1
|
Weil PP, Pembaur A, Wirth B, Oetjen E, Büsscher H, Zirngibl K, Czarnetzki M, Braun S, Cremers JF, Gödde D, Degener S, Postberg J. Histone variant H3.5 in testicular cell differentiation and its interactions with histone chaperones. Sci Rep 2024; 14:30564. [PMID: 39702777 DOI: 10.1038/s41598-024-83206-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
Testicular cell differentiation is a highly regulated process, essential for male reproductive health. The histone variant H3.5 is apparently a critical player in this intricate orchestra of cell types, but its regulation and function remains poorly understood. To elucidate its role, we fractionized testicular cells using c-Kit/CD117 as a separation marker and analyzed H3.5 expression. Further, we investigated the regulation of H3.5 expression using public data repositories. We explored DNA methylation patterns in specific regions of the H3-5 gene and assessed H3-5 copy number gain in seminoma specimens. Additionally, we examined the testicular localization of H3.5 and its histone chaperone interactions to understand its regulation at the protein level. We used qRT-PCR, MeDIP, and qPCR to study H3.5 expression and DNA methylation in various cell types. H3-5 copy number gain was analyzed using qPCR. Protein interactions were investigated through fluorescence-2-hybrid assays in baby hamster kidney cells. H3.5 is primarily enriched in spermatocytes. DNA methylation of a CpG island overlapping the H3-5 promoter appeared to be involved in the tissue-specific regulation of H3.5 expression. Elevated H3.5 expression was observed in seminoma specimens, suggesting a potential link to testicular tumors. H3-5 copy number gain was associated with elevated H3.5 expression in seminoma specimens. Furthermore, we identified physical interactions between H3.5 and histone chaperones Asf1a and Asf1b, HIRA, CAF p150 and DAXX, shedding light on the protein-level regulation of H3.5. These findings provide valuable insights into the molecular mechanisms governing testicular cell differentiation and the potential role of H3.5 in testicular pathologies.
Collapse
Affiliation(s)
- Patrick Philipp Weil
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| | - Anton Pembaur
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| | - Beatrice Wirth
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| | - Eda Oetjen
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| | - Hannes Büsscher
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| | - Klemens Zirngibl
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| | - Malte Czarnetzki
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| | - Stella Braun
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| | - Jann-Frederik Cremers
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Münster, Germany
| | - Daniel Gödde
- Chair of Pathology, Centre for Clinical and Translational Research (ZFKM), Helios University Hospital Wuppertal, Witten/Herdecke University, Heusnerstr. 40, 42283, Wuppertal, Germany
| | - Stephan Degener
- Chair of Urology, Centre for Clinical and Translational Research (ZFKM), Helios University Hospital Wuppertal, Witten/Herdecke University, Heusnerstr. 40, 42283, Wuppertal, Germany
| | - Jan Postberg
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany.
| |
Collapse
|
2
|
Caneparo C, Carignan L, Lonina E, Goulet SM, Pellerin FA, Chabaud S, Bordeleau F, Bolduc S, Pelletier M. Impact of Endocrine Disruptors on the Genitourinary Tract. J Xenobiot 2024; 14:1849-1888. [PMID: 39728407 DOI: 10.3390/jox14040099] [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: 08/30/2024] [Revised: 11/04/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Over the last decades, the human species has seen an increase in the incidence of pathologies linked to the genitourinary tract. Observations in animals have allowed us to link these increases, at least in part, to changes in the environment and, in particular, to an increasing presence of endocrine disruptors. These can be physical agents, such as light or heat; natural products, such as phytoestrogens; or chemicals produced by humans. Endocrine disruptors may interfere with the signaling pathways mediated by the endocrine system, particularly those linked to sex hormones. These factors and their general effects are presented before focusing on the male and female genitourinary tracts by describing their anatomy, development, and pathologies, including bladder and prostate cancer.
Collapse
Affiliation(s)
- Christophe Caneparo
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, Geneva University Hospitals, University of Geneva, CH-1205 Geneva, Switzerland
| | - Laurence Carignan
- Oncology Division, CHU de Québec-Université Laval Research Center and Université Laval Cancer Research Center, Quebec, QC G1R 3S3, Canada
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
| | - Elena Lonina
- Infectious and Immune Diseases Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
- Intersectorial Centre for Endocrine Disruptors Analysis, Institut National de La Recherche Scientifique (INRS), Montreal, QC H4V 1B7, Canada
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University; ARThrite Research Center, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Sarah-Maude Goulet
- Infectious and Immune Diseases Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
- Intersectorial Centre for Endocrine Disruptors Analysis, Institut National de La Recherche Scientifique (INRS), Montreal, QC H4V 1B7, Canada
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University; ARThrite Research Center, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Felix-Antoine Pellerin
- Oncology Division, CHU de Québec-Université Laval Research Center and Université Laval Cancer Research Center, Quebec, QC G1R 3S3, Canada
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
| | - Stéphane Chabaud
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
| | - François Bordeleau
- Oncology Division, CHU de Québec-Université Laval Research Center and Université Laval Cancer Research Center, Quebec, QC G1R 3S3, Canada
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Stéphane Bolduc
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
- Department of Surgery, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Martin Pelletier
- Infectious and Immune Diseases Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
- Intersectorial Centre for Endocrine Disruptors Analysis, Institut National de La Recherche Scientifique (INRS), Montreal, QC H4V 1B7, Canada
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University; ARThrite Research Center, Université Laval, Quebec, QC G1V 0A6, Canada
| |
Collapse
|
3
|
Repetto F, Perrino CM, Hirsch MS. Intratesticular Mullerian Serous Borderline Tumor With Microinvasion: A Rare Tumor and Review of the Literature. Int J Surg Pathol 2024; 32:1567-1573. [PMID: 38509773 DOI: 10.1177/10668969241232702] [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] [Indexed: 03/22/2024]
Abstract
Ovarian-type (ie, Mullerian) epithelial tumors occurring in the testicular and paratesticular regions are exceptionally rare, with only a handful reported worldwide. Serous tumors are the most frequently encountered subtype among these rare tumors. The pathogenesis of these tumors within the testicular and paratesticular regions remains a subject of intrigue and debate, with various hypotheses attempting to explain their presence in the paratestis region, where most tumors occur. However, our understanding of the pathogenesis of intratesticular tumors is limited. To date, 11 known examples of intratesticular serous Mullerian tumors have been reported globally. In this report, we present an extraordinary tumor, an intratesticular Mullerian serous borderline tumor with foci of microinvasion, in a 38-year-old male patient. This tumor exhibits histological features similar to their ovarian counterparts and is confirmed through an immunohistochemical panel. Our report underscores the extreme rarity of these tumors, emphasizes the importance of heightened awareness among clinicians and pathologists, and provides valuable insights into their complex development and histogenesis. This contribution aims to enhance diagnostic precision and optimize therapeutic strategies for similar tumors.
Collapse
Affiliation(s)
| | - Carmen M Perrino
- Department of Pathology, Lahey Hospital & Medical Center, Burlington, MA, USA
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Pera MF. A brief chronicle of research on human pluripotent stem cells. Bioessays 2024; 46:e2400092. [PMID: 39058898 DOI: 10.1002/bies.202400092] [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: 04/13/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024]
Abstract
Today, human pluripotent stem cell technologies find widespread application across biomedical research, as models for early human development, as platforms for functional human genomics, as tools for the study of disease, drug screening and toxicology, and as a renewable source of cellular therapeutics for a range of intractable diseases. The foundations of this human pluripotent stem cell revolution rest on advances in a wide range of disciplines, including cancer biology, assisted reproduction, cell culture and organoid technology, somatic cell nuclear transfer, primate embryology, single-cell biology, and gene editing. This review surveys the slow emergence of the study of human pluripotency and the exponential growth of the field during the past several decades.
Collapse
Affiliation(s)
- Martin F Pera
- JAX Mammalian Genetics, The Jackson Laboratory, Bar Harbor, Maine, USA
| |
Collapse
|
5
|
Zhang B, Kapur P, Koduru PR, Jia L. Retroperitoneal Sarcomatoid Yolk Sac Tumor in a Chemotherapy-Naive Patient With Testicular Postpubertal Type Teratoma: A Rare Case Report With Emphasis on Molecular Features. Int J Surg Pathol 2024; 32:1537-1543. [PMID: 38377960 DOI: 10.1177/10668969241231973] [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] [Indexed: 02/22/2024]
Abstract
Sarcomatoid yolk sac tumor is a very rare histologic type of testicular germ cell tumor and is mainly reported in testicular germ cell tumor patients who receive chemotherapy. Herein, we report an extremely rare concurrent retroperitoneal sarcomatoid yolk sac tumor in a man with a testicular postpuberal teratoma before he received chemotherapy. A 37-year-old man initially presented with a persistent abdominal pain. Subsequent imaging studies revealed a 9.6-cm retroperitoneal mass, and 2 testicular masses (3.1 cm and 0.9 cm in greatest dimension, respectively). His serum tumor markers were within normal ranges. His radical orchiectomy demonstrated a postpubertal type teratoma with an adjacent scarring nodule. Later, his retroperitoneal tumor showed spindle tumor cells embedded in predominantly myxoid and focally fibrous stroma with diffuse and strong immunoreactivity for keratin AE1/AE3, SALL4 and glypican 3. No tumor necrosis or brisk mitotic figures were observed. A diagnosis of sarcomatoid yolk sac tumor was rendered. Fluorescence in situ hybridization analysis of his retroperitoneal sarcomatoid yolk sac tumor revealed polysomy 12 and MYC amplification, whereas no evidence of isochromosome 12p [i(12p)], and DNA sequencing showed 6 mutations per megabase (muts/Mb), and the somatic alterations included ARAF amplification and ATR I774Yfs*5. Considering its rarity, sarcomatoid yolk sac tumor may pose diagnostic challenges. Therefore, relevant clinicoradiologic information and ancillary work up, including immunohistochemistry and molecular studies, may be helpful for the accurate classification. Our tumor further raises awareness of this rare event, expands the spectrum of its clinical presentation, and explores the molecular features.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Prasad R Koduru
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Liwei Jia
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
6
|
Alshwayyat S, Soudi MS, Qaddoura MT, Alshwayyat TA, Ababneh O, Hanifa H, Odat RM, AlAzab RS. Comparative analysis of testicular and nontesticular choriocarcinoma: a population-based study. Ann Med Surg (Lond) 2024; 86:6951-6959. [PMID: 39649900 PMCID: PMC11623855 DOI: 10.1097/ms9.0000000000002702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/18/2024] [Indexed: 12/11/2024] Open
Abstract
Background Germ cell tumors (GCTs) are common solid tumors in young men, originating in the testicles or outside the gonads. Choriocarcinoma, a rare and aggressive subtype, primarily affects females but can also occur in males. Treatment options depend on the stage and location of the tumor, with early recognition being crucial for better outcomes. Comparative studies between testicular and nontesticular choriocarcinoma are crucial for understanding distinct features and prognoses. Methods The study utilized SEER*Stat software to extract data and applied statistical methods such as χ 2 analysis and Kaplan-Meier method. Inclusion criteria focused on patients diagnosed with choriocarcinoma between 2000 and 2018, while exclusion criteria eliminated cases without histological confirmation or with other tumors. Results Among 363 patients, 270 (74.4%) had testicular CC, and 93 (25.6%) had nontesticular CC. Notably, testicular CC was more common in white patients, which could indicate demographic or environmental factors at play. Patients with testicular CC were more likely to undergo surgery, suggesting a significant treatment trend. It is worth exploring whether patient preferences or observed postsurgery improvements contribute to this pattern. Testicular CC had a higher 5-year OS rate of 54% versus 29%, and a higher 5-year CSS rate of 56.3% versus 31.9%, respectively. Conclusion This study reveals distinct characteristics and treatment responses in testicular and nontesticular choriocarcinoma, emphasizing the need for personalized management based on subtype. Our findings highlight racial disparities in incidence and the efficacy of surgical intervention for both types, while chemotherapy benefits extragonadal cases and radiotherapy's role requires further evaluation.
Collapse
Affiliation(s)
- Sakhr Alshwayyat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Maen S. Soudi
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Marwa T. Qaddoura
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Tala A. Alshwayyat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Obada Ababneh
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Hamdah Hanifa
- Faculty of Medicine, University of Kalamoon, Al-Nabk, Syria
| | - Ramez M. Odat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Rami S. AlAzab
- Department of Surgery, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
7
|
Fichtner A, Nettersheim D, Bremmer F. Pathogenesis and pathobiology of testicular germ cell tumours: a view from a developmental biological perspective with guidelines for pathological diagnostics. Histopathology 2024; 85:701-715. [PMID: 38922953 DOI: 10.1111/his.15249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/26/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Abstract
Testicular germ cell tumours (GCT) are divided into three different subtypes (types I-III) regarding to their developmental origin, histological differences and molecular features. Type I GCT develop from disturbed primordial germ cells and most commonly occur in children and young adolescents, which is why they are referred to as prepubertal GCT. Type II GCT develop from a non-invasive germ cell neoplasia in situ (GCNIS) and show an isochromosome 12p (i12p) or gain of 12p material as a common and characteristic molecular alteration. Type III GCT originate from distorted postpubertal germ cells (e.g. spermatogonia) in adult patients and have changes on chromosome 9 with amplification of the DMRT1 gene. Type I GCT encompass prepubertal-type teratomas and yolk-sac tumours (YST). Type II GCT include seminoma, embryonal carcinoma, choriocarcinoma, postpubertal-type teratoma and postpubertal-type YST. Types I and II GCT both show similar morphology, but are separated from each other by the detection of a GCNIS and an i12p in type II GCT. For type II GCT it is especially important to detect non-seminomatous elements, as these tumours have a worse biological behaviour and need a different treatment to seminomas. In contrast to types I and II GCT, type III tumours are equivalent to spermatocytic tumours and usually occur in elderly men, with few exceptions in young adults. The development of types I and II GCT seems to depend not upon driver mutations, but rather on changes in the epigenetic landscape. Furthermore, different pluripotency associated factors (e.g. OCT3/4, SOX2, SOX17) play a crucial role in GCT development and can be used as immunohistochemical markers allowing to distinguish the different subtypes from each other in morphologically challenging tissue specimens. Especially in metastatic sites, a morphological and immunohistochemical diagnostic algorithm is important to detect small subpopulations of each non-seminomatous GCT subtype, which are associated with a poorer prognosis and need a different treatment. Furthermore, primary extragonadal GCT of the retroperitoneum or mediastinum develop from misguided germ cells during embryonic development, and might be challenging to detect in small tissue biopsies due to their rarity at corresponding sites. This review article summarises the pathobiological and developmental aspects of the three different types of testicular GCT that can be helpful in the histopathological examination of tumour specimens by pathologists.
Collapse
Affiliation(s)
| | - Daniel Nettersheim
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Center for Integrated Oncology Aachen, Bonn, Cologne Düsseldorf (CIO ABCD), Lighthouse Project: Germ Cell Tumours, Düsseldorf, Germany
| | - Felix Bremmer
- Institute of Pathology, University Medical Center, Göttingen, Germany
| |
Collapse
|
8
|
Zhang K, Song J, Zhang Y, Chen X, Chao M. Comparison of clinical characteristics of testicular tumor between children and adult population: a retrospective analysis. BMC Cancer 2024; 24:1287. [PMID: 39415133 PMCID: PMC11484207 DOI: 10.1186/s12885-024-12984-2] [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: 08/01/2024] [Accepted: 09/24/2024] [Indexed: 10/18/2024] Open
Abstract
OBJECTIVE Testicular tumor (TT) is a uncommon disease posing serious health problem. There are differences in some aspects between adult and pediatric TT. The study was to compare their differences of clinical and histological characteristics through the analysis of the long-term experiences in TT patients from two institutions. MATERIALS AND METHODS The clinical data of hospitalized patients was collected and analyzed retrospectively from January 2014 to January 2024 at a pediatric and an adult institution, respectively. The data included composition, gender, age, initial presentation, tumor size, tumor markers, pathological diagnosis. RESULTS A total of 195 hospitalized patients were included. There were 135 children and 60 adult with TT, respectively. Of these children, patients were aged from 1 month to 14 years, with a mean age of 2.32 years. More cases (37.04%) were diagnosed at age younger than 1 years. 69 cases were left-sided, 65 cases were right-side and only 1 case was bilateral. Pediatric TTs mainly included 82 prepubertal teratomas, 37 had prepubertal yolk sac tumors and 3 mixed malignant germ tumors. Testicular surgeries included testicular-sparing surgery (TSS) (n = 73), radical orchiectomy (n = 60), and testicular biopsy (n = 2). There were 24 patients receiving postoperative chemotherapy. Adult TTs mainly contained 17 seminomas, 10 prepubertal teratomas,7 postpubertal teratomas, 6 stromal tumors and 3 embryonal carcinomas. The average age was 34.08 years. There were 29 right-sided, 27 left-sided and 4 bilateral tumors. TSS (n = 26), radical orchiectomy (n = 33), and testicular biopsy (n = 1) were performed in these TT patients. Only 6 patients received postoperative chemotherapy. The most common symptom was a painless scrotal mass at initial diagnosis in both groups. In addition, we found that significant differences were explored between histological type and age, tumor size (P < 0.05). Yolk sac tumor and seminoma were the most common malignant TT in pediatric and adult population, respectively. After two year follow-up, two children with yolk sac tumor and 4 adults with seminoma died of their diseases. CONCLUSIONS The majority of pediatric cases were benign compared to adult. The most common type was prepubertal teratoma and yolk sac tumor. Pediatric TTs often occurred under the age of 1 year. Seminomas and prepubertal teratomas were commonly found in adult TTs, especially for young adult. We found that pediatric tumor type was associated with age and tumor size. TSS should be considered for benign TTs based on frozen biopsy findings in children.
Collapse
Affiliation(s)
- Kaiping Zhang
- Department of Urology, Anhui Provincial Children's Hospital, Children's Hospital of Fudan University (Affiliated Anhui Branch), No. 39 Wangjing East Road, Hefei, 230000, Anhui Province, People's Republic of China
| | - Jian Song
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University; Anhui Province Key Laboratory of Genitourinary Diseases, No. 39 Jixi Road, Hefei, 230000, Anhui Province, People's Republic of China
| | - Yin Zhang
- Department of Urology, Anhui Provincial Children's Hospital, Children's Hospital of Fudan University (Affiliated Anhui Branch), No. 39 Wangjing East Road, Hefei, 230000, Anhui Province, People's Republic of China
| | - Xianguo Chen
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University; Anhui Province Key Laboratory of Genitourinary Diseases, No. 39 Jixi Road, Hefei, 230000, Anhui Province, People's Republic of China.
| | - Min Chao
- Department of Urology, Anhui Provincial Children's Hospital, Children's Hospital of Fudan University (Affiliated Anhui Branch), No. 39 Wangjing East Road, Hefei, 230000, Anhui Province, People's Republic of China.
| |
Collapse
|
9
|
Fichtner A, Nettersheim D, Bremmer F. [Histopathological analysis of germ cell tumours: aspects to consider]. Aktuelle Urol 2024. [PMID: 39236760 DOI: 10.1055/a-2363-4744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Germ cell tumours (GCT) are the most common testicular tumours and form a heterogenous group of tumours with different biological behaviour. Based on differences regarding patient age at first manifestation, morphological characteristics and molecular changes, they are divided into three different groups of GCTs (type I-III). For treatment and prognosis, an exact histopathological analysis of an orchiectomy resection specimen is very important, including the specification of all different histological subtypes with their proportional distribution. This article describes the approach of the preparation of a resection specimen and the histopathological analysis of a testicular tumour. In addition, it describes cases in which additional tools like immunohistochemical and molecular analyses are necessary. Furthermore, the current WHO classification of testicular GCT is discussed.
Collapse
Affiliation(s)
- Alexander Fichtner
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Daniel Nettersheim
- Klinik für Urologie, Urologisches Forschungslabor, Translationale Uro-Onkologie, Medizinische Fakultät und Universitätskrankenhaus Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Lighthouse Projekt: Keimzelltumoren, Zentrum für integrierte Onkologie Aachen, Bonn und Köln-Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Felix Bremmer
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
10
|
Demirci A, Başar H. Can serum tumor marker densities according to tumor volume and testicle size be used to predict progression in patients with testicular cancer? Curr Urol 2024; 18:218-224. [PMID: 39219631 PMCID: PMC11337983 DOI: 10.1097/cu9.0000000000000212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/21/2023] [Indexed: 09/04/2024] Open
Abstract
Background The objective of this study is to determine the role of tumor marker density (TMD) values such as alpha-fetoprotein tumor volume ratio (ATVR), beta-human chorionic gonadotropin tumor volume ratio (βTVR), alpha-fetoprotein testicle size ratio (ATSR), beta-human chorionic gonadotropin testicle size ratio (βTSR), lactate dehydrogenase tumor volume ratio (LTVR), and lactate dehydrogenase testicle size ratio (LTSR) in the determination of progression-free survival (PFS) in patients with testicular cancer. Materials and methods A retrospective study was conducted of 95 patients followed-up in our clinic with a diagnosis of testicular cancer between January 2015 and August 2022. Patients were grouped according to clinical stage, as either early stage (n = 50) or advanced stage (n = 45). Clinical and pathological data and TMD values for all patients were recorded. Results The median age of patients was 35 years (21-63 years). All TMDs except LTVR in advanced stage patients were found to be significantly higher than those of early stage patients (p < 0.05). Median ATVR (2.58 vs. 0.0), ATSR (0.63 vs. 0.03), βTVR (0.9 vs. 0.009), and βTSR (0.18 vs. 0.007) of the nonseminoma patients were found to be significantly higher than those of the seminoma patients, respectively (p < 0.001). Progression-free survival (months) was decreased in seminoma patients with high values of βTVR (11.3 ± 1.9 vs. 35.2 ± 0.7), βTSR (16.2 ± 3.4 vs. 35.2 ± 0.75), LTVR (17.7 ± 3.4 vs. 35.2 ± 0.7), and LTSR (21.5 ± 3.13 vs. 35.09 ± 0.8) (p < 0.001). Decreased PFS (months) was associated with higher values of ATVR (5.37 ± 0.7 vs. 35.05 ± 0.93), βTVR (7.4 ± 1.5 vs. 34.6 ± 1.3), ATSR (5.37 ± 0.75 vs. 35.05 ± 0.9), βTSR (7 ± 1.5 vs. 34.6 ± 1.3), and LTSR (7.9 ± 1.2 vs. 34.3 ± 1.5) in nonseminoma patients (p < 0.001). Based on multivariate analysis, βTVR-LTVR and ATVR-ATSR were determined to be independent risk factors for reduced PFS in seminoma and nonseminoma patients, respectively (p < 0.05). Conclusions The results of this study suggest that the calculation of TMDs could be a promising and simple method for prediction of PFS among testicular cancer patients.
Collapse
Affiliation(s)
- Aykut Demirci
- Department of Urology, Dr. Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, Ankara, Turkey
| | | |
Collapse
|
11
|
Tatanis V, Veroutis D, Pantelis P, Theocharous G, Sarlanis H, Georgiou A, Mulita F, Peteinaris A, Natsos A, Moulavasilis N, Kavantzas N, Kotsinas A, Adamakis I. Cellular Senescence in Germ Cell Neoplasia In Situ (GCNIS) and Other Histological Types of Testicular Cancer. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1108. [PMID: 39064537 PMCID: PMC11278860 DOI: 10.3390/medicina60071108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: The presence and contribution of senescent cells in premalignant lesions is well documented, but not in germ cell neoplasia in situ. The purpose of this study is to identify the presence of senescent cells in pre-malignant testicular conditions and in different histological types of testicular cancer. Materials and Methods: Thirty patients who underwent orchiectomy due to testicular tumors were included. Formalin-fixed paraffin-embedded (FFPE) testicular tissue for each patient was available. Sections from these specimens were examined by immunohistochemical analysis with the following markers: GL13 for cellular senescence, p21WAF1/Cip1 for cell cycle arrest, and Ki67 for cell proliferation. Results: Thirteen (43.3%) suffered from seminoma with a mean total proportion of GCNIS senescence of 20.81 ± 6.81%. In the group of embryonal testicular tumors, nine (30%) patients were included, with an average rate of 6.64 ± 5.42% of senescent cells in GCNIS. One (3.3%) patient suffered from chondrosarcoma in which 7.9% of GL13+ cells were detected in GCNIS. Four (13.4%) patients suffered from teratoma and three (10%) from yolk sac tumors, while GCNIS senescence was detected in a range of 4.43 ± 1.78% and 3.76 ± 1.37%, respectively. Conclusions: Cellular senescence was detected in both germ cell neoplasia in situ and testicular cancer, but was more prevalent within the premalignant lesions.
Collapse
Affiliation(s)
- Vasileios Tatanis
- Department of Urology, University of Patras, 26504 Patras, Greece; (V.T.); (A.P.); (A.N.)
| | - Dimitris Veroutis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece; (D.V.); (P.P.); (G.T.); (A.K.)
| | - Pavlos Pantelis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece; (D.V.); (P.P.); (G.T.); (A.K.)
| | - George Theocharous
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece; (D.V.); (P.P.); (G.T.); (A.K.)
| | - Helen Sarlanis
- Department of Pathology, Medical School, National and Kapodistrian University, 10680 Athens, Greece; (H.S.); (A.G.); (N.K.)
| | - Alexandros Georgiou
- Department of Pathology, Medical School, National and Kapodistrian University, 10680 Athens, Greece; (H.S.); (A.G.); (N.K.)
| | - Francesk Mulita
- Department of Surgery, University Hospital of Patras, 26504 Patras, Greece
| | - Angelis Peteinaris
- Department of Urology, University of Patras, 26504 Patras, Greece; (V.T.); (A.P.); (A.N.)
| | - Anastasios Natsos
- Department of Urology, University of Patras, 26504 Patras, Greece; (V.T.); (A.P.); (A.N.)
| | - Napoleon Moulavasilis
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10680 Athens, Greece; (N.M.); (I.A.)
| | - Nikolaos Kavantzas
- Department of Pathology, Medical School, National and Kapodistrian University, 10680 Athens, Greece; (H.S.); (A.G.); (N.K.)
| | - Athanassios Kotsinas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece; (D.V.); (P.P.); (G.T.); (A.K.)
| | - Ioannis Adamakis
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10680 Athens, Greece; (N.M.); (I.A.)
| |
Collapse
|
12
|
Misgar RA, Islam Mir SU, Mir MH, Bashir MI, Wani AI, Masoodi SR. Germ Cell Tumors in 46, XY Gonadal Dysgenesis. Indian J Endocrinol Metab 2024; 28:424-428. [PMID: 39371651 PMCID: PMC11451959 DOI: 10.4103/ijem.ijem_373_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 02/15/2023] [Indexed: 10/08/2024] Open
Abstract
Introduction To present the clinical data, investigative profile, management, and follow-up of patients with 46, XY gonadal dysgenesis with germ cell tumors from the endocrine unit of a tertiary care university hospital. Materials and Methods This retrospective study included 3 cases of 46, XY gonadal dysgenesis with germ cell tumors evaluated and managed at the Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Kashmir, over a period of 13 years from (September 2008 to December 2021). Results Over a period of 13 years, we diagnosed and managed 7 patients with 46, XY gonadal dysgenesis. This included 4 patients with pure gonadal dysgenesis (PGD; Swyer syndrome), 2 patients with mixed gonadal dysgenesis (MGD), and one patient with partial gonadal dysgenesis. Out of these 7 patients, three patients developed germ cell tumors, one patient with MGD, and two patients with pure PGD (Swyer syndrome). In all three patients, germ cell tumor was the first presentation of DSD. The patient with MGD presented with primary amenorrhea and virilization, while the two patients with PGD presented as phenotypic females with primary amenorrhea and pelvic mass. All three patients developed seminomatous cancers. Patient with MGD developed seminoma and the two patients with PGD (Swyer syndrome) developed dysgerminoma. The patients were managed with bilateral gonadectomy with removal of the tumor. In addition, the 2 patients with PGD (Swyer syndrome) received combined chemotherapy. On a follow up ranging from 1 to 10 years, all three patients are disease free. Conclusions we conclude that germ cell tumors may be the first presentation of 46, XY gonadal dysgenesis. In all phenotypic females with primary amenorrhea and dysgerminoma, karyotype is a must to uncover the diagnosis of PGD. In addition virilization may be clue to the presence of germ cell tumor in a patient with 46, XY gonadal dysgenesis.
Collapse
Affiliation(s)
- Raiz A. Misgar
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Kashmir, India
| | - Sajad U. Islam Mir
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Kashmir, India
| | - Mohmad H. Mir
- Department of Medical Oncology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Kashmir, India
| | - Mir I. Bashir
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Kashmir, India
| | - Arshad I. Wani
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Kashmir, India
| | - Shariq R. Masoodi
- Department of Endocrinology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Kashmir, India
| |
Collapse
|
13
|
Whaley RD, Ulbright TM. Primitive Embryonic-Type Neuroectodermal/Glandular Complexes in Testicular Germ Cell Tumors: A Mimic of Embryonic-Type Neuroectodermal Tumor. Am J Surg Pathol 2024; 48:909-917. [PMID: 38712621 DOI: 10.1097/pas.0000000000002241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Embryonic-type neuroectodermal elements are often intimately mixed with primitive endodermal-type glands, like those of yolk sac tumors, in germ cell neoplasia in situ (GCNIS)-derived germ cell tumors of the testis. Because the primitive glands mimic tubules or rosettes of embryonic-type neuroectodermal elements, these embryonic-type neuroectodermal/glandular complexes may be misinterpreted as pure lesions of embryonic-type neuroectodermal elements, which, if of sufficient size, may lead to a diagnosis of embryonic-type neuroectodermal tumor, despite that the criteria of the World Health Organization for a "somatic-type malignancy" are not met. A diagnosis of embryonic-type neuroectodermal tumor in the testis may lead to retroperitoneal lymphadenectomy even in clinical stage I patients, and in postchemotherapy resections indicates a poor prognosis. The distinction of the neuroectodermal and glandular elements is not always straightforward based on morphology alone. We, therefore, studied 34 testis-derived germ cell tumors with embryonic-type neuroectodermal/glandular complexes and 2 purely glandular yolk sac tumors to characterize the immunophenotypes and determine an efficient immunohistochemical panel to aid in this differential. We found that GFAP, synaptophysin, and paired-like homeobox 2B (PHOX2B) expression was specific to embryonic-type neuroectodermal elements, although PHOX2B had poor sensitivity. In contrast, positive reactions with antibodies directed against AFP, villin, and CDX2 were specific for the glandular elements, although CDX2 had poor sensitivity. Other markers, including AE1/AE3 cytokeratin, SALL4, glypican 3, SOX2, SOX11, CD56, INSM1, and neurofilament, proved less helpful because of their nonspecificity and/or poor sensitivity. We conclude that the optimal immunohistochemical panel for distinguishing the components of embryonic-type neuroectodermal/glandular complexes includes stains for synaptophysin, GFAP, villin, and AFP.
Collapse
Affiliation(s)
- Rumeal D Whaley
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Thomas M Ulbright
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
14
|
Fazekas FE, Ujfaludi Z, Biró K, Páhi ZG, Buzogány I, Sükösd F, Pankotai T, Beöthe T. Complex treatment of residual metastatic germ cell cancer: A single center experience. J Biotechnol 2024; 389:61-67. [PMID: 38692356 DOI: 10.1016/j.jbiotec.2024.04.018] [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: 03/27/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Testicular cancer is the most common solid malignancy among men aged 15-35. Radical orchiectomy and platinum-based chemotherapy (BEP) are curative in the majority of patients, including advanced, metastatic cases. According to current urooncology guidelines all non-seminoma patients harbouring post-chemotherapy residual masses of ≥ 1 cm should undergo salvage retroperitoneal lymph node dissection (RPLND). However, only 10% of residual tumors contain viable disease. OBJECTIVE To assess patient outcomes and complications considering different treatment regimens and clinical characteristics. MATERIALS AND METHODS In a retrospective cross-sectional study patients (n=127) who underwent postchemotherapy RPLND between 2007 and 2023 at our referral center were evaluated. The patients received systemic treatment at various oncology centers. The number of BEP cycles received were occasionally different from standard. Only patients with normal postchemotherapy serum tumor markers and primary testicular or extragonadal germ cell neoplasms were included. Treatment groups were established according to the number of BEP cycles received, and the extent of RPLND (bilateral or modified template). Treatment outcomes and complications were assessed. RESULTS Standard 3-4 courses of BEP were received by 100 (78,7%) patients, while 11 (8,7%) patients underwent less, and 16 (12,6%) more courses than standard. On histopathologic evaluation viable germ cell tumor, teratoma, and necrosis/fibrosis was present in 26 (20,5%), 67 (52,7%) and 34 (26,8%) of specimen, respectively. In the 5-6 BEP series subgroup high rate of viable disease (37,5%) was found and significantly more nephrectomies were performed, than other chemotherapy subgroups. Extratesticular GCT, viable disease in residual mass or progression after RPLND indicated lower survival. Mild (Clavien-Dindo I-II) or no postoperative complications were reported in 93,7% of cases. CONCLUSIONS The study suggests no significant benefit from exceeding 3-4 courses of BEP. Timely salvage RPLND should be performed in high volume centers for optimal treatment outcomes with acceptable complication rates. Adherence to the Heidenreich criteria is advisable where practical.
Collapse
Affiliation(s)
| | - Zsuzsanna Ujfaludi
- Institute of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary; Competence Centre of the Life Sciences Cluster of the Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, Dugonics tér, 13, Szeged H-6720, Hungary
| | - Krisztina Biró
- National Institute of Oncology, Dept. of Genitourinary Oncology and Clinical Pharmacology, Budapest, Hungary
| | - Zoltán Gábor Páhi
- Institute of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary; Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Genome Integrity and DNA Repair Core Group, Budapesti út 9, Szeged H-6728, Hungary; Competence Centre of the Life Sciences Cluster of the Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, Dugonics tér, 13, Szeged H-6720, Hungary
| | - István Buzogány
- Péterfy Sándor Hospital, Dept. of Urology, Budapest, Hungary
| | - Farkas Sükösd
- Institute of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Tibor Pankotai
- Institute of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary; Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Genome Integrity and DNA Repair Core Group, Budapesti út 9, Szeged H-6728, Hungary; Competence Centre of the Life Sciences Cluster of the Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, Dugonics tér, 13, Szeged H-6720, Hungary.
| | - Tamás Beöthe
- Péterfy Sándor Hospital, Dept. of Urology, Budapest, Hungary.
| |
Collapse
|
15
|
Sköld C, Jansson AK, Glimelius I. Malignant ovarian and testicular germ cell tumors: Common characteristics but different prognoses. J Intern Med 2024; 295:715-734. [PMID: 38468475 DOI: 10.1111/joim.13778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Both ovarian and testicular germ cell tumors (GCTs) arise from the primordial germ cell and share many similarities. Both malignancies affect mainly young patients, show remarkable responsiveness to cisplatin-based therapy, and have an excellent prognosis, which also highlights the importance of minimizing long-term side effects. However, certain differences can be noted: The spreading of the disease differs, and the staging system and treatment recommendations are dissimilar. Moreover, the prognosis for ovarian GCTs is significantly inferior to that for testicular cancer, as exemplified in this review comparing the survival in Swedish patients diagnosed with testicular (1995-2022) and ovarian (1990-2018) GCTs. The 5-year overall survival in ovarian GCTs was 85.2%, versus 98.2% for testicular GCTs. How can this be explained? One reason may be the difference in knowledge, experience, and evidence because the incidence rate of testicular cancer is more than 15 times that of ovarian GCTs. Given the rarity of the disease in women and the lack of established guidelines, a comprehensive understanding of the disease and treatment decisions is challenging. The main objective of this review is to derive insights from testicular GCTs (seminoma and non-seminoma) by reviewing etiological, tumor biological, and clinical knowledge, and to thereafter suggest actions for ovarian GCTs based on this. We hypothesize that by adopting specific treatment strategies from testicular GCTs-including de-escalating adjuvant chemotherapy for low-risk patients and implementing more standardized and intensive treatment protocols in cases of relapse-we can improve the prognosis and minimize long-term side effects in ovarian GCT patients.
Collapse
Affiliation(s)
- Camilla Sköld
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
| | - Anna K Jansson
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
| | - Ingrid Glimelius
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
- Department of Medicine, Clinical Epidemiology Unit, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
16
|
Kilic I, Acosta AM, Idrees MT. Evolution of Testicular Germ Cell Tumors in the Molecular Era With Histogenetic Implications. Adv Anat Pathol 2024; 31:206-214. [PMID: 38525515 DOI: 10.1097/pap.0000000000000438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
The current WHO classification of testicular germ cell tumors is based on the pathogenesis of the tumors driven by different genomic events. The germ cell neoplasia in situ is the precursor lesion for all malignant germ cell tumors. The current understanding of pathogenesis is that the developmental and environmental factors with the erasure of parental genomic imprinting lead to the development of abnormal gonocytes that settle in the "spermatogonial Niche" in seminiferous tubules. The abnormal primordial germ cells in the seminiferous tubules give rise to pre-GCNIS cells under the influence of TPSY and OCT4 genes. The whole genome duplication events give rise to germ cell neoplasia in situ, which further acquires alterations in 12p along with NRAS and KRAS mutations to produce seminoma. A subset of seminomas acquires KIT mutation and does not differentiate further. The remaining KIT-stable seminomas differentiate to nonseminomatous GCTs after obtaining recurrent chromosomal losses, epigenetic modification, and posttranscriptional regulation by multiple genes. Nonseminomatous germ cell tumors also develop directly from differentiated germ cell neoplasia in situ. TP53 pathway with downstream drivers may give rise to somatic-type malignancies of GCT. The GCTs are remarkably sensitive to cisplatin-based combination chemotherapy; however, resistance to cisplatin develops in up to 8% of tumors and appears to be driven by TP53/MDM2 gene mutations. Serum and Plasma miRNAs show promise in diagnosing, managing, and following up on these tumors. The mechanisms underlying the development of most tumors have been elucidated; however, additional studies are required to pinpoint the events directing specific characteristics. Advances in identifying specific molecular markers have been seen recently and may be adopted as gold standards in the future.
Collapse
Affiliation(s)
- Irem Kilic
- Department of Pathology, Indiana University, Indianapolis, IN
| | | | | |
Collapse
|
17
|
K AR, Arumugam S, Muninathan N, Baskar K, S D, D DR. P53 Gene as a Promising Biomarker and Potential Target for the Early Diagnosis of Reproductive Cancers. Cureus 2024; 16:e60125. [PMID: 38864057 PMCID: PMC11165294 DOI: 10.7759/cureus.60125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/10/2024] [Indexed: 06/13/2024] Open
Abstract
One of the crucial aspects of cancer research is diagnosis with specificity and accuracy. Early cancer detection mostly helps make appropriate decisions regarding treatment and metastasis. The well-studied transcription factor tumor suppressor protein p53 is essential for maintaining genetic integrity. p53 is a key tumor suppressor that recognizes the carcinogenic biological pathways and eradicates them by apoptosis. A wide range of carcinomas, especially gynecological such as ovarian, cervical, and endometrial cancers, frequently undergo TP53 gene mutations. This study evaluates the potential of the p53 gene as a biological marker for the diagnosis of reproductive system neoplasms. Immunohistochemistry of p53 is rapid, easy to accomplish, cost-effective, and preferred by pathologists as a surrogate for the analysis of TP53 mutation. Thus, this review lays a groundwork for future efforts to develop techniques using p53 for the early diagnosis of cancer.
Collapse
Affiliation(s)
- Aswathi R K
- Medical Biochemistry, Meenakshi Academy of Higher Education and Research, Chennai, IND
| | - Suresh Arumugam
- Central Research Laboratory, Meenakshi Medical College Hospital and Research Institute, Kanchipuram, IND
| | - Natrajan Muninathan
- Central Research Laboratory, Meenakshi Medical College Hospital and Research Institute, Kanchipuram, IND
| | - Kuppusamy Baskar
- Central Research Laboratory, Meenakshi Medical College Hospital and Research Institute, Kanchipuram, IND
| | - Deepthi S
- Research and Development, Meenakshi Academy of Higher Education and Research, Chennai, IND
| | - Dinesh Roy D
- Centre for Advanced Genetic Studies, Genetika, Thiruvananthapuram, IND
| |
Collapse
|
18
|
Saliyeva S, Boranbayeva R, Bulegenova M, Zhumadullayev B, Nurzhanova G, Manzhuova L. Outcomes of pediatric extracranial germ cell tumors: A single center experience in a developing country. Rare Tumors 2024; 16:20363613231216567. [PMID: 38566825 PMCID: PMC10986168 DOI: 10.1177/20363613231216567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 11/08/2023] [Indexed: 04/04/2024] Open
Abstract
The purpose of this study was to analyze the outcomes of extracranial GCT in children in a developing country and to assess prognostic factors. The data on 141 children (<18 years old) with extracranial GCT, confirmed histopathologically, collected over the past 9 years (from February 2013 to June 2022) were retrospectively studied. The patients underwent the same therapy with platinum-containing chemotherapy regimens. In the malignant GCT group, OS and EFS were 81.0 ± 4% and 73 ± 5%, respectively. OS and EFS in the teratoma group were 90 ± 5% and 85 ± 6%. In univariate analysis, parameters like stage of disease, tumor localization, AFP level ≥10,000 ng/mL, serum AFP kinetics and resection status were found to be statistically significant prognostic factors. In the multivariate analysis, the significant adverse factors were the resection status, initial AFP level ≥10,000 ng/mL and serum AFP kinetics slow down (p = .000). Good survival rates can be achieved in developing countries with adequate compliance with treatment protocols. The analysis demonstrates high efficacy of platinum-containing chemotherapy regimens. In our opinion, the protocol used in high-income countries can be implemented in low-income countries with the financial support from the government. The qualification of specialists is also important.
Collapse
Affiliation(s)
- Symbat Saliyeva
- Oncology/hematology Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
- Department of Children's Diseases, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Riza Boranbayeva
- Oncology/hematology Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
- Department of Children's Diseases, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Minira Bulegenova
- Oncology/hematology Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
- Department of Children's Diseases, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Bakhram Zhumadullayev
- Oncology/hematology Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
| | - Gaukhar Nurzhanova
- Oncology/hematology Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
| | - Lyazat Manzhuova
- Oncology/hematology Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
- Department of Children's Diseases, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| |
Collapse
|
19
|
Favero D, Oing C, Seidel C, Rescigno P, Catalano F, Cremante M, Rebuzzi SE, Gatto F, Rosti G, Ferone D, Fornarini G, Cocchiara F. Hyperthyroidism in non-seminomatous testicular germ cell tumors: two case reports and literature review. Front Oncol 2024; 14:1338438. [PMID: 38601761 PMCID: PMC11004277 DOI: 10.3389/fonc.2024.1338438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/26/2024] [Indexed: 04/12/2024] Open
Abstract
Background Human chorionic gonadotropin (hCG)-induced hyperthyroidism is a rare paraneoplastic syndrome observed in non-seminomatous testicular germ cell tumors, due to a cross-reaction between the β-subunit of hCG with the thyroid-stimulating hormone receptor. The precise prevalence of this paraneoplastic phenomenon is unclear as, in the majority of cases, hyperthyroidism remains subclinical. Case presentation Here, we present two cases of advanced metastatic non-seminomatous testicular germ cell tumors where patients exhibited signs and symptoms of thyrotoxicosis at primary diagnosis due to excessive serum β-hCG elevation, with complete remission of symptomatology after the start of oncological treatments and no signs of relapse at the time of publication of this report. Additionally, we provide a comprehensive review of the existing literature concerning this uncommon occurrence. Conclusion Despite being a rare event, the presence of hyperthyroidism or thyrotoxicosis without clear etiology in a young man should lead to consider less frequent causes such as testicular tumors. Even if patients typically have mild symptoms that resolve after chemotherapy, in rare cases, it can be a life-threatening condition that requires prompt recognition and specific intervention.
Collapse
Affiliation(s)
- Diletta Favero
- Department of Internal Medicine and Medical Specialties (Di.M.I.), School of Medicine, University of Genova, Genova, Italy
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genova, Italy
| | - Christoph Oing
- Translational and Clinical Research Institute, Centre for Cancer, Newcastle University, Newcastle upon Tyne, United Kingdom
- Mildred Scheel Cancer Career Centre HaTriCS4, University Cancer Centre Hamburg, University Medical Centre Eppendorf, Hamburg, Germany
| | - Christoph Seidel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Centre Eppendorf, Hamburg, Germany
| | - Pasquale Rescigno
- Translational and Clinical Research Institute, Centre for Cancer, Newcastle University, Newcastle upon Tyne, United Kingdom
- Candiolo Cancer Institute Fondazione del Piemonte per l’Oncologia-Istituto di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), Candiolo, Italy
| | - Fabio Catalano
- Medical Oncology Unit 1, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genova, Italy
| | - Malvina Cremante
- Medical Oncology Unit 1, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genova, Italy
| | - Sara Elena Rebuzzi
- Medical Oncology Unit, Ospedale San Paolo, Savona, Italy
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy
| | - Federico Gatto
- Endocrinology Unit, Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genova, Italy
| | - Giovanni Rosti
- Oncology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Diego Ferone
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (Di.M.I.), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genova, Italy
| | - Francesco Cocchiara
- Endocrinology Unit, Department of Internal Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
20
|
Mohin M, Dey S, Ray R, Wasim Sk F, Das O, Chatterjee U. Testicular Mixed Teratoma and Yolk Sac Tumor, Prepubertal Type: A Case Report with Summary of Prior Published Cases. Fetal Pediatr Pathol 2024; 43:182-187. [PMID: 37946365 DOI: 10.1080/15513815.2023.2279132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Testicular mixed germ cell tumor is common in the post-pubertal age, less so in prepuberty. There are only 3 reports of prepubertal mixed teratoma and yolk sac tumor. Two of these cases had immature teratoma component and were in the neonatal age group. The third case in a toddler had a mature teratoma component. CASE REPORT An 18-month-old boy presented with a testicular mass. Serum AFP was elevated (2200 ng/ml). The orchidectomy specimen contained a yolk-sac tumor and a small epidermoid cyst, indicating a mature teratomatous component. CONCLUSION We report a testicular mixed teratoma and yolk sac tumor, prepubertal type along with summary of prior published cases. There is only one report describing this combination of mature teratoma with yolk sac tumor in the prepubertal testis.
Collapse
Affiliation(s)
- Md Mohin
- Department of Pathology, Institute of Postgraduate Medical Education & Research (IPGME&R), Kolkata, India
| | - Soumya Dey
- Department of Pathology, Institute of Postgraduate Medical Education & Research (IPGME&R), Kolkata, India
| | - Raktim Ray
- Department of Pathology, Institute of Postgraduate Medical Education & Research (IPGME&R), Kolkata, India
| | - Firdous Wasim Sk
- Department of Pathology, Institute of Postgraduate Medical Education & Research (IPGME&R), Kolkata, India
| | - Oindrila Das
- Department of Pathology, Institute of Postgraduate Medical Education & Research (IPGME&R), Kolkata, India
| | - Uttara Chatterjee
- Department of Pathology, Institute of Postgraduate Medical Education & Research (IPGME&R), Kolkata, India
| |
Collapse
|
21
|
Fichtner A, Marx A, Ströbel P, Bremmer F. Primary germ cell tumours of the mediastinum: A review with emphasis on diagnostic challenges. Histopathology 2024; 84:216-237. [PMID: 37994540 DOI: 10.1111/his.15090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/24/2023]
Abstract
This article will review current aspects of the histopathological, immunohistochemical and molecular analysis of primary mediastinal germ cell tumours (PMGCTs) as well as their aetiological, epidemiological, clinical and therapeutic features. PMGCTs represent an important differential diagnosis in the spectrum of mediastinal tumours, and their diagnosis is usually made on small tissue samples from core needle biopsies in combination with diagnostic imaging and serum tumour markers. As in lymphomas, a small biopsy is often the only viable tumour sample available from these patients, as they receive chemotherapy prior to eventual surgical resection. Pathologists therefore need to apply an efficient combination of immunohistochemical markers to confirm the diagnosis of a PMGCT and to exclude morphological mimics.
Collapse
Affiliation(s)
- Alexander Fichtner
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Alexander Marx
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Felix Bremmer
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
22
|
Lu X, Luo Y, Nie X, Zhang B, Wang X, Li R, Liu G, Zhou Q, Liu Z, Fan L, Hotaling JM, Zhang Z, Bo H, Guo J. Single-cell multi-omics analysis of human testicular germ cell tumor reveals its molecular features and microenvironment. Nat Commun 2023; 14:8462. [PMID: 38123589 PMCID: PMC10733385 DOI: 10.1038/s41467-023-44305-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
Seminoma is the most common malignant solid tumor in 14 to 44 year-old men. However, its molecular features and tumor microenvironment (TME) is largely unexplored. Here, we perform a series of studies via genomics profiling (single cell multi-omics and spatial transcriptomics) and functional examination using seminoma samples and a seminoma cell line. We identify key gene expression programs share between seminoma and primordial germ cells, and further characterize the functions of TFAP2C in promoting tumor invasion and migration. We also identify 15 immune cell subtypes in TME, and find that subtypes with exhaustion features were located closer to the tumor region through combined spatial transcriptome analysis. Furthermore, we identify key pathways and genes that may facilitate seminoma disseminating beyond the seminiferous tubules. These findings advance our knowledge of seminoma tumorigenesis and produce a multi-omics atlas of in situ human seminoma microenvironment, which could help discover potential therapy targets for seminoma.
Collapse
Affiliation(s)
- Xiaojian Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yanwei Luo
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xichen Nie
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Bailing Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyan Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Ran Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Guangmin Liu
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Qianyin Zhou
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Zhizhong Liu
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Liqing Fan
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, China
| | - James M Hotaling
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Zhe Zhang
- Department of Urology, Peking University Third Hospital, Beijing, China.
- Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China.
| | - Hao Bo
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China.
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, China.
| | - Jingtao Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
23
|
Młynarczyk G, Domian N, Kasacka I. Changes in adhesion molecules: β-catenin, E-cadherin and Galectin-3 in cells of testicular seminoma. Front Oncol 2023; 13:1269637. [PMID: 38144531 PMCID: PMC10739379 DOI: 10.3389/fonc.2023.1269637] [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: 07/30/2023] [Accepted: 11/23/2023] [Indexed: 12/26/2023] Open
Abstract
Introduction The most common testicular tumors are seminomas. They are characterized by rapid growth and a very high potential for metastasis to other organs. Mutual interactions of tumor cells play an important role in the invasiveness and metastatic capacity, in which complexes of adhesion proteins play a special role. There is a lack of studies on changes in these molecules and their behaviour in testicular cancer. The aim of the study was immunohistochemical identification and evalutaion of adhesive molecules β-catenin, E-cadherin, galectin-3 in testicular cancer - seminoma. Methods Tests were performed on sections of testicular cancer - seminoma in comparison with unchanged tissue samples as a control. Material was taken from 30 patients who underwent orchiectomy. Immunohistochemistry and PCR were used to identify β-catenin, E-cadherin and galectin-3 and gene expression. Results Immunoreactivity and expression of β-catenin and E-cadherin in seminomas were markedly decreased compared to non-cancerous testicular tissue. Galectin-3 immunoreactivity was found in both control and cancerous tissue, but in different location. In non-cancerous tissue, it was localized in the cytoplasm of the cells of the seminiferous tubules, in seminomas it was localized mainly in the endothelium. The expression of the Lgals3 gene encoding galectin-3 in seminomas was slightl higher in relation to the tissue unchanged by the carcinogenetic process. Conclusions The results of the study suggest a significant role of β-catenin, E-cadherin and galectin-3 in the carcinogenesis of seminomas and may indicate new aspects of the patomechanism of seminomas formation, and thus time lead to better understand the biology of these tumors.
Collapse
Affiliation(s)
| | - Natalia Domian
- Department of Histology and Cytophysiology, Medical University of Białystok, Bialystok, Poland
| | - Irena Kasacka
- Department of Histology and Cytophysiology, Medical University of Białystok, Bialystok, Poland
| |
Collapse
|
24
|
Bremmer F. [Teratoma with somatic-type malignancy : Analyses to characterize the mutations, DNA methylation, and proteome]. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:109-112. [PMID: 38047949 DOI: 10.1007/s00292-023-01282-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 12/05/2023]
Affiliation(s)
- F Bremmer
- Institut für Pathologie, Universitätsmedizin Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Deutschland.
| |
Collapse
|
25
|
Wei R, Zhang X, Li X, Wen J, Liu H, Fu J, Li L, Zhang W, Liu Z, Yang Y, Zou K. A rapid and stable spontaneous reprogramming system of Spermatogonial stem cells to Pluripotent State. Cell Biosci 2023; 13:222. [PMID: 38041111 PMCID: PMC10693117 DOI: 10.1186/s13578-023-01150-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 10/20/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND The scarcity of pluripotent stem cells poses a major challenge to the clinical application, given ethical and biosafety considerations. While germline stem cells commit to gamete differentiation throughout life, studies demonstrated the spontaneous acquisition of pluripotency by spermatogonial stem cells (SSCs) from neonatal testes at a low frequency (1 in 1.5 × 107). Notably, this process occurs without exogenous oncogenes or chemical supplementation. However, while knockout of the p53 gene accelerates the transformation of SSCs, it also increases risk and hampers their clinical use. RESULTS We report a transformation system that efficiently and stably convert SSCs into pluripotent stem cells around 10 passages with the morphology similar to that of epiblast stem cells, which convert to embryonic stem (ES) cell-like colonies after change with ES medium. Epidermal growth factor (EGF), leukemia inhibitory factor (LIF) and fresh mouse embryonic fibroblast feeder (MEF) are essential for transformation, and addition of 2i (CHIR99021 and PD0325901) further enhanced the pluripotency. Transcriptome analysis revealed that EGF activated the RAS signaling pathway and inhibited p38 to initiate transformation, and synergically cooperated with LIF to promote the transformation. CONCLUSION This system established an efficient and safe resource of pluripotent cells from autologous germline, and provide new avenues for regenerative medicine and animal cloning.
Collapse
Affiliation(s)
- Rui Wei
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiaoyu Zhang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiaoxiao Li
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jian Wen
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China
| | - Hongyang Liu
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jiqiang Fu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China
| | - Li Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China
| | - Wenyi Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China
| | - Zhen Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China
| | - Yang Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
26
|
Urbini M, Bleve S, Schepisi G, Menna C, Gurioli G, Gianni C, De Giorgi U. Biomarkers for Salvage Therapy in Testicular Germ Cell Tumors. Int J Mol Sci 2023; 24:16872. [PMID: 38069192 PMCID: PMC10706346 DOI: 10.3390/ijms242316872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
The outcome of metastatic testicular germ cell tumor patients has been dramatically improved by cisplatin-based chemotherapy combinations. However, up to 30% of patients with advanced disease relapse after first-line therapy and require salvage regimens, which include treatments with conventional-dose chemotherapy or high-dose chemotherapy with autologous stem cell transplantation. For these patients, prognosis estimation represents an essential step in the choice of medical treatment but still remains a complex challenge. The available histological, clinical, and biochemical parameters attempt to define the prognosis, but they do not reflect the tumor's molecular and pathological features and do not predict who will exhibit resistance to the several treatments. Molecular selection of patients and validated biomarkers are highly needed in order to improve current risk stratification and identify novel therapeutic approaches for patients with recurrent disease. Biomolecular biomarkers, including microRNAs, gene expression profiles, and immune-related biomarkers are currently under investigation in testicular germ cell tumors and could potentially hold a prominent place in the future treatment selection and prognostication of these tumors. The aim of this review is to summarize current scientific data regarding prognostic and predictive biomarkers for salvage therapy in testicular germ cell tumors.
Collapse
Affiliation(s)
- Milena Urbini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Sara Bleve
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (S.B.); (G.S.); (C.M.); (C.G.); (U.D.G.)
| | - Giuseppe Schepisi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (S.B.); (G.S.); (C.M.); (C.G.); (U.D.G.)
| | - Cecilia Menna
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (S.B.); (G.S.); (C.M.); (C.G.); (U.D.G.)
| | - Giorgia Gurioli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Caterina Gianni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (S.B.); (G.S.); (C.M.); (C.G.); (U.D.G.)
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (S.B.); (G.S.); (C.M.); (C.G.); (U.D.G.)
| |
Collapse
|
27
|
Jiang S, Dong K, Li K, Liu J, Du X, Huang C, Jiao Y, Han Y, Yang J, Liao X, Li Y, Zhang T, Li S, Lv Z, Gao Y. Extracranial Germ Cell Tumors in Children: Ten Years of Experience in Three Children's Medical Centers in Shanghai. Cancers (Basel) 2023; 15:5412. [PMID: 38001671 PMCID: PMC10670163 DOI: 10.3390/cancers15225412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/28/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
OBJECTIVE The aim was to describe the clinical features of extracranial germ cell tumors (GCTs) in pediatrics and study the clinical risk factors related to survival for malignant germ cell tumors (MGCTs) in order to optimize therapeutic options. METHODS The clinical data of children with extracranial GCTs in three children's medical centers in Shanghai were retrospectively analyzed. RESULTS In total, 1007 cases of extracranial GCTs diagnosed between 2010 and 2019 were included in this study, including teratomas (TERs) 706 (70.11%) and MGCTs 301 (29.89%). There were twice as many TER cases as MGCT cases. Approximately 50% of children with GCTs were <3 years old (43.39% for TERs, 67.13% for MGCTs). GCTs in children of different ages show differences in tumor anatomical locations and pathological subtypes. The 5-year event-free survival (EFS) and overall survival (OS) of all patients with MGCTs were 82.33% (95% CI, 77.32%, 86.62%) and 94.13% (95% CI, 90.02%, 96.69%), respectively. The multivariate Cox regression analysis identified a primary site in the mediastinum and alpha fetoprotein (AFP) levels ≥10,000 ng/mL as independent adverse prognostic factors (p < 0.0.0001, χ2 = 23.6638, p = 0.0225, χ2 = 5.2072.). There were no significant differences in OS among children receiving various chemotherapy regimens, such as the BEP, PEB, JEB and other regimens (VBP/VIP and AVCP/IEV) (p < 0.05). CONCLUSIONS The clinical features of GCTs in Chinese pediatrics are similar to those reported in children in Europe and America. The age distribution of pathological types and primary sites in GCTs reflect the developmental origin of type I and type II GCTs transformed from mismigration primordial germ cells (PGCs). Optimizing the current platinum-based chemotherapy regimens and exploring the treatment strategies for MGCTs of the mediastinum are future research directions.
Collapse
Affiliation(s)
- Shayi Jiang
- Department of Hematology and Oncology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355 Luding Road, Shanghai 200061, China; (S.J.); (X.D.); (C.H.); (Y.J.); (J.Y.); (X.L.); (Y.L.); (T.Z.); (S.L.)
| | - Kuiran Dong
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai 201102, China; (K.D.); (K.L.)
| | - Kai Li
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai 201102, China; (K.D.); (K.L.)
| | - Jiangbin Liu
- Department of Pediatric Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355 Luding Road, Shanghai 200061, China;
| | - Xin Du
- Department of Hematology and Oncology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355 Luding Road, Shanghai 200061, China; (S.J.); (X.D.); (C.H.); (Y.J.); (J.Y.); (X.L.); (Y.L.); (T.Z.); (S.L.)
| | - Can Huang
- Department of Hematology and Oncology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355 Luding Road, Shanghai 200061, China; (S.J.); (X.D.); (C.H.); (Y.J.); (J.Y.); (X.L.); (Y.L.); (T.Z.); (S.L.)
| | - Yangyang Jiao
- Department of Hematology and Oncology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355 Luding Road, Shanghai 200061, China; (S.J.); (X.D.); (C.H.); (Y.J.); (J.Y.); (X.L.); (Y.L.); (T.Z.); (S.L.)
| | - Yali Han
- Department of Hematology and Oncology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, No. 1678 Dongfang Road, Shanghai 200127, China;
| | - Jingwei Yang
- Department of Hematology and Oncology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355 Luding Road, Shanghai 200061, China; (S.J.); (X.D.); (C.H.); (Y.J.); (J.Y.); (X.L.); (Y.L.); (T.Z.); (S.L.)
| | - Xuelian Liao
- Department of Hematology and Oncology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355 Luding Road, Shanghai 200061, China; (S.J.); (X.D.); (C.H.); (Y.J.); (J.Y.); (X.L.); (Y.L.); (T.Z.); (S.L.)
| | - Yanhua Li
- Department of Hematology and Oncology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355 Luding Road, Shanghai 200061, China; (S.J.); (X.D.); (C.H.); (Y.J.); (J.Y.); (X.L.); (Y.L.); (T.Z.); (S.L.)
| | - Ting Zhang
- Department of Hematology and Oncology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355 Luding Road, Shanghai 200061, China; (S.J.); (X.D.); (C.H.); (Y.J.); (J.Y.); (X.L.); (Y.L.); (T.Z.); (S.L.)
| | - Shanshan Li
- Department of Hematology and Oncology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355 Luding Road, Shanghai 200061, China; (S.J.); (X.D.); (C.H.); (Y.J.); (J.Y.); (X.L.); (Y.L.); (T.Z.); (S.L.)
| | - Zhibao Lv
- Department of Pediatric Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, No. 355 Luding Road, Shanghai 200061, China;
| | - Yijin Gao
- Department of Hematology and Oncology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, No. 1678 Dongfang Road, Shanghai 200127, China;
| |
Collapse
|
28
|
Kirchner K, Seidel C, Paulsen FO, Sievers B, Bokemeyer C, Lessel D. Further Association of Germline CHEK2 Loss-of-Function Variants with Testicular Germ Cell Tumors. J Clin Med 2023; 12:7065. [PMID: 38002677 PMCID: PMC10672725 DOI: 10.3390/jcm12227065] [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: 07/14/2023] [Revised: 10/17/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Testicular germ cell tumors (TGCTs) represent the most frequent malignancy in young adult men and have one the highest heritability rates among all cancers. A recent multicenter case-control study identified CHEK2 as the first moderate-penetrance TGCT predisposition gene. Here, we analyzed CHEK2 in 129 TGCT cases unselected for age of onset, histology, clinical outcome, and family history of any cancer, and the frequency of identified variants was compared to findings in 27,173 ancestry-matched cancer-free men. We identified four TGCT cases harboring a P/LP variant in CHEK2 (4/129, 3.10%), which reached statistical significance (p = 0.0191; odds ratio (OR), 4.06; 95% CI, 1.59-10.54) as compared to the control group. Cases with P/LP variants in CHEK2 developed TGCT almost 6 years earlier than individuals with CHEK2 wild-type alleles (5.67 years; 29.5 vs. 35.17). No association was found between CHEK2 status and further clinical and histopathological characteristics, including histological subtypes, the occurrence of aggressive TGCT, family history of TGCT, and family history of any cancer. In addition, we found significant enrichment for the low-penetrance CHEK2 variant p.Ile157Thr (p = 0.0259; odds ratio (OR), 3.69; 95% CI, 1.45-9.55). Thus, we provide further independent evidence of CHEK2 being a moderate-penetrance TGCT predisposition gene.
Collapse
Affiliation(s)
- Kira Kirchner
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (K.K.); (B.S.)
| | - Christoph Seidel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (C.S.); (F.-O.P.); (C.B.)
| | - Finn-Ole Paulsen
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (C.S.); (F.-O.P.); (C.B.)
| | - Bianca Sievers
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (K.K.); (B.S.)
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (C.S.); (F.-O.P.); (C.B.)
| | - Davor Lessel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (K.K.); (B.S.)
- Institute of Human Genetics, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| |
Collapse
|
29
|
Bremmer F, Pongratanakul P, Skowron M, Che Y, Richter A, Küffer S, Reuter-Jessen K, Bohnenberger H, Pauls S, Kresbach C, Schüller U, Stühler K, Ströbel P, Albers P, Nettersheim D. Characterizing the mutational burden, DNA methylation landscape, and proteome of germ cell tumor-related somatic-type malignancies to identify the tissue-of-origin, mechanisms of therapy resistance, and druggable targets. Br J Cancer 2023; 129:1580-1589. [PMID: 37726478 PMCID: PMC10645790 DOI: 10.1038/s41416-023-02425-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 08/22/2023] [Accepted: 09/04/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Germ cell tumors (GCT) might undergo transformation into a somatic-type malignancy (STM), resulting in a cell fate switch to tumors usually found in somatic tissues, such as rhabdomyosarcomas or adenocarcinomas. STM is associated with a poor prognosis, but the molecular and epigenetic mechanisms triggering STM are still enigmatic, the tissue-of-origin is under debate and biomarkers are lacking. METHODS To address these questions, we characterized a unique cohort of STM tissues on mutational, epigenetic and protein level using modern and high-throughput methods like TSO assays, 850k DNA methylation arrays and mass spectrometry. RESULTS AND CONCLUSIONS For the first time, we show that based on DNA methylation and proteome data carcinoma-related STM more closely resemble yolk-sac tumors, while sarcoma-related STM resemble teratoma. STM harbor mutations in FGF signaling factors (FGF6/23, FGFR1/4) highlighting the corresponding pathway as a therapeutic target. Furthermore, STM utilize signaling pathways, like AKT, FGF, MAPK, and WNT to mediate molecular functions coping with oxidative stress, toxin transport, DNA helicase activity, apoptosis and the cell cycle. Collectively, these data might explain the high therapy resistance of STM. Finally, we identified putative novel biomarkers secreted by STM, like EFEMP1, MIF, and DNA methylation at specific CpG dinucleotides.
Collapse
Affiliation(s)
- Felix Bremmer
- Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Pailin Pongratanakul
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Margaretha Skowron
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Yue Che
- Department of Urology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Annika Richter
- Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Stefan Küffer
- Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Hanibal Bohnenberger
- Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Stella Pauls
- Molecular Proteomics Laboratory (MPL), Biological and Medical Research Centre (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Catena Kresbach
- Institute of Neuropathology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Schüller
- Institute of Neuropathology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory (MPL), Biological and Medical Research Centre (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Peter Albers
- Department of Urology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel Nettersheim
- Department of Urology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
30
|
Patel D, Tayade S, Sharma S, Reddy LS. Immature Teratoma: A Case Report of a Monster Tumor in the Pediatric Age Group. Cureus 2023; 15:e48989. [PMID: 38111418 PMCID: PMC10726097 DOI: 10.7759/cureus.48989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/18/2023] [Indexed: 12/20/2023] Open
Abstract
Immature teratoma is a rare type of germ cell tumor containing embryonic tissues that may be malignant. It usually occurs in young women and affects the ovaries. Teratomas exhibit benign clinical behavior, but they can return as teratomas or with malignant components, and in a small subset of individuals, the prognosis may be deadly. We will discuss a case of a 9-year-old female child who presented with pain and a huge lump in the lower abdomen that was suggestive of an ovarian dermoid cyst or a germ cell tumor on computed tomography (CT) abdomen pelvis and underwent exploratory laparotomy and debulking surgery. Histopathology results indicated that she had a grade 3 immature teratoma. Postoperatively, the patient received 3 cycles of bleomycin, etoposide, and cisplatin (BEP) as adjuvant chemotherapy with a good response. She is currently under regular follow-up and has no evidence of recurrence or metastasis. This case illustrates the importance of early diagnosis and treatment of immature teratoma, which can be cured with surgery and chemotherapy. It also highlights the challenges of managing such a large tumor in a pediatric patient.
Collapse
Affiliation(s)
- Drashti Patel
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Surekha Tayade
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sakshi Sharma
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Lucky Srivani Reddy
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
31
|
Das DV, Jabbar PK, Gomez R, Nambisan B, Bhuvitha MS, Nair A, Jayakumari C. Prevalence, distribution, and risk markers for the development of gonadal germ cell tumors in patients with certain types of disorders of sexual differentiation with Y chromosome - A retrospective study. Indian J Cancer 2023; 60:464-474. [PMID: 38078466 DOI: 10.4103/ijc.ijc_1218_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 02/04/2021] [Indexed: 01/24/2024]
Abstract
PURPOSE To study the prevalence, subtypes, and risk markers for the development of gonadal germ cell tumors (GCT's) among disorders of sexual differentiation (DSD) patients with the Y chromosome. MATERIALS AND METHOD Design: A retrospective review of the patient's case records from 2010 to 2020 in Government Medical College, Thiruvananthapuram, India was studied. The study participants included 54 subjects with DSD containing the Y chromosome. Demographic data, external masculinization scoring, associated congenital anomalies, karyotyping, intraoperative findings such as gonadal location and internal genital ducts, histopathology of the resected gonads, and its immunohistochemistry were collected. The prevalence of gonadal GCT's was estimated from paraffin-embedded gonadectomy samples (S = 82). RESULTS The median age of occurrence of gonadal GCT's was 18 years. The prevalence of malignant gonadal GCT's was highest among the PAIS group (19.2%) followed by gonadal dysgenesis (15.8% each in MGD and CGD) and least among CAIS (7.7%) (p < 0.01). The most common type of malignant gonadal GCT's in the descending order of frequency was dysgerminoma, seminoma, mixed GCT, and yolk sac tumor. Multivariance logistic analysis showed post-puberty and the presence of congenital anomalies were associated with the occurrence of gonadal GCT's ( P < 0.01). CONCLUSION The overall prevalence of gonadal GCT's (malignant and premalignant) among DSD with Y chromosomes is nearly 25%. Dysgerminoma is the most common malignant gonadal GCT's. Age at or above 18 years and the presence of congenital anomalies like renal agenesis, retroperitoneal vascular defects, and congenital diaphragmatic hernia were independent risk markers for the development of gonadal GCT's.
Collapse
Affiliation(s)
- Darvin V Das
- Department of Endocrinology, Government Medical College, Thiruvanathapuram, Kerala, India
| | - P K Jabbar
- Department of Endocrinology, Government Medical College, Thiruvanathapuram, Kerala, India
| | - Ramesh Gomez
- Department of Endocrinology, Government Medical College, Thiruvanathapuram, Kerala, India
| | - Bindu Nambisan
- Department of Obstetrics and Gynecology, Government Medical College, Thiruvanathapuram, Kerala, India
| | - M S Bhuvitha
- Department of Pathology, Government Medical College, Thiruvanathapuram, Kerala, India
| | - Abilash Nair
- Department of Endocrinology, Government Medical College, Thiruvanathapuram, Kerala, India
| | - C Jayakumari
- Department of Endocrinology, Government Medical College, Thiruvanathapuram, Kerala, India
| |
Collapse
|
32
|
Abdellateif MS, Bayoumi AK, Mohammed MA. c-Kit Receptors as a Therapeutic Target in Cancer: Current Insights. Onco Targets Ther 2023; 16:785-799. [PMID: 37790582 PMCID: PMC10544070 DOI: 10.2147/ott.s404648] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/19/2023] [Indexed: 10/05/2023] Open
Abstract
c-Kit is a type III receptor tyrosine kinase (RTK) that has an essential role in various biological functions including gametogenesis, melanogenesis, hematopoiesis, cell survival, and apoptosis. c-KIT aberrations, either overexpression or loss-of-function mutations, have been implicated in the pathogenesis and development of many cancers, including gastrointestinal stromal tumors, mastocytosis, acute myeloid leukemia, breast, thyroid, and colorectal cancer, making c-KIT an attractive molecular target for the treatment of cancers. Therefore, a lot of effort has been put into investigating the utility of tyrosine kinase inhibitors for the management of c-KIT mutated tumors. This review of the literature illustrates the role of c-KIT mutations in many cancers, aiming to provide insights into the role of TKIs as a therapeutic option for cancer patients with c-KIT aberrations. In conclusion, c-KIT is implicated in different types of cancer, and it could be a successful molecular target; however, proper detection of the underlying mutation type is required before starting the appropriate personalized therapy.
Collapse
Affiliation(s)
- Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| | - Ahmed K Bayoumi
- Paediatric Oncology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
- Children’s Cancer Hospital 57357, Cairo, 11617, Egypt
| | - Mohammed Aly Mohammed
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| |
Collapse
|
33
|
García-Andrade F, Vigueras-Villaseñor RM, Chávez-Saldaña MD, Rojas-Castañeda JC, Bahena-Ocampo IU, Aréchaga-Ocampo E, Flores-Fortis M, Díaz-Chávez J, Herrera LA, Landero-Huerta DA. Molecular Characterization of Patients with Cryptorchidism: Preliminary Search for an Expression Profile Related to That of Testicular Germ-Cell Tumors. Diagnostics (Basel) 2023; 13:3020. [PMID: 37761387 PMCID: PMC10529510 DOI: 10.3390/diagnostics13183020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/30/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Cryptorchidism (CO) is a risk factor for the development of testicular germ-cell tumors (TGCT). This is supported by reports showing the persistence of gonocytes in CO patients. These cells are proposed to be related to the development of germ-cell neoplasia in situ (GCNIS), which is considered the precursor stage/lesion of TGCT. Therefore, it is proposed that some patients with CO could express some molecular markers related to TGCT. In this study, we analyzed testicular tissue samples from CO, TGCT, and controls. We determined the expression of POU5F1, PLAP, and KIT by immunohistochemistry and that of the hsa-miR-371-373 cluster, hsa-miR-367, and LATS2, PTEN, and IGFR1 genes by RT-qPCR. We then carried out a bioinformatic analysis to identify other possible candidate genes as tumor biomarkers. We found that 16.7% (2/12) of the CO patients presented increased expression of POU5F1, KIT, PLAP, hsa-miR-371-373, and hsa-miR-367 and decreased expression of LATS2 and IGF1R. Finally, the genes ARID4B, GALNT3, and KPNA6 were identified as other possible candidate tumor biomarkers. This is the first report describing the expression of the hsa-miR-371-373 cluster, hsa-miR-367, LATS2, and IGF1R in the testicular tissues of two CO patients with cells immune-positive to POU5F1, PLAP, and KIT, which is similar to what is observed in TGCT.
Collapse
Affiliation(s)
- Fabiola García-Andrade
- Laboratorio de Biología de la Reproducción, Instituto Nacional de Pediatría, Ciudad de Mexico 04530, Mexico
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Ciudad de Mexico 09310, Mexico
| | | | | | | | - Ivan Uriel Bahena-Ocampo
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Ciudad de Mexico 09310, Mexico
| | - Elena Aréchaga-Ocampo
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, Ciudad de Mexico 05348, Mexico
| | - Mauricio Flores-Fortis
- Posgrado en Ciencias Naturales e Ingeniería, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, Ciudad de Mexico 05348, Mexico
| | - José Díaz-Chávez
- Unidad de Investigación en Cáncer, Instituto de Investigaciones Biomédicas-Universidad Nacional Autónoma de México, Instituto Nacional de Cancerología, Ciudad de Mexico 14080, Mexico
| | - Luis Alonso Herrera
- Unidad de Investigación en Cáncer, Instituto de Investigaciones Biomédicas-Universidad Nacional Autónoma de México, Instituto Nacional de Cancerología, Ciudad de Mexico 14080, Mexico
- Escuela de Medicina y Ciencias de la Salud-Tecnológico de Monterrey, Ciudad de Mexico 14380, Mexico
| | | |
Collapse
|
34
|
Törzsök P, Oswald D, Dieckmann KP, Angerer M, Scherer LC, Tymoszuk P, Kunz Y, Pinggera GM, Lusuardi L, Horninger W, Pichler R. Subsets of preoperative sex hormones in testicular germ cell cancer: a retrospective multicenter study. Sci Rep 2023; 13:14604. [PMID: 37669975 PMCID: PMC10480169 DOI: 10.1038/s41598-023-41915-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/04/2023] [Indexed: 09/07/2023] Open
Abstract
Preoperative homeostasis of sex hormones in testicular germ cell tumor (TGCT) patients is scarcely characterized. We aimed to explore regulation of sex hormones and their implications for histopathological parameters and prognosis in TGCT using a data-driven explorative approach. Pre-surgery serum concentrations of luteinizing hormone (LH), follicle-stimulating hormone (FSH), testosterone (T), estradiol (E2) and prolactin were measured in a retrospective multicenter TGCT cohort (n = 518). Clusters of patients were defined by latent class analysis. Clinical, pathologic and survival parameters were compared between the clusters by statistical hypothesis testing, Random Forest modeling and Peto-Peto test. Cancer tissue expression of sex hormone-related genes was explored in the publicly available TCGA cohort (n = 149). We included 354 patients with pure seminoma and 164 patients with non-seminomatous germ cell tumors (NSGCT), with a median age of 36 years. Three hormonal clusters were defined: 'neutral' (n = 228) with normal sex hormone homeostasis, 'testicle' (n = 91) with elevated T and E2, low pituitary hormones, and finally 'pituitary' subset (n = 103) with increased FSH and LH paralleled by low-to-normal levels of the gonadal hormones. Relapse-free survival in the hormonal subsets was comparable (p = 0.64). Cancer tissue expression of luteinizing hormone- and follicle-stimulating hormone-coding genes was significantly higher in seminomas, while genes of T and E2 biosynthesis enzymes were strongly upregulated in NSGCT. Substantial percentages of TGCT patients are at increased risk of sex hormone dysfunction at primary diagnosis before orchiectomy. TGCT may directly influence systemic hormonal homeostasis by in-situ synthesis of sex hormones.
Collapse
Affiliation(s)
- Péter Törzsök
- Department of Urology and Andrology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - David Oswald
- Department of Urology and Andrology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | | | - Markus Angerer
- Department of Urology, Hodentumorzentrum, Asklepios Klinik Altona, Hamburg, Germany
| | - Lukas Christian Scherer
- Department of Urology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | | | - Yannic Kunz
- Department of Urology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Germar-Michael Pinggera
- Department of Urology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Lukas Lusuardi
- Department of Urology and Andrology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Wolfgang Horninger
- Department of Urology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Renate Pichler
- Department of Urology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| |
Collapse
|
35
|
Elesawy AE, Abulsoud AI, Moustafa HAM, Elballal MS, Sallam AAM, Elazazy O, El-Dakroury WA, Abdel Mageed SS, Abdelmaksoud NM, Midan HM, Shahin RK, Elrebehy MA, Nassar YA, Elazab IM, Elballal AS, Elballal MS, Doghish AS. miRNAs orchestration of testicular germ cell tumors - Particular emphasis on diagnosis, progression and drug resistance. Pathol Res Pract 2023; 248:154612. [PMID: 37327566 DOI: 10.1016/j.prp.2023.154612] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/18/2023]
Abstract
Testicular cancer (TC) is one of the most frequently incident solid tumors in males. A growing prevalence has been documented in developed countries. Although recent advances have made TC an exceedingly treatable cancer, numerous zones in TC care still have divisive treatment decisions. In addition to physical examination and imaging techniques, conventional serum tumor markers have been traditionally used for the diagnosis of testicular germ cell tumors (TGCT). Unlike other genital and urinary tract tumors, recent research methods have not been broadly used in TGCTs. Even though several challenges in TC care must be addressed, a dedicated group of biomarkers could be particularly beneficial to help classify patient risk, detect relapse early, guide surgery decisions, and tailor follow-up. Existing tumor markers (Alpha-fetoprotein, human chorionic gonadotrophin, and lactate dehydrogenase) have limited accuracy and sensitivity when used as diagnostic, prognostic, or predictive markers. At present, microRNAs (miRNA or miR) play a crucial role in the process of several malignancies. The miRNAs exhibit pronounced potential as novel biomarkers since they reveal high stability in body fluids, are easily detected, and are relatively inexpensive in quantitative assays. In this review, we aimed to shed light on the recent novelties in developing microRNAs as diagnostic and prognostic markers in TC and discuss their clinical applications in TC management.
Collapse
Affiliation(s)
- Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt.
| | - Hebatallah Ahmed Mohamed Moustafa
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Al-Aliaa M Sallam
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | | | - Heba M Midan
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Reem K Shahin
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Yara A Nassar
- Biology Department, School of Biotechnology, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Ibrahim M Elazab
- Biochemistry Department, Faculty of Pharmacy, Tanta University, Egypt
| | - Ahmed S Elballal
- Department of Dentistry, Medical Administration, University of Sadat City Menoufia 32897, Egypt
| | | | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| |
Collapse
|
36
|
Yu H, Feng B, Zhang Y, Lyu J. Development and validation of a nomogram for predicting the overall survival of patients with testicular cancer. Cancer Med 2023; 12:15567-15578. [PMID: 37264772 PMCID: PMC10417196 DOI: 10.1002/cam4.6203] [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: 03/28/2023] [Revised: 04/25/2023] [Accepted: 05/23/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND The purpose of this study was to develop and validate a nomogram to predict survival in testicular cancer patients. METHODS Testicular cancer patients diagnosed between 2004 and 2015 from the Surveillance, Epidemiology, and End Results (SEER) database were selected for this study. A random sampling method was used to divide patients into training and validation cohorts, which accounted for 30% and 70% of the total sample, respectively. The nomogram was developed using the training cohort and evaluated using the C index, calibration chart, and area under the receiver operating characteristic curve (AUC). RESULTS Seven risk factors that affect the survival of testicular cancer patients (AJCC stage, marital status, age at diagnosis, race, SEER historic stage A, surgery status, and origin) were identified using Cox proportional hazard regression analysis. The nomogram has a higher C index (0.897) and AUC when compared with the AJCC staging system. The results of the calibration chart of the nomogram show that the predicted survival of testicular cancer patients at 3, 5, and 10 years after diagnosis is very close to their actual survival. CONCLUSIONS We developed and validated a nomogram for predicting the survival rate of testicular cancer patients at 3, 5, and 10 years after diagnosis. This nomogram has better discrimination, calibration, and clinical validity than the AJCC staging system. This indicates that the nomogram can be used to predict the survival of testicular cancer patients effectively, and provide a reference for patient treatment strategies.
Collapse
Affiliation(s)
- Haohui Yu
- Department of Medical AdministrationThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Bin Feng
- Department of Medical AdministrationThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Yunrui Zhang
- Department of Medical AdministrationThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Jun Lyu
- Department of Medical AdministrationThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| |
Collapse
|
37
|
Funke K, Einsfelder U, Hansen A, Arévalo L, Schneider S, Nettersheim D, Stein V, Schorle H. Genome-scale CRISPR screen reveals neddylation to contribute to cisplatin resistance of testicular germ cell tumours. Br J Cancer 2023; 128:2270-2282. [PMID: 37024667 PMCID: PMC10241889 DOI: 10.1038/s41416-023-02247-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Type II testicular germ cell tumours (TGCT) are the most prevalent tumours in young men. Patients suffering from cisplatin-resistant TGCTs are facing very poor prognosis demanding novel therapeutic options. Neddylation is a known posttranslational modification mediating many important biological processes, including tumorigenesis. Overactivation of the neddylation pathway promotes carcinogenesis and tumour progression in various entities by inducing proteasomal degradation of tumour suppressors (e.g., p21, p27). METHODS We used a genome-scale CRISPR/Cas9 activation screen to identify cisplatin resistance factors. TGCT cell lines were treated with the neddylation inhibitor (MLN4924)/cisplatin/combination and investigated for changes in viability (XTT assay), apoptosis/cell cycle (flow cytometry) as well as in the transcriptome (3'mRNA sequencing). RESULTS NAE1 overexpression was detected in cisplatin-resistant colonies from the CRISPR screen. Inhibition of neddylation using MLN4924 increased cisplatin cytotoxicity in TGCT cell lines and sensitised cisplatin-resistant cells towards cisplatin. Apoptosis, G2/M-phase cell cycle arrest, γH2A.X/P27 accumulation and mesoderm/endoderm differentiation were observed in TGCT cells, while fibroblast cells were unaffected. CONCLUSIONS We identified overactivation of neddylation as a factor for cisplatin resistance in TGCTs and highlighted the additive effect of NAE1 inhibition by MLN4924 in combination with cisplatin as a novel treatment option for TGCTs.
Collapse
Affiliation(s)
- Kai Funke
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Ulf Einsfelder
- Institute of Physiology II, University Hospital Bonn, Bonn, Germany
| | - Aylin Hansen
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Lena Arévalo
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Simon Schneider
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Daniel Nettersheim
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Valentin Stein
- Institute of Physiology II, University Hospital Bonn, Bonn, Germany
| | - Hubert Schorle
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
38
|
Pinto MT, Eiras Martins G, Vieira AGS, Galvão JMS, de Pádua Souza C, Macedo CRPD, Lopes LF. Molecular Biology of Pediatric and Adult Ovarian Germ Cell Tumors: A Review. Cancers (Basel) 2023; 15:cancers15112990. [PMID: 37296950 DOI: 10.3390/cancers15112990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 06/12/2023] Open
Abstract
Ovarian germ cell tumors (OGCTs) are rare in adults; indeed, they occur predominantly in children, adolescents, and young adults, and they account for approximately 11% of cancer diagnoses in these groups. Because OGCTs are rare tumors, our current understanding of them is sparse; this is because few studies have investigated the molecular basis of pediatric and adult cancers. Here, we review the etiopathogenesis of OGCTs in children and adults, and we address the molecular landscape of these tumors, including integrated genomic analysis, microRNAs, DNA methylation, the molecular implications of treatment resistance, and the development of in vitro and in vivo models. An elucidation of potential molecular alterations may provide a novel field for understanding the pathogenesis, tumorigenesis, diagnostic markers, and genetic peculiarity of the rarity and complexity of OGCTs.
Collapse
Affiliation(s)
| | - Gisele Eiras Martins
- Brazilian Childhood Germ Cell Tumor Study Group, The Brazilian Pediatric Oncology Society (SOBOPE), Barretos 14784400, Brazil
- Children's Cancer Hospital from Hospital de Amor, Barretos 14784400, Brazil
| | - Ana Glenda Santarosa Vieira
- Brazilian Childhood Germ Cell Tumor Study Group, The Brazilian Pediatric Oncology Society (SOBOPE), Barretos 14784400, Brazil
- Children's Cancer Hospital from Hospital de Amor, Barretos 14784400, Brazil
| | | | | | - Carla Renata Pacheco Donato Macedo
- Brazilian Childhood Germ Cell Tumor Study Group, The Brazilian Pediatric Oncology Society (SOBOPE), Barretos 14784400, Brazil
- Pediatric Oncology Department, IOP/GRAACC/Federal University of Sao Paulo, Sao Paulo 04038001, Brazil
| | - Luiz Fernando Lopes
- Brazilian Childhood Germ Cell Tumor Study Group, The Brazilian Pediatric Oncology Society (SOBOPE), Barretos 14784400, Brazil
- Children's Cancer Hospital from Hospital de Amor, Barretos 14784400, Brazil
| |
Collapse
|
39
|
Petrovic M, Simic T, Djukic T, Radic T, Savic-Radojevic A, Zekovic M, Durutovic O, Janicic A, Milojevic B, Kajmakovic B, Zivkovic M, Bojanic N, Bumbasirevic U, Coric V. The Polymorphisms in GSTO Genes ( GSTO1 rs4925, GSTO2 rs156697, and GSTO2 rs2297235) Affect the Risk for Testicular Germ Cell Tumor Development: A Pilot Study. Life (Basel) 2023; 13:1269. [PMID: 37374052 DOI: 10.3390/life13061269] [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: 02/15/2023] [Revised: 04/24/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Members of the omega class of glutathione transferases (GSTs), GSTO1, and GSTO2, catalyze a range of reduction reactions as a part of the antioxidant defense system. Polymorphisms of genes encoding antioxidant proteins and the resultant altered redox profile have already been associated with the increased risk for testicular germ cell cancer (GCT) development. The aim of this pilot study was to assess the individual, combined, haplotype, and cumulative effect of GSTO1rs4925, GSTO2rs156697, and GSTO2rs2297235 polymorphisms with the risk for testicular GCT development, in 88 patients and 96 matched controls, through logistic regression models. We found that carriers of the GSTO1*C/A*C/C genotype exhibited an increased risk for testicular GCT development. Significant association with increased risk of testicular GCT was observed in carriers of GSTO2rs2297235*A/G*G/G genotype, and in carriers of combined GSTO2rs156697*A/G*G/G and GSTO2rs2297235*A/G*G/G genotypes. Haplotype H7 (GSTO1rs4925*C/GSTO2rs2297235*G/GSTO2rs156697*G) exhibited higher risk of testicular GCT, however, without significant association (p > 0.05). Finally, 51% of testicular GCT patients were the carriers of all three risk-associated genotypes, with 2.5-fold increased cumulative risk. In conclusion, the results of this pilot study suggest that GSTO polymorphisms might affect the protective antioxidant activity of GSTO isoenzymes, therefore predisposing susceptible individuals toward higher risk for testicular GCT development.
Collapse
Affiliation(s)
- Milos Petrovic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Tatjana Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Department of Medical Sciences, Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| | - Tatjana Djukic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Tanja Radic
- Institute of Mental Health, 11000 Belgrade, Serbia
| | - Ana Savic-Radojevic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Milica Zekovic
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Otas Durutovic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandar Janicic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Bogomir Milojevic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Boris Kajmakovic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Marko Zivkovic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Nebojsa Bojanic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Uros Bumbasirevic
- Clinic of Urology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Vesna Coric
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
40
|
Jabłońska I, Goławski M, Nowicka E, Drosik-Rutowicz K, Trybus A, Tarnawski R, Miszczyk M. Intracranial Germinoma-Association between Delayed Diagnosis, Altered Clinical Manifestations, and Prognosis. Cancers (Basel) 2023; 15:2789. [PMID: 37345126 DOI: 10.3390/cancers15102789] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Intracranial germinoma is a rare malignant neoplasm of the central nervous system (CNS) that occurs in children and young adults. The aim of our study was to assess the initial manifestation of the disease, and to find differences in outcomes dependent on time of diagnosis. METHODS The study group consisted of 35 consecutive patients (adults and children) who were treated for intracranial germinoma with radiotherapy at a tertiary centre, and their data were retrospectively collected. We evaluated time from the first symptoms to diagnosis and divided patients into early and delayed diagnosis groups. Delayed diagnosis has been defined as the time from initial presentation to final diagnosis longer than six months. RESULTS A total of 17 (48.6%) of the patients had delayed diagnoses. Patient survival data spanned a median of six (interquartile range 3-12) years. At the time of the diagnosis, patients presented exclusively neurological symptoms in 16 (45.7%) cases, exclusively endocrinological symptoms in five (14.3%) cases, and mixed symptoms in the remaining cases (n = 14; 40.0%). Patients with neurological symptoms had shorter time (p < 0.001) from first symptoms to the final diagnosis (5.91 months) than in patients without them (19.44 months). The delayed diagnosis group presented significantly smaller tumour size (mean maximal dimension 2.35 cm) compared to early diagnosis group (3.1 cm). The 5-year and 10-year survival rates of our patients were 94.3% and 83.4%, respectively. Patients with a delayed diagnosis (n = 17) had a significantly worse (p = 0.02) 10-year OS (63%) compared to the early diagnosis group (n = 18; OS = 100%). Importantly, in five patients (14.29%), initial manifestation occurred before radiological signs of the disease. CONCLUSION Our study stresses the need for timely diagnosis in intracranial germinoma, as a delay has a significant impact on the prognosis. In particular, if the tumour is small or causes exclusively endocrinological symptoms, the diagnosis may be difficult and delayed.
Collapse
Affiliation(s)
- Iwona Jabłońska
- IIIrd Department of Radiotherapy and Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland
| | - Marcin Goławski
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| | - Elżbieta Nowicka
- IIIrd Department of Radiotherapy and Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland
| | - Katarzyna Drosik-Rutowicz
- Ist Department of Radiotherapy and Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland
| | - Anna Trybus
- IIIrd Department of Radiotherapy and Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland
| | - Rafał Tarnawski
- IIIrd Department of Radiotherapy and Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland
| | - Marcin Miszczyk
- IIIrd Department of Radiotherapy and Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland
| |
Collapse
|
41
|
Xu L, Pierce JL, Sanchez A, Chen KS, Shukla AA, Fustino NJ, Stuart SH, Bagrodia A, Xiao X, Guo L, Krailo MD, Shaikh F, Billmire DF, Pashankar F, Bestrashniy J, Oosterhuis JW, Gillis AJM, Xie Y, Teot L, Mora J, Poynter JN, Rakheja D, Looijenga LHJ, Draper BW, Frazier AL, Amatruda JF. Integrated genomic analysis reveals aberrations in WNT signaling in germ cell tumors of childhood and adolescence. Nat Commun 2023; 14:2636. [PMID: 37149691 PMCID: PMC10164134 DOI: 10.1038/s41467-023-38378-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 04/26/2023] [Indexed: 05/08/2023] Open
Abstract
Germ cell tumors (GCTs) are neoplasms of the testis, ovary and extragonadal sites that occur in infants, children, adolescents and adults. Post-pubertal (type II) malignant GCTs may present as seminoma, non-seminoma or mixed histologies. In contrast, pre-pubertal (type I) GCTs are limited to (benign) teratoma and (malignant) yolk sac tumor (YST). Epidemiologic and molecular data have shown that pre- and post-pubertal GCTs arise by distinct mechanisms. Dedicated studies of the genomic landscape of type I and II GCT in children and adolescents are lacking. Here we present an integrated genomic analysis of extracranial GCTs across the age spectrum from 0-24 years. Activation of the WNT pathway by somatic mutation, copy-number alteration, and differential promoter methylation is a prominent feature of GCTs in children, adolescents and young adults, and is associated with poor clinical outcomes. Significantly, we find that small molecule WNT inhibitors can suppress GCT cells both in vitro and in vivo. These results highlight the importance of WNT pathway signaling in GCTs across all ages and provide a foundation for future efforts to develop targeted therapies for these cancers.
Collapse
Affiliation(s)
- Lin Xu
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Population & Data Sciences, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Joshua L Pierce
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Angelica Sanchez
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kenneth S Chen
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Abhay A Shukla
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nicholas J Fustino
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Blank Children's Hospital, Des Moines, IA, USA
| | - Sarai H Stuart
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Aditya Bagrodia
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Urology, University of California San Diego, San Diego, CA, USA
| | - Xue Xiao
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Population & Data Sciences, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lei Guo
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Population & Data Sciences, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mark D Krailo
- Department of Preventative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
- Children's Oncology Group, Monrovia, CA, USA
| | - Furqan Shaikh
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | - Farzana Pashankar
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - Ad J M Gillis
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Yang Xie
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Population & Data Sciences, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lisa Teot
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Jaume Mora
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - Jenny N Poynter
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Dinesh Rakheja
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Bruce W Draper
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA, USA
| | - A Lindsay Frazier
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - James F Amatruda
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, CA, USA.
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
42
|
Gayer FA, Henkel M, Luft J, Reichardt SD, Fichtner A, Legler TJ, Reichardt HM. The Subtype Identity of Testicular Cancer Cells Determines Their Immunostimulatory Activity in a Coculture Model. Cancers (Basel) 2023; 15:cancers15092619. [PMID: 37174085 PMCID: PMC10177190 DOI: 10.3390/cancers15092619] [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: 03/21/2023] [Revised: 04/20/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Testicular germ cell cancer (TGCC) is subdivided into several subtypes. While seminomatous germ cell tumors (SGCT) are characterized by an intensive infiltration of immune cells which constitute a pro-inflammatory tumor micromilieu (TME), immune cells in non-seminomatous germ cell tumors (NSGCT) are differently composed and less abundant. Previously, we have shown that the seminomatous cell line TCam-2 promotes T cell and monocyte activation in a coculture model, resulting in mutual interactions between both cell types. Here we set out to compare this feature of TCam-2 cells with the non-seminomatous cell line NTERA-2. Peripheral blood T cells or monocytes cocultured with NTERA-2 cells failed to secrete relevant amounts of pro-inflammatory cytokines, and significantly downregulated the expression of genes encoding activation markers and effector molecules. In contrast, immune cells cocultured with TCam-2 cells produced IL-2, IL-6 and TNFα, and strongly upregulated the expression of multiple pro-inflammatory genes. Furthermore, the expression of genes involved in proliferation, stemness and subtype specification remained unaltered in NTERA-2 cells during coculture with T cells or monocytes, indicating the absence of mutual interactions. Collectively, our findings uncover fundamental differences between SGCT and NSGCT in their capability to generate a pro-inflammatory TME, which possibly impacts the clinical features and prognosis of both TGCC subtypes.
Collapse
Affiliation(s)
- Fabian A Gayer
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
- Clinic of Urology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Miriam Henkel
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Juliane Luft
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Sybille D Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Alexander Fichtner
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Tobias J Legler
- Department of Transfusion Medicine, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Holger M Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, 37073 Göttingen, Germany
| |
Collapse
|
43
|
Kunvariya AD, Dave SA, Modi ZJ, Patel PK, Sagar SR. Exploration of multifaceted molecular mechanism of angiotensin-converting enzyme 2 (ACE2) in pathogenesis of various diseases. Heliyon 2023; 9:e15644. [PMID: 37153428 PMCID: PMC10160752 DOI: 10.1016/j.heliyon.2023.e15644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 05/09/2023] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) is a homolog of ACE (a transmembrane bound dipeptidyl peptidase enzyme). ACE2 converts angiotensinogen to the heptapeptide angiotensin-(1-7). ACE2 and its product, angiotensin-(1-7), have counteracting effects against the adverse actions of other members of renin-angiotensin system (RAS). ACE2 and its principal product, angiotensin-(1-7), were considered an under recognized arm of the RAS. The COVID-19 pandemic brought to light this arm of RAS with special focus on ACE2. Membrane bound ACE2 serves as a receptor for SARS-CoV-2 viral entry through spike proteins. Apart from that, ACE2 is also involved in the pathogenesis of various other diseases like cardiovascular disease, cancer, respiratory diseases, neurodegenerative diseases and infertility. The present review focuses on the molecular mechanism of ACE2 in neurodegenerative diseases, cancer, cardiovascular disease, infertility and respiratory diseases, including SARS-CoV-2. This review summarizes unveiled roles of ACE2 in the pathogenesis of various diseases which further provides intriguing possibilities for the use of ACE2 activators and RAS modulating agents for various diseases.
Collapse
Affiliation(s)
- Aditi D. Kunvariya
- Department of Pharmaceutical Chemistry, L.J. Institute of Pharmacy, L J University, Ahmedabad 382 210, India
| | - Shivani A. Dave
- Department of Pharmaceutical Chemistry, L.J. Institute of Pharmacy, L J University, Ahmedabad 382 210, India
| | - Zeal J. Modi
- Department of Pharmaceutical Chemistry, L.J. Institute of Pharmacy, L J University, Ahmedabad 382 210, India
| | - Paresh K. Patel
- Department of Pharmaceutical Chemistry, L.J. Institute of Pharmacy, L J University, Ahmedabad 382 210, India
| | - Sneha R. Sagar
- Department of Pharmaceutical Chemistry, L.J. Institute of Pharmacy, L J University, Ahmedabad 382 210, India
- Corresponding author.
| |
Collapse
|
44
|
Radhakrishnan K, Luu M, Iaria J, Sutherland JM, McLaughlin EA, Zhu HJ, Loveland KL. Activin and BMP Signalling in Human Testicular Cancer Cell Lines, and a Role for the Nucleocytoplasmic Transport Protein Importin-5 in their Crosstalk. Cells 2023; 12:cells12071000. [PMID: 37048077 PMCID: PMC10093041 DOI: 10.3390/cells12071000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/05/2023] [Accepted: 03/09/2023] [Indexed: 03/29/2023] Open
Abstract
Testicular germ cell tumours (TGCTs) are the most common malignancy in young men. Originating from foetal testicular germ cells that fail to differentiate correctly, TGCTs appear after puberty as germ cell neoplasia in situ cells that transform through unknown mechanisms into distinct seminoma and non-seminoma tumour types. A balance between activin and BMP signalling may influence TGCT emergence and progression, and we investigated this using human cell line models of seminoma (TCam-2) and non-seminoma (NT2/D1). Activin A- and BMP4-regulated transcripts measured at 6 h post-treatment by RNA-sequencing revealed fewer altered transcripts in TCam-2 cells but a greater responsiveness to activin A, while BMP4 altered more transcripts in NT2/D1 cells. Activin significantly elevated transcripts linked to pluripotency, cancer, TGF-β, Notch, p53, and Hippo signalling in both lines, whereas BMP4 altered TGF-β, pluripotency, Hippo and Wnt signalling components. Dose-dependent antagonism of BMP4 signalling by activin A in TCam-2 cells demonstrated signalling crosstalk between these two TGF-β superfamily arms. Levels of the nuclear transport protein, IPO5, implicated in BMP4 and WNT signalling, are highly regulated in the foetal mouse germline. IPO5 knockdown in TCam-2 cells using siRNA blunted BMP4-induced transcript changes, indicating that IPO5 levels could determine TGF-β signalling pathway outcomes in TGCTs.
Collapse
Affiliation(s)
- Karthika Radhakrishnan
- Centre for Reproductive Health, Hudson Institute of Medical Research, 27-31 Kanooka Grove, Clayton, VIC 3168, Australia
- Correspondence: (K.R.); (K.L.L.)
| | - Michael Luu
- Centre for Reproductive Health, Hudson Institute of Medical Research, 27-31 Kanooka Grove, Clayton, VIC 3168, Australia
| | - Josie Iaria
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC 3050, Australia
| | - Jessie M. Sutherland
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, NSW 2305, Australia
- Hunter Medical Research Institute (HMRI), New Lambton Heights, NSW 2305, Australia
| | - Eileen A. McLaughlin
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, NSW 2305, Australia
- Hunter Medical Research Institute (HMRI), New Lambton Heights, NSW 2305, Australia
- Faculty of Science, Medicine and Health, University of Wollongong, Gwynneville, NSW 2500, Australia
| | - Hong-Jian Zhu
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC 3050, Australia
| | - Kate L. Loveland
- Centre for Reproductive Health, Hudson Institute of Medical Research, 27-31 Kanooka Grove, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3800, Australia
- Correspondence: (K.R.); (K.L.L.)
| |
Collapse
|
45
|
Barchi M, Guida E, Dolci S, Rossi P, Grimaldi P. Endocannabinoid system and epigenetics in spermatogenesis and testicular cancer. VITAMINS AND HORMONES 2023; 122:75-106. [PMID: 36863802 DOI: 10.1016/bs.vh.2023.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
In mammals, male germ cell development starts during fetal life and is carried out in postnatal life with the formation of sperms. Spermatogenesis is the complex and highly orderly process during which a group of germ stem cells is set at birth, starts to differentiate at puberty. It proceeds through several stages: proliferation, differentiation, and morphogenesis and it is strictly regulated by a complex network of hormonal, autocrine and paracrine factors and it is associated with a unique epigenetic program. Altered epigenetic mechanisms or inability to respond to these factors can impair the correct process of germ development leading to reproductive disorders and/or testicular germ cell cancer. Among factors regulating spermatogenesis an emerging role is played by the endocannabinoid system (ECS). ECS is a complex system comprising endogenous cannabinoids (eCBs), their synthetic and degrading enzymes, and cannabinoid receptors. Mammalian male germ cells have a complete and active ECS which is modulated during spermatogenesis and that crucially regulates processes such as germ cell differentiation and sperm functions. Recently, cannabinoid receptor signaling has been reported to induce epigenetic modifications such as DNA methylation, histone modifications and miRNA expression. Epigenetic modifications may also affect the expression and function of ECS elements, highlighting the establishment of a complex mutual interaction. Here, we describe the developmental origin and differentiation of male germ cells and testicular germ cell tumors (TGCTs) focusing on the interplay between ECS and epigenetic mechanisms involved in these processes.
Collapse
Affiliation(s)
- Marco Barchi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Eugenia Guida
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Susanna Dolci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Pellegrino Rossi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Paola Grimaldi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
46
|
Guo CC, Czerniak B. Reprint of: somatic-type malignancies in testicular germ cell tumors. Hum Pathol 2023; 133:102-114. [PMID: 36890027 DOI: 10.1016/j.humpath.2023.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 06/30/2022] [Indexed: 03/08/2023]
Abstract
The development of somatic-type malignancy (SM) in testicular germ cell tumor represents a major challenge in the diagnosis and treatment of testicular cancer. Most SMs are derived from teratoma, and the remainder is associated with yolk sac tumor. They occur more frequently in metastases than in primary testicular tumors. SMs demonstrate a variety of histologic types, including sarcoma, carcinoma, embryonic-type neuroectodermal tumor, nephroblastoma-like tumor, and hematologic malignancy. Sarcoma, particularly rhabdomyosarcoma, accounts for the majority of SMs in the primary testicular tumors, whereas carcinoma, particularly adenocarcinoma, is the most common SM in metastases. Although SMs derived from testicular germ cell tumors mimic their histologic counterparts in other organs with overlapping immunohistochemical profiles, isochromosome 12p is present in most SMs, which can be useful in the differential diagnosis. The presence of SM in the primary testicular tumor may not worsen the outcome, but the development of SM in metastasis is associated with a poor prognosis. Furthermore, somatic-type carcinoma is likely associated with a worse prognosis than somatic-type sarcoma. Although SMs respond poorly to the cisplatin-based chemotherapy, timely surgical resection is an effective treatment for most patients.
Collapse
Affiliation(s)
- Charles C Guo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77401, USA.
| | - Bogdan Czerniak
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77401, USA
| |
Collapse
|
47
|
Partenope C, Pozzobon G, Weber G, Carceller F, Albanese A. Implications of deferred diagnosis of paediatric intracranial germ cell tumours. Pediatr Blood Cancer 2023; 70:e30168. [PMID: 36582128 DOI: 10.1002/pbc.30168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/31/2022]
Abstract
AIMS This study analysed the clinical features of a cohort of children with intracranial germ cell tumours (IC-GCTs). We retrospectively reviewed timelag between symptoms onset, clinic-radiological findings, diagnosis and outcomes. METHODS Symptoms at diagnosis were divided into four groups: (1) raised intracranial pressure (RICP); (2) visual impairment; (3) endocrinopathies; (4) other. Total diagnostic interval (TDI), defined as the interval between symptom onset (including retrospective recall of symptoms) and definitive diagnosis of IC-GCT, was calculated and compared to survival rates. RESULTS Our cohort included 55 children with median follow-up of 78.9 months (0.5-249.9). The majority (63.6%) had germinomas and 10.9% were metastatic at diagnosis. IC-GCTs were suprasellar (41.8%), pineal (36.4%), bifocal (12.7%) or in atypical sites (9.1%). The most common presenting symptoms were related to RICP (43.6%); however, by the time of tumour diagnosis, 50.9% of patients had developed endocrine dysfunctions. All pineal GCTs manifested with RICP or visual impairment. All suprasellar GCTs presented with endocrinopathies. TDI ranged between 0.25 and 58.5 months (median 4 months). Pineal GCTs had the shortest TDI (median TDI 1 month versus 24 months in suprasellar GCTs, p < .001). TDI > 6 months was observed in 47.3% of patients and was significantly associated with endocrine presenting symptoms. No statistically significant difference was found in progression-free survival and overall survival between patients with TDI > 6 months and with TDI ≤ 6 months. CONCLUSION Approximately half of the IC-GCT patients in this cohort had TDI > 6 months. These presented mostly with endocrine deficits. TDI > 6 months was not associated with increased relapse or mortality rates.
Collapse
Affiliation(s)
- Cristina Partenope
- Department of Pediatrics, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Department of Paediatric Endocrinology, Royal Marsden NHS Foundation Trust, London, UK
| | - Gabriella Pozzobon
- Department of Pediatrics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giovanna Weber
- Department of Pediatrics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fernando Carceller
- Department of Paediatric and Adolescent Neuro-Oncology and Drug Development Team, Children and Young People's Unit, Royal Marsden Hospital and Institute of Cancer Research, Sutton, UK
| | - Assunta Albanese
- Department of Paediatric Endocrinology, Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
48
|
Ozgun G, Nappi L. Primary Mediastinal Germ Cell Tumors: A Thorough Literature Review. Biomedicines 2023; 11:biomedicines11020487. [PMID: 36831022 PMCID: PMC9953372 DOI: 10.3390/biomedicines11020487] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/31/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Primary mediastinal germ cell tumors (PMGCTs) are a rare type of cancer affecting young adults. They have different molecular and clinical features compared to testicular germ cell tumors. Non-seminoma PMGCTs have the shortest 5-year overall survival and the poorest prognosis among all of the germ cell tumor presentations, while seminomas share the same survival and prognosis as their testicular counterparts. There is an unmet need for better treatment options for patients with non-seminoma PMGCTs in both first-line and salvage therapy, as the available options are associated with underwhelming outcomes. Identifying biological and genetic factors to predict treatment responses would be helpful in improving the survival of these patients.
Collapse
|
49
|
Roelen BAJ, Chuva de Sousa Lopes SM. Stay on the road: from germ cell specification to gonadal colonization in mammals. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210259. [PMID: 36252219 PMCID: PMC9574628 DOI: 10.1098/rstb.2021.0259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The founder cells of the gametes are primordial germ cells (PGCs). In mammals, PGCs are specified early during embryonic development, at the boundary between embryonic and extraembryonic tissue, long before their later residences, the gonads, have developed. Despite the differences in form and behaviour when differentiated into oocytes or sperm cells, in the period between specification and gonadal colonization, male and female PGCs are morphologically indistinct and largely regulated by similar mechanisms. Here, we compare different modes and mechanisms that lead to the formation of PGCs, putting in context protocols that are in place to differentiate both human and mouse pluripotent stem cells into PGC-like cells. In addition, we review important aspects of the migration of PGCs to the gonadal ridges, where they undergo further sex-specific differentiation. Defects in migration need to be effectively corrected, as misplaced PGCs can become tumorigenic. Concluding, a combination of in vivo studies and the development of adequate innovative in vitro models, ensuring both robustness and standardization, are providing us with the tools for a greater understanding of the first steps of gametogenesis and to develop disease models to study the origin of germ cell tumours. This article is part of the theme issue ‘Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom’.
Collapse
Affiliation(s)
- Bernard A J Roelen
- Anatomy and Physiology, Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584CL Utrecht, The Netherlands.,Department of Biosciences, Biotechnologies & Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Susana M Chuva de Sousa Lopes
- Department of Biosciences, Biotechnologies & Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy.,Department of Anatomy and Embryology, Leiden University Medical Centre, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| |
Collapse
|
50
|
Burmeister A, Stephan A, Alves Avelar LA, Müller MR, Seiwert A, Höfmann S, Fischer F, Torres-Gomez H, Hoffmann MJ, Niegisch G, Bremmer F, Petzsch P, Köhrer K, Albers P, Kurz T, Skowron MA, Nettersheim D. Establishment and Evaluation of Dual HDAC/BET Inhibitors as Therapeutic Options for Germ Cell Tumors and Other Urological Malignancies. Mol Cancer Ther 2022; 21:1674-1688. [PMID: 35999659 PMCID: PMC9630828 DOI: 10.1158/1535-7163.mct-22-0207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/18/2022] [Accepted: 08/15/2022] [Indexed: 01/07/2023]
Abstract
Urological malignancies represent major challenges for clinicians, with annually rising incidences. In addition, cisplatin treatment induced long-term toxicities and the development of therapy resistance emphasize the need for novel therapeutics. In this study, we analyzed the effects of novel histone deacetylase (HDAC) and bromodomain and extraterminal domain-containing (BET) inhibitors to combine them into a potent HDAC-BET-fusion molecule and to understand their molecular mode-of-action. Treatment of (cisplatin-resistant) germ cell tumors (GCT), urothelial, renal, and prostate carcinoma cells with the HDAC, BET, and dual inhibitors decreased cell viability, induced apoptosis, and affected the cell cycle. Furthermore, a dual inhibitor considerably decreased tumor burden in GCT xenograft models. On a molecular level, correlating RNA- to ATAC-sequencing data indicated a considerable induction of gene expression, accompanied by site-specific changes of chromatin accessibility after HDAC inhibitor application. Upregulated genes could be linked to intra- and extra-cellular trafficking, cellular organization, and neuronal processes, including neuroendocrine differentiation. Regarding chromatin accessibility on a global level, an equal distribution of active or repressed DNA accessibility has been detected after HDAC inhibitor treatment, questioning the current understanding of HDAC inhibitor function. In summary, our HDAC, BET, and dual inhibitors represent a new treatment alternative for urological malignancies. Furthermore, we shed light on new molecular and epigenetic mechanisms of the tested epi-drugs, allowing for a better understanding of the underlying modes-of-action and risk assessment for the patient.
Collapse
Affiliation(s)
- Aaron Burmeister
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Alexa Stephan
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Leandro A. Alves Avelar
- Department of Pharmaceutical and Medical Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Melanie R. Müller
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andrea Seiwert
- Department of Pharmaceutical and Medical Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stefan Höfmann
- Department of Pharmaceutical and Medical Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Fabian Fischer
- Department of Pharmaceutical and Medical Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hector Torres-Gomez
- Department of Pharmaceutical and Medical Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michèle J. Hoffmann
- Department of Urology, Urological Research Laboratory, Bladder Cancer Group, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Guenter Niegisch
- Department of Urology, Urological Research Laboratory, Bladder Cancer Group, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Department of Urology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Felix Bremmer
- Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Patrick Petzsch
- Genomics and Transcriptomics Laboratory (GTL), Biological and Medical Research Center (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Karl Köhrer
- Genomics and Transcriptomics Laboratory (GTL), Biological and Medical Research Center (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Peter Albers
- Department of Urology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Thomas Kurz
- Department of Pharmaceutical and Medical Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Margaretha A. Skowron
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Corresponding Authors: Daniel Nettersheim, University Hospital Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany. Phone: 49-021-1811-5844; E-mail: ; and Margaretha A. Skowron,
| | - Daniel Nettersheim
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,Corresponding Authors: Daniel Nettersheim, University Hospital Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany. Phone: 49-021-1811-5844; E-mail: ; and Margaretha A. Skowron,
| |
Collapse
|