netherlands
| | - Rafael Molina
- Laboratory of Biochemistry, Hospital Clinico Provincial, Barcelona, Spain
| | - Hans Jørgen Nielsen
- Department of Surgical Gastroenterology, Hvidovre Hospital, Copenhagen, Denmark
| | | | - Axel Semjonow
- Prostate Center, Department of Urology, University Clinic Muenster, Muenster, Germany
| | - Ie-Ming Shih
- Departments of Pathology and Oncology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Paul Sibley
- Siemens Medical Solutions Diagnostics, Glyn Rhonwy, Llanberis, Gwynedd, UK
| | | | - Carsten Stephan
- Department of Urology, Charité Hospital, Universitätsmedizin Berlin, Berlin, Germany
| | - Lori Sokoll
- Departments of Pathology and Oncology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Barry R Hoffman
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - Eleftherios P Diamandis
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | | |
Collapse
|
44
|
|
45
|
de Jong J, Stoop H, Gillis AJM, van Gurp RJHLM, van de Geijn GJM, Boer MD, Hersmus R, Saunders PTK, Anderson RA, Oosterhuis JW, Looijenga LHJ. Differential expression of SOX17 and SOX2 in germ cells and stem cells has biological and clinical implications. J Pathol 2008; 215:21-30. [PMID: 18348160 DOI: 10.1002/path.2332] [Citation(s) in RCA: 160] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Combined action of SOX and POU families of transcription factors plays major roles in embryonic development. In embryonic stem cells, the combination of SOX2 and POU5F1 (OCT3/4) is essential for maintaining the undifferentiated state by activating pluripotency-linked genes, and inhibition of genes involved in differentiation. Besides embryonic stem cells, POU5F1 is also present in early germ cells, primordial germ cells, and gonocytes, where it has a role in suppression of apoptosis. Here we demonstrate that SOX2 is absent in germ cells of human fetal gonads, and as expected carcinoma in situ (CIS), ie the precursor lesion of testicular germ cell tumours of adolescents and adults (TGCTs), and seminoma. Based on genome-wide expression profiling, SOX17 was found to be present, instead of SOX2, in early germ cells and their malignant counterparts, CIS and seminoma. Immunohistochemistry, western blot analysis, and quantitative RT-PCR showed that SOX17 is a suitable marker to distinguish seminoma from embryonal carcinoma, confirmed in representative cell lines. Aberrant SOX2 expression can be present in Sertoli cells when associated with CIS, which can be misdiagnosed as embryonal carcinoma. In conclusion, this study demonstrates the absence of SOX2 in human embryonic and malignant germ cells, which express SOX17 in conjunction with POU5F1. This finding has both diagnostic and developmental biological implications. It allows the identification of seminoma-like cells from embryonal carcinoma based on a positive marker and might be the explanation for the different function of POU5F1 in normal and malignant germ cells versus embryonic stem cells.
Collapse
Affiliation(s)
- J de Jong
- Department of Pathology, Erasmus MC-University Medical Center Rotterdam, Josephine Nefkens Institute, Daniel den Hoed Cancer Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Sung MT, Zhang S, MacLennan GT, Lopez-Beltran A, Montironi R, Wang M, Tan PH, Cheng L. Histogenesis of clear cell adenocarcinoma in the urinary tract: evidence of urothelial origin. Clin Cancer Res 2008; 14:1947-55. [PMID: 18381932 DOI: 10.1158/1078-0432.ccr-07-4147] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Clear cell adenocarcinoma in the urinary tract is a rare entity with an appearance resembling its counterpart in the female genital tract. Although several theories have been proposed about its origin, its exact histogenesis has remained uncertain. EXPERIMENTAL DESIGN We integrated molecular genetic evaluation by fluorescence in situ hybridization and X-chromosome inactivation with conventional morphologic and immunohistochemical analyses in 12 patients with clear cell adenocarcinomas in the urinary tract. RESULTS Concurrent urothelial carcinoma or urothelial carcinoma in situ was present in six cases (50%) and foci of cystitis glandularis were observed in four cases (33%). Neither intestinal metaplasia nor Müllerian component was identified in any case. Cytoplasmic expression of alpha-methylacyl-CoA racemase was demonstrable in 10 of 12 tumors (83%). Moderate to diffuse immunostaining for cytokeratin 7 was identified in all 12 tumors (100%), whereas only 3 of 12 (25%) tumors showed positive immunostaining for cytokeratin 20. Focal uroplakin III staining was seen in 6 of 12 tumors (50%). In five cases (42%), focal to moderate CD10 immunoreactivity was observed. Immunostains for OCT4 and CDX2 were completely negative in all tumors. In UroVysion fluorescence in situ hybridization assays, all tumors displayed chromosomal alterations similar to those commonly found in urothelial carcinoma. Identical patterns of nonrandom X-chromosome inactivation in concurrent clear cell adenocarcinoma and urothelial neoplasia were identified in two informative female cases. CONCLUSIONS Our findings support an urothelial origin for most clear cell adenocarcinomas of the urinary tract, despite their morphologic resemblance to certain Müllerian-derived tumors of the female genital tract.
Collapse
Affiliation(s)
- Ming-Tse Sung
- Department of Pathology, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Disgerminoma «anaplásico» de ovario en una mujer de 23 años de edad. Estudio inmunohistoquímico de un caso, con énfasis en el diagnóstico diferencial. CLINICA E INVESTIGACION EN GINECOLOGIA Y OBSTETRICIA 2008. [DOI: 10.1016/s0210-573x(08)73055-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
48
|
Panagopoulos I, Möller E, Isaksson M, Mertens F. A PCR/restriction digestion assay for the detection of the transcript variants 1 and 2 ofPOU5F1. Genes Chromosomes Cancer 2008; 47:521-9. [DOI: 10.1002/gcc.20555] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
49
|
Hombach-Klonisch S, Paranjothy T, Wiechec E, Pocar P, Mustafa T, Seifert A, Zahl C, Gerlach KL, Biermann K, Steger K, Hoang-Vu C, Schulze-Osthoff K, Los M. Cancer stem cells as targets for cancer therapy: selected cancers as examples. Arch Immunol Ther Exp (Warsz) 2008; 56:165-80. [PMID: 18512024 DOI: 10.1007/s00005-008-0023-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 05/17/2008] [Indexed: 12/16/2022]
Abstract
It is becoming increasingly evident that cancer constitutes a group of diseases involving altered stem-cell maturation/differentiation and the disturbance of regenerative processes. The observed malignant transformation is merely a symptom of normal differentiation processes gone astray rather than the primary event. This review focuses on the role of cancer stem cells (CSCs) in three common but also relatively under-investigated cancers: head and neck, ovarian, and testicular cancer. For didactic purpose, the physiology of stem cells is first introduced using hematopoietic and mesenchymal stem cells as examples. This is followed by a discussion of the (possible) role of CSCs in head and neck, ovarian, and testicular cancer. Aside from basic information about the pathophysiology of these cancers, current research results focused on the discovery of molecular markers specific to these cancers are also discussed. The last part of the review is largely dedicated to signaling pathways active within various normal and CSC types (e.g. Nanog, Nestin, Notch1, Notch2, Oct3 and 4, Wnt). Different elements of these pathways are also discussed in the context of therapeutic opportunities for the development of targeted therapies aimed at CSCs. Finally, alternative targeted anticancer therapies arising from recently identified molecules with cancer-(semi-)selective capabilities (e.g. apoptin, Brevinin-2R) are considered.
Collapse
|
50
|
Pectasides D, Pectasides E, Kassanos D. Germ cell tumors of the ovary. Cancer Treat Rev 2008; 34:427-41. [PMID: 18378402 DOI: 10.1016/j.ctrv.2008.02.002] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 02/07/2008] [Accepted: 02/14/2008] [Indexed: 11/16/2022]
Abstract
PURPOSE Malignant ovarian germ cell tumors (MOGCTS) are rare but curable at all stages of disease. This review gives an outline of the management of this disease. METHODS We performed a literature search in the PubMed of almost all relevant articles concerning MOGCTs on pathology, prognostic factors, surgery, post-operative therapy and late effects of therapy. The available literature is mainly composed of retrospective reviews and articles. RESULTS Prognostic factors include stage, amount of residual tumor, histologic type and raised tumor markers. For patients with early stage disease, cure rates approach 100%, while for those with advanced-stage disease are at least 75%. Appropriate surgical treatment for patients where fertility needs to be preserved consists in laparotomy with unilateral salpingo-oophorectomy (USO) and resection of all visible disease. For patients with advanced-stage disease, the role and the extent of debulking surgery remain controversial despite its routine use. However, it is suggested a benefit from minimal residual disease at completion of primary surgical cytoreduction with both non-platinum and platinum-based chemotherapy regimens. Second-look surgery clearly is not indicated in patients with early stage non-dysgerminoma or in all patients with dysgerminoma. However, teratoma patients may benefit from secondary cytoreduction. Three courses of bleomycin, etoposide and cisplatin (BEP) is the current standard adjuvant chemotherapy and four courses of BEP are recommended in case of bulky residual tumor after surgery. More evidence is required to show that surveillance is a safe option. There is a hint that high-dose chemotherapy may play a role in relapsed patients. The majority of MOGCTs patients who undergo fertility-sparing surgery and chemotherapy retain their gonadal and reproductive function. There is an increasing concern about life-threatening long-term effects of treatment. CONCLUSION MOGCTs are rare neoplasms that affect girls and young women and have excellent prognosis at all stages of disease with optimal therapy. The majority of MOGCTs patients retain their reproductive function.
Collapse
Affiliation(s)
- D Pectasides
- Second Department of Internal Medicine, Propaeduetic, Oncology Section, University of Athens, "Attikon" University Hospital, Haidari, 1 Rimini, Athens, Greece.
| | | | | |
Collapse
|