1
|
Alberts AR, Dinkelman-Smit M, Zuiverloon TCM, Boormans JL. Testicular Germ Cell Tumor Etiology: Still a Black Box? Eur Urol 2024:S0302-2838(24)02742-8. [PMID: 39732603 DOI: 10.1016/j.eururo.2024.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/30/2024]
Affiliation(s)
- Arnout R Alberts
- Department of Urology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Marij Dinkelman-Smit
- Department of Urology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Joost L Boormans
- Department of Urology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
2
|
Salazar-Mejía CE, Villarreal-González RV, Vidal-Gutiérrez O, de la Cruz-de la Cruz C, Guadarrama-Rendón E, Alvarado-Ruiz SA, Guerra-Garza AS, Quiroz-Huerta R, Salazar-Salazar KA, Alvarez-Villalobos NA. Immune Checkpoint Inhibitors in Patients with Testicular Cancer: A Systematic Review. J Adolesc Young Adult Oncol 2024. [PMID: 39718949 DOI: 10.1089/jayao.2024.0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024] Open
Abstract
Germ cell tumors (GCTs) are chemosensitive neoplasms with high cure rates; however, a small group of patients present tumors with refractory chemotherapy, with a dismal prognosis and few effective management options. Although immune checkpoint inhibitors (ICIs) are approved for use in chemotherapy refractory GCT, the evidence supporting this indication remains scarce. Original research studies were included on patients with GCTs refractory to chemotherapy treated with ICI up to December 2023. Comprehensive search strategies databases and MeSH keywords were used to locate eligible literature. Study characteristics, participant demographics, and oncological outcomes were recorded. A total of 13 studies (n = 106) were included, five single-patient case reports, one retrospective cohort, six-phase II randomized controlled trials (RCTs), and an abstract from the preliminary results of a phase II RCT. Most of the studies evaluated did not request biomarkers as inclusion criteria. Median overall response rate across studies was 3.4% (range, 0-57) and 0% (range, 0-6) in retrospective cohort and phase II studies. Progressive disease as the best response was present in most patients, with 75% (range, 0-82.9) in the overall population and 82% (range, 75 -83) in the retrospective cohort and phase II trials. Some of the most durable clinical responses documented in this systematic review corresponded to high tumor mutational burden (TMB-H) or high microsatellite instability (MSI-H)/dMMR tumors. Retrospective cohorts and clinical trials evaluating ICIs for the treatment of chemo-refractory GCTs documented limited activity of these drugs as a single intervention in patients not selected by biomarkers, with a tendency to better results described in those with TMB-H or MSI-H/dMMR tumors.
Collapse
Affiliation(s)
- Carlos Eduardo Salazar-Mejía
- Oncology Service, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario "Dr. José Eleuterio González," Faculty of Medicine, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Rosalaura Virginia Villarreal-González
- Oncology Service, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario "Dr. José Eleuterio González," Faculty of Medicine, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Oscar Vidal-Gutiérrez
- Oncology Service, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario "Dr. José Eleuterio González," Faculty of Medicine, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Carlos de la Cruz-de la Cruz
- Department of Internal Medicine, Christus Muguerza Hospital Alta Especialidad, Universidad de Monterrey, Monterrey, Mexico
| | - Estefanía Guadarrama-Rendón
- Oncology Service, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario "Dr. José Eleuterio González," Faculty of Medicine, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Sofia Alejandra Alvarado-Ruiz
- Faculty of Medicine, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Andrea Sarahi Guerra-Garza
- Faculty of Medicine, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Ramiro Quiroz-Huerta
- Faculty of Medicine, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Karina Alicia Salazar-Salazar
- Faculty of Medicine, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | | |
Collapse
|
3
|
Karlberg B, Kirchgaessner R, Lee J, Peterkort M, Beckman L, Goecks J, Ellrott K. SyntheVAEiser: augmenting traditional machine learning methods with VAE-based gene expression sample generation for improved cancer subtype predictions. Genome Biol 2024; 25:309. [PMID: 39696541 DOI: 10.1186/s13059-024-03431-3] [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: 10/11/2023] [Accepted: 10/30/2024] [Indexed: 12/20/2024] Open
Abstract
The accuracy of machine learning methods is often limited by the amount of training data that is available. We proposed to improve machine learning training regimes by augmenting datasets with synthetically generated samples. We present a method for synthesizing gene expression samples and test the system's capabilities for improving the accuracy of categorical prediction of cancer subtypes. We developed SyntheVAEiser, a variational autoencoder based tool that was trained and tested on over 8000 cancer samples. We have shown that this technique can be used to augment machine learning tasks and increase performance of recognition of underrepresented cohorts.
Collapse
Affiliation(s)
- Brian Karlberg
- Biomedical Engineering, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239-3098, USA
| | - Raphael Kirchgaessner
- Biomedical Engineering, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239-3098, USA
| | - Jordan Lee
- Biomedical Engineering, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239-3098, USA
| | - Matthew Peterkort
- Biomedical Engineering, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239-3098, USA
| | - Liam Beckman
- Biomedical Engineering, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239-3098, USA
| | - Jeremy Goecks
- Biomedical Engineering, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239-3098, USA
- Department of Machine Learning, Moffitt Cancer Center, Tampa, USA
| | - Kyle Ellrott
- Biomedical Engineering, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR, 97239-3098, USA.
| |
Collapse
|
4
|
Li X, Ding R, Liu Z, Teixeira WMS, Ye J, Tian L, Li H, Guo S, Yao K, Ma Z, Liu Z. A predictive system comprising serum microRNAs and radiomics for residual retroperitoneal masses in metastatic nonseminomatous germ cell tumors. Cell Rep Med 2024; 5:101843. [PMID: 39672156 DOI: 10.1016/j.xcrm.2024.101843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/18/2024] [Accepted: 11/06/2024] [Indexed: 12/15/2024]
Abstract
Predicting the histopathology of residual retroperitoneal masses (RMMs) before post-chemotherapy retroperitoneal lymph node dissection in metastatic nonseminomatous germ cell tumors (NSGCTs) can guide individualized treatment and minimize complications. Previous single approach-based models perform poorly in validation. Herein, we introduce a machine learning model that evolves from a single-dimensional tumor diameter to incorporate high-dimensional radiomic features, with its effectiveness assessed using the macro-average area under the receiver operating characteristic curves (AUCs). In addition, we utilize more precise and specific microRNAs (miRNAs), not common clinical indicators, to construct an integrated radiomics-miRNA predictive system, achieving an AUC of 0.91 (0.80-0.99) in the prospective test set. We further develop a web-based dynamic nomogram for swift and precise calculation of the histopathological probabilities of RMMs based on radiomic scores and serum miRNA levels. The radiomics-miRNA integrated system offers a promising tool to select personalized treatments for patients with metastatic NSGCT.
Collapse
Affiliation(s)
- Xiangdong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Renjie Ding
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Zhenhua Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Wilhem M S Teixeira
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Jingwei Ye
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Li Tian
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Haojiang Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Shengjie Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Kai Yao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Zikun Ma
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| | - Zhuowei Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| |
Collapse
|
5
|
Destefanis E, Sighel D, Dalfovo D, Gilmozzi R, Broso F, Cappannini A, Bujnicki J, Romanel A, Dassi E, Quattrone A. The three YTHDF paralogs and VIRMA are strong cross-histotype tumor driver candidates among m 6A core genes. NAR Cancer 2024; 6:zcae040. [PMID: 39411658 PMCID: PMC11474903 DOI: 10.1093/narcan/zcae040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/04/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
N6-Methyladenosine (m6A) is the most abundant internal modification in mRNAs. Despite accumulating evidence for the profound impact of m6A on cancer biology, there are conflicting reports that alterations in genes encoding the m6A machinery proteins can either promote or suppress cancer, even in the same tumor type. Using data from The Cancer Genome Atlas, we performed a pan-cancer investigation of 15 m6A core factors in nearly 10000 samples from 31 tumor types to reveal underlying cross-tumor patterns. Altered expression, largely driven by copy number variations at the chromosome arm level, results in the most common mode of dysregulation of these factors. YTHDF1, YTHDF2, YTHDF3 and VIRMA are the most frequently altered factors and the only ones to be uniquely altered when tumors are grouped according to the expression pattern of the m6A factors. These genes are also the only ones with coherent, pan-cancer predictive power for progression-free survival. On the contrary, METTL3, the most intensively studied m6A factor as a cancer target, shows much lower levels of alteration and no predictive power for patient survival. Therefore, we propose the non-enzymatic YTHDF and VIRMA genes as preferred subjects to dissect the role of m6A in cancer and as priority cancer targets.
Collapse
Affiliation(s)
- Eliana Destefanis
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Denise Sighel
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Davide Dalfovo
- Laboratory of Bioinformatics and Computational Biology, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Riccardo Gilmozzi
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Francesca Broso
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Andrea Cappannini
- Laboratory of Bioinformatics and Protein Engineering, International Institute of Molecular and Cell Biology in Warsaw, PL-02-109 Warsaw, Poland
| | - Janusz M Bujnicki
- Laboratory of Bioinformatics and Protein Engineering, International Institute of Molecular and Cell Biology in Warsaw, PL-02-109 Warsaw, Poland
| | - Alessandro Romanel
- Laboratory of Bioinformatics and Computational Biology, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Erik Dassi
- Laboratory of RNA Regulatory Networks, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Alessandro Quattrone
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| |
Collapse
|
6
|
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
|
7
|
Vieira Cardoso II, Nunes Rosa M, Antunes Moreno D, Barbosa Tufi LM, Pereira Ramos L, Bourdeth Pereira LA, Silva L, Soares Galvão JM, Tosi IC, Van Helvoort Lengert A, Cavalcanti Da Cruz M, Teixeira SA, Reis RM, Lopes LF, Tomazini Pinto M. Cisplatin‑resistant germ cell tumor models: An exploration of the epithelial‑mesenchymal transition regulator SLUG. Mol Med Rep 2024; 30:228. [PMID: 39392037 PMCID: PMC11484538 DOI: 10.3892/mmr.2024.13352] [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: 02/28/2024] [Accepted: 06/27/2024] [Indexed: 10/12/2024] Open
Abstract
Germ cell tumors (GCTs) constitute diverse neoplasms arising in the gonads or extragonadal locations. Testicular GCTs (TGCTs) are the predominant solid tumors in adolescents and young men. Despite cisplatin serving as the primary therapeutic intervention for TGCTs, 10‑20% of patients with advanced disease demonstrate resistance to cisplatin‑based chemotherapy, and epithelial‑mesenchymal transition (EMT) is a potential contributor to this resistance. EMT is regulated by various factors, including the snail family transcriptional repressor 2 (SLUG) transcriptional factor, and, to the best of our knowledge, remains unexplored within TGCTs. Therefore, the present study investigated the EMT transcription factor SLUG in TGCTs. In silico analyses were performed to investigate the expression of EMT markers in TGCTs. In addition, a cisplatin‑resistant model for TGCTs was developed using the NTERA‑2 cell line, and a mouse model was also established. Subsequently, EMT was assessed both in vitro and in vivo within the cisplatin‑resistant models using quantitative PCR and western blot analyses. The results of the in silico analysis showed that the different histologies exhibited distinct expression profiles for EMT markers. Seminomas exhibited a lower expression of EMT markers, whereas embryonal carcinomas and mixed GCT demonstrated high expression. Notably, patients with lower SLUG expression had longer median progression‑free survival (46.4 months vs. 28.0 months, P=0.022). In the in vitro analysis, EMT‑associated genes [fibronectin; vimentin (VIM); actin, α2, smooth muscle; collagen type I α1; transforming growth factor‑β1; and SLUG] were upregulated in the cisplatin‑resistant NTERA‑2 (NTERA‑2R) cell line after 72 h of cisplatin treatment. Consistent with this finding, the NTERA‑2R mouse model demonstrated a significant upregulation in the expression levels of VIM and SLUG. In conclusion, the present findings suggested that SLUG may serve a crucial role in connecting EMT with the development of cisplatin resistance, and targeting SLUG may be a putative therapeutic strategy to mitigate cisplatin resistance.
Collapse
Affiliation(s)
| | - Marcela Nunes Rosa
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
| | - Daniel Antunes Moreno
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
| | | | - Lorrayne Pereira Ramos
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
| | | | - Lenilson Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
| | | | - Isabela Cristiane Tosi
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
| | | | | | | | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
- Life and Health Sciences Research Institute Medical School, University of Minho, 710057 Braga, Portugal
| | - Luiz Fernando Lopes
- Barretos Children's Cancer Hospital, Hospital de Amor, Barretos, São Paulo 14784400, Brazil
| | - Mariana Tomazini Pinto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784400, Brazil
- Barretos Children's Cancer Hospital, Hospital de Amor, Barretos, São Paulo 14784400, Brazil
| |
Collapse
|
8
|
Tateo V, Thompson ZJ, Gilbert SM, Cortessis VK, Daneshmand S, Masterson TA, Feldman DR, Pierorazio PM, Prakash G, Heidenreich A, Albers P, Necchi A, Spiess PE. Epidemiology and Risk Factors for Testicular Cancer: A Systematic Review. Eur Urol 2024:S0302-2838(24)02685-X. [PMID: 39542769 DOI: 10.1016/j.eururo.2024.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/07/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND AND OBJECTIVE Testicular germ cell tumors (TGCTs) are globally rare, although incidence significantly varies across global geographic regions and ethnicities. Recent decades have seen an unexplained increase in incidence. This review investigates the changing epidemiology of TGCT and identifies key risk factors. METHODS A systematic review following the Preferred Reporting Items for Systematic Reviews and Meta-analyses 2020 statement was conducted. After screening and risk-of-bias assessment, 53 reports on significant and updated topics on TGCT epidemiology and risk factors were included for narrative synthesis. Of these, 26 were selected for quantitative synthesis. KEY FINDINGS AND LIMITATIONS Projections suggest a continued increase in global TGCT incidence, even in populations with historically low incidence. Genetic predisposition, particularly single-nucleotide polymorphisms, accounts for approximately 44% of TGCT heritability. In utero exposure to endocrine-disrupting chemicals, cryptorchidism, infertility, high height, behavioral factors such as marijuana consumption, and environmental or occupational exposures to potentially harmful substances are associated with higher TGCT risk, with variable strength of evidence. Meta-analyses confirmed a significant association between prenatal/early-life risk factors and TGCT incidence (odds ratio 1.44). Limitations include constrained evidence quality, heterogeneity in study types, and a limited volume of data supporting each topic. CONCLUSIONS AND CLINICAL IMPLICATIONS TGCT pathogenesis is influenced by genetic predisposition and exposures during early life. The rising incidence may reflect socioeconomic changes and migration patterns, which determine variation in population exposure to risk factors. TGCT epidemiology remains controversial and requires further research and the implementation of optimal screening programs considering the rising incidence and consequent impact on global health and socioeconomic systems. ADVANCING PRACTICE What does this study add? . PATIENT SUMMARY .
Collapse
Affiliation(s)
- Valentina Tateo
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Zachary J Thompson
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Scott M Gilbert
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Victoria K Cortessis
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Siamak Daneshmand
- Department of Urology, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Timothy A Masterson
- Department of Urology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Darren R Feldman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Phillip M Pierorazio
- Division of Urology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gagan Prakash
- Department of Surgical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India; Homi Bhabha National Institute, Mumai, Maharashtra, India
| | - Axel Heidenreich
- Department of Urology, University Hospital Cologne, Cologne, Germany
| | - Peter Albers
- Department of Urology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andrea Necchi
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Philippe E Spiess
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
9
|
Bian Z, Chen B, Shi G, Yuan H, Zhou Y, Jiang B, Li L, Su H, Zhang Y. Single-cell landscape identified SERPINB9 as a key player contributing to stemness and metastasis in non-seminomas. Cell Death Dis 2024; 15:812. [PMID: 39528470 PMCID: PMC11555415 DOI: 10.1038/s41419-024-07220-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/27/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Embryonal carcinoma (EC), characterized by a high degree of stemness similar to that of embryonic stem cells, is the most malignant subtype within non-seminomatous testicular germ cell tumors (TGCTs). However, the mechanisms underlying its malignancy remain unknown. In this study, we employed single-cell RNA sequencing to analyze four non-seminoma samples. Our differential expression analysis revealed high expression of SERPINB9 in metastatic EC cells. We conducted in vitro experiments to further investigate SERPINB9's role in the progression of EC. Functionally, the knockdown of SERPINB9 in NCCIT and NTERA-2 leads to a diminished migratory capability and decreased cis-platin resistance, as demonstrated by Transwell migration assay and drug sensitivity assay. Moreover, embryoid bodies showed reduced size and lower OCT4 expression, alongside heightened expression of differentiation markers AFP, ACTA2, and CD57 in shSERPINB9 cells. In vivo, the role of SERPINB9 in maintaining cancer stemness was validated by the limiting dilution assay. Mechanistically, Bulk RNA-seq further showed downregulation of ERK1/2 signaling and WNT signaling pathways with concomitant upregulation of differentiation pathways subsequent to SERPINB9 knockdown. Additionally, the analysis indicated increased levels of cytokines linked to tertiary lymphoid structures (TLS), such as IL6, IL11, IL15, CCL2, CCL5, and CXCL13 in shSERPINB9 cells, which were further validated by ELISA. Our research indicates that SERPINB9 plays a key role in driving tumor progression by enhancing tumor stemness and suppressing TLS. This study stands as the first to elucidate the molecular signature of non-seminomas at a single-cell level, presenting a wealth of promising targets with substantial potential for informing the development of future therapeutic interventions.
Collapse
Affiliation(s)
- Zhouliang Bian
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, PR China
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, PR China
| | - Biying Chen
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, PR China
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, PR China
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Haihua Yuan
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, PR China
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, PR China
| | - Yue Zhou
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Bin Jiang
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, PR China.
| | - Long Li
- Department of Urology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.
| | - Hengchuan Su
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, PR China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China.
| | - Yanjie Zhang
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, PR China.
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, PR China.
| |
Collapse
|
10
|
Ferraresso M, Bailey S, Alonso-Crisostomo L, Ward D, Panayi C, Scurlock ZGL, Saini HK, Smith SP, Nicholson JC, Enright AJ, Scarpini CG, Coleman N, Murray MJ. Replenishing co-downregulated miR-100-5p and miR-125b-5p in malignant germ cell tumors causes growth inhibition through cell cycle disruption. Mol Oncol 2024. [PMID: 39522951 DOI: 10.1002/1878-0261.13757] [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: 11/29/2023] [Revised: 09/12/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
MicroRNAs (miRNAs) are short, nonprotein-coding RNAs, and their expression is dysregulated in malignant germ cell tumors (GCTs). Here, we investigated the causes and consequences of downregulated miR-99a-5p/miR-100-5p (functionally identical) and miR-125b-5p levels in malignant GCTs regardless of age, site, or subtype. Quantitative RT-PCR was used to assess miR-99a-5p/miR-100-5p, miR-125b-5p, and associated gene expression in malignant GCT tissues/cell lines [seminoma (Sem), yolk sac tumor (YST), embryonal carcinoma (EC)]. Cells were treated with demethylating 5-azacytidine and pyrosequencing was performed. Combination miR-100-5p/miR-125b-5p mimic replenishment was used to treat malignant GCT cells. Global messenger RNA (mRNA) targets of the replenished miRNAs were identified and Metascape used to study pathway effects. We found that expression levels of miR-99a-5p/miR-100-5p and miR-125b-5p, their respective pri-miRNAs, and associated genes from chromosomes 11 and 21 (chr11/chr21) were downregulated and highly correlated in malignant GCT cells. Treatment with 5-azacytidine caused upregulation of these miRNAs, with pyrosequencing revealing hypermethylation of their chr11/chr21 loci, likely contributing to miR-100-5p/miR-125b-5p downregulation. Combination miR-100-5p/miR-125b-5p mimic replenishment resulted in growth inhibition in Sem/YST cells, with miR-100-5p/miR-125b-5p mRNA targets enriched in downregulated genes, which were involved in cell cycle (confirmed by flow cytometry) and signaling pathways. Knockdown of the miR-100-5p/miR-125b-5p target tripartite motif containing 71 (TRIM71kd) recapitulated miR-100-5p/miR-125b-5p replenishment, with growth inhibition and cell cycle disruption of Sem/YST/EC cells. Further, replenishment led to reduced lin-28 homolog A (LIN28A) levels and concomitant increases in let-7 (MIRLET7B) tumor suppressor miRNAs, creating a sustained reversion of cell phenotype. In summary, combination miR-100-5p/miR-125b-5p mimic replenishment or TRIM71kd caused growth inhibition in malignant GCT cells via cell cycle disruption. Further studies are now warranted, including mimic treatment alongside conventional platinum-based chemotherapy.
Collapse
Affiliation(s)
| | | | | | - Dawn Ward
- Department of Pathology, University of Cambridge, UK
| | | | | | - Harpreet K Saini
- EMBL-European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | | | - James C Nicholson
- Department of Paediatrics, University of Cambridge, Cambridge University Hospitals NHS Foundation Trust, UK
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, UK
| | | | | | - Nicholas Coleman
- Department of Pathology, University of Cambridge, UK
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, UK
| | - Matthew J Murray
- Department of Pathology, University of Cambridge, UK
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, UK
| |
Collapse
|
11
|
Morris JP, Baslan T, Soltis DE, Soltis PS, Fox DT. Integrating the Study of Polyploidy Across Organisms, Tissues, and Disease. Annu Rev Genet 2024; 58:297-318. [PMID: 39227132 PMCID: PMC11590481 DOI: 10.1146/annurev-genet-111523-102124] [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] [Indexed: 09/05/2024]
Abstract
Polyploidy is a cellular state containing more than two complete chromosome sets. It has largely been studied as a discrete phenomenon in either organismal, tissue, or disease contexts. Increasingly, however, investigation of polyploidy across disciplines is coalescing around common principles. For example, the recent Polyploidy Across the Tree of Life meeting considered the contribution of polyploidy both in organismal evolution over millions of years and in tumorigenesis across much shorter timescales. Here, we build on this newfound integration with a unified discussion of polyploidy in organisms, cells, and disease. We highlight how common polyploidy is at multiple biological scales, thus eliminating the outdated mindset of its specialization. Additionally, we discuss rules that are likely common to all instances of polyploidy. With increasing appreciation that polyploidy is pervasive in nature and displays fascinating commonalities across diverse contexts, inquiry related to this important topic is rapidly becoming unified.
Collapse
Affiliation(s)
- John P Morris
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA;
| | - Timour Baslan
- Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Systems Pharmacology and Translational Therapeutics and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical Sciences and Penn Vet Cancer Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Douglas E Soltis
- Florida Museum of Natural History, University of Florida, Gainesville, Florida, USA;
- Biodiversity Institute, University of Florida, Gainesville, Florida, USA
- Polyploidy Integration and Innovation Institute
- Department of Biology, University of Florida, Gainesville, Florida, USA;
| | - Pamela S Soltis
- Biodiversity Institute, University of Florida, Gainesville, Florida, USA
- Florida Museum of Natural History, University of Florida, Gainesville, Florida, USA;
- Polyploidy Integration and Innovation Institute
| | - Donald T Fox
- Department of Pharmacology and Cancer Biology, Duke Regeneration Center, and Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, USA;
- Polyploidy Integration and Innovation Institute
| |
Collapse
|
12
|
Oliver TRW, Behjati S. Developmental Dysregulation of Childhood Cancer. Cold Spring Harb Perspect Med 2024; 14:a041580. [PMID: 38692740 PMCID: PMC11529852 DOI: 10.1101/cshperspect.a041580] [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: 05/03/2024]
Abstract
Most childhood cancers possess distinct clinicopathological profiles from those seen in adulthood, reflecting their divergent mechanisms of carcinogenesis. Rather than depending on the decades-long, stepwise accumulation of changes within a mature cell that defines adult carcinomas, many pediatric malignancies emerge rapidly as the consequence of random errors during development. These errors-whether they be genetic, epigenetic, or microenvironmental-characteristically block maturation, resulting in phenotypically primitive neoplasms. Only an event that falls within a narrow set of spatiotemporal parameters will forge a malignant clone; if it occurs too soon then the event might be lethal, or negatively selected against, while if it is too late or in an incorrectly primed precursor cell then the necessary intracellular conditions for transformation will not be met. The precise characterization of these changes, through the study of normal tissues and tumors from patients and model systems, will be essential if we are to develop new strategies to diagnose, treat, and perhaps even prevent childhood cancer.
Collapse
Affiliation(s)
- Thomas R W Oliver
- Department of Histopathology and Cytology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, Cambridgeshire CB2 0QQ, United Kingdom
- Wellcome Sanger Institute, Hinxton, Cambridgeshire CB10 1RQ, United Kingdom
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, Cambridgeshire CB10 1RQ, United Kingdom
- Department of Paediatrics, University of Cambridge, Cambridge, Cambridgeshire CB2 0QQ, United Kingdom
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, Cambridgeshire CB2 0QQ, United Kingdom
| |
Collapse
|
13
|
Shokry D, Khan MW, Powell C, Johnson S, Rennels BC, Boyd RI, Sun Z, Fazal Z, Freemantle SJ, Parker MH, Vieson MD, Samuelson JP, Spinella MJ, Singh R. Refractory testicular germ cell tumors are highly sensitive to the targeting of polycomb pathway demethylases KDM6A and KDM6B. Cell Commun Signal 2024; 22:528. [PMID: 39482699 PMCID: PMC11529429 DOI: 10.1186/s12964-024-01912-3] [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: 08/27/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024] Open
Abstract
Testicular germ cell tumors (TGCTs) can be treated with cisplatin-based therapy. However, a clinically significant number of cisplatin-resistant patients die from progressive disease as no effective alternatives exist. Curative cisplatin therapy results in acute and life-long toxicities in the young TGCT patient population providing a rationale to decrease cisplatin exposure. In contrast to genetic alterations, recent evidence suggests that epigenetics is a major driving factor for TGCT formation, progression, and response to chemotherapy. Hence, targeting epigenetic pathways with "epidrugs" is one potential relatively unexplored strategy to advance TGCT treatment beyond cisplatin. In this report, we demonstrate for the first time that targeting polycomb demethylases KDM6A and KDM6B with epidrug GSK-J4 can treat both cisplatin-sensitive and -resistant TGCTs. While GSK-J4 had minimal effects alone on TGCT tumor growth in vivo, it dramatically sensitized cisplatin-sensitive and -resistant TGCTs to cisplatin. We validated KDM6A/KDM6B as the target of GSK-J4 since KDM6A/KDM6B genetic depletion had a similar effect to GSK-J4 on cisplatin-mediated anti-tumor activity and transcriptome alterations. Pharmacologic and genetic targeting of KDM6A/KDM6B potentiated or primed the p53-dominant transcriptional response to cisplatin, with also evidence for basal activation of p53. Further, several chromatin modifier genes, including BRD4, lysine demethylases, chromodomain helicase DNA binding proteins, and lysine methyltransferases, were repressed with cisplatin only in KDM6A/KDM6B-targeted cells, implying that KDM6A/KDM6B inhibition sets the stage for extensive chromatin remodeling of TGCT cells upon cisplatin treatment. Our findings demonstrate that targeting polycomb demethylases is a new potent pharmacologic strategy for treating cisplatin resistant TGCTs that warrants clinical development.
Collapse
Affiliation(s)
- Doha Shokry
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- Department of Anatomy and Embryology, Alexandria University, Alexandria, Egypt
| | - Mehwish W Khan
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Christine Powell
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Samantha Johnson
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Brayden C Rennels
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Raya I Boyd
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Zhengyang Sun
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Zeeshan Fazal
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Sarah J Freemantle
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
| | - Maryanna H Parker
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Miranda D Vieson
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Veterinary Clinical Medicine, University of Illinois Urbana-Champaign, Urbana, IL, 61802, USA
| | - Jonathan P Samuelson
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Michael J Spinella
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61802, USA.
- Cancer Center of Illinois, University of Illinois Urbana-Champaign, Urbana, IL, 61802, USA.
| | - Ratnakar Singh
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA.
| |
Collapse
|
14
|
Ní Leathlobhair M, Frangou A, Kinnersley B, Cornish AJ, Chubb D, Lakatos E, Arumugam P, Gruber AJ, Law P, Tapinos A, Jakobsdottir GM, Peneva I, Sahli A, Smyth EM, Ball RY, Sylva R, Benes K, Stark D, Young RJ, Lee ATJ, Wolverson V, Houlston RS, Sosinsky A, Protheroe A, Murray MJ, Wedge DC, Verrill C. Genomic landscape of adult testicular germ cell tumours in the 100,000 Genomes Project. Nat Commun 2024; 15:9247. [PMID: 39461959 PMCID: PMC11513037 DOI: 10.1038/s41467-024-53193-6] [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: 03/16/2023] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Testicular germ cell tumours (TGCT), which comprise seminoma and non-seminoma subtypes, are the most common cancers in young men. In this study, we present a comprehensive whole genome sequencing analysis of adult TGCTs. Leveraging samples from participants recruited via the UK National Health Service and data from the Genomics England 100,000 Genomes Project, our results provide an extended description of genomic elements underlying TGCT pathogenesis. This catalogue offers a comprehensive, high-resolution map of copy number alterations, structural variation, and key global genome features, including mutational signatures and analysis of extrachromosomal DNA amplification. This study establishes correlations between genomic alterations and histological diversification, revealing divergent evolutionary trajectories among TGCT subtypes. By reconstructing the chronological order of driver events, we identify a subgroup of adult TGCTs undergoing relatively late whole genome duplication. Additionally, we present evidence that human leukocyte antigen loss is a more prevalent mechanism of immune disruption in seminomas. Collectively, our findings provide valuable insights into the developmental and immune modulatory processes implicated in TGCT pathogenesis and progression.
Collapse
Affiliation(s)
- Máire Ní Leathlobhair
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland.
| | - Anna Frangou
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ben Kinnersley
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- University College London Cancer Institute, 72 Huntley Street, London, UK
| | - Alex J Cornish
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Daniel Chubb
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Eszter Lakatos
- Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, Gothenburg, Sweden
| | | | - Andreas J Gruber
- Department of Biology, University of Konstanz, Universitaetsstrasse 10, D-78464, Konstanz, Germany
| | - Philip Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Avraam Tapinos
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK
| | - G Maria Jakobsdottir
- Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Christie Hospital, The Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Iliana Peneva
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Atef Sahli
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Evie M Smyth
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Richard Y Ball
- Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
| | - Rushan Sylva
- Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Ksenija Benes
- Department of Pathology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Dan Stark
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Robin J Young
- Weston Park Cancer Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Alexander T J Lee
- Christie Hospital, The Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | | | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | | | - Andrew Protheroe
- Department of Oncology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Matthew J Murray
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Pathology, University of Cambridge, Cambridge, UK.
| | - David C Wedge
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Division of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
- Christie Hospital, The Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| | - Clare Verrill
- NIHR Oxford Biomedical Research Centre, Oxford, UK.
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
15
|
Shokry D, Khan MW, Powell C, Johnson S, Rennels BC, Boyd RI, Sun Z, Fazal Z, Freemantle SJ, Parker MH, Vieson MD, Samuelson JP, Spinella MJ, Singh R. Refractory testicular germ cell tumors are highly sensitive to the targeting of polycomb pathway demethylases KDM6A and KDM6B. RESEARCH SQUARE 2024:rs.3.rs-4986186. [PMID: 39483904 PMCID: PMC11527238 DOI: 10.21203/rs.3.rs-4986186/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Testicular germ cell tumors (TGCTs) can be treated with cisplatin-based therapy. However, a clinically significant number of cisplatin-resistant patients die from progressive disease as no effective alternatives exist. Curative cisplatin therapy results in acute and life-long toxicities in the young TGCT patient population providing a rationale to decrease cisplatin exposure. In contrast to genetic alterations, recent evidence suggests that epigenetics is a major driving factor for TGCT formation, progression, and response to chemotherapy. Hence, targeting epigenetic pathways with "epidrugs" is one potential relatively unexplored strategy to advance TGCT treatment beyond cisplatin. In this report, we demonstrate for the first time that targeting polycomb demethylases KDM6A and KDM6B with epidrug GSK-J4 can treat both cisplatin-sensitive and -resistant TGCTs. While GSK-J4 had minimal effects alone on TGCT tumor growth in vivo, it dramatically sensitized cisplatin-sensitive and -resistant TGCTs to cisplatin. We validated KDM6A/KDM6B as the target of GSK-J4 since KDM6A/KDM6B genetic depletion had a similar effect to GSK-J4 on cisplatin-mediated anti-tumor activity and transcriptome alterations. Pharmacologic and genetic targeting of KDM6A/KDM6B potentiated or primed the p53-dominant transcriptional response to cisplatin, with also evidence for basal activation of p53. Further, several chromatin modifier genes, including BRD4, lysine demethylases, chromodomain helicase DNA binding proteins, and lysine methyltransferases, were repressed with cisplatin only in KDM6A/KDM6B-targeted cells, implying that KDM6A/KDM6B inhibition sets the stage for extensive chromatin remodeling of TGCT cells upon cisplatin treatment. Our findings demonstrate that targeting polycomb demethylases is a new potent pharmacologic strategy for treating cisplatin resistant TGCTs that warrants clinical development.
Collapse
|
16
|
Alwosaibai K, Alruwaii ZI, Mashhour M, Almsned FM, Asraf R, Alrsheedy W, Alessa A, Almohanna H, Selwi W, Azam F. Dysgerminomas: germ cell tumors exhibit high expression of PD-L1 and associated with high TILs and good prognosis. Sci Rep 2024; 14:24191. [PMID: 39406772 PMCID: PMC11480429 DOI: 10.1038/s41598-024-74192-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Ovarian germ cell tumors (OVGCTs) account for 28% of all diagnosed ovarian cancers, and malignant germ cell tumors specifically account for approximately 13% of diagnosed ovarian cancers in Saudi Arabia. Although most germ cell tumor patients have a high survival rate, patients who experience tumor recurrence have a poor prognosis and present with more aggressive and chemoresistant tumors. The use of immunotherapeutic agents such as PD-L1/PD-1 inhibitors for OVGCTs remains very limited because few studies have described the immunological characteristics of these tumors. This study is the first to investigate PD-L1 expression in ovarian germ cell tumors and explore the role of PD-L1 expression in tumor microenvironment cells and genetic alterations. A total of 34 ovarian germ cell tumors were collected from pathology archives. The collected tumor tissues included ten dysgerminomas, five yolk sac tumors, five immature teratomas, and one mature teratoma, and the remaining samples were mixed germ cell tumors. The tumors were analyzed using immunohistochemical analysis to determine PD-L1 expression, immune cell infiltration and cancer stem cell populations and their correlation with clinical outcome. Furthermore, the genetic alterations in different subtypes of germ cell tumors were correlated with PD-L1 expression and clinical outcome. Datasets for testicular germ cells (TGCTs) were retrieved from The Cancer Genome Atlas (TCGA) and analyzed using cBioPortal (cbioportal.org) and Gene Expression Profiling Interactive Analysis (GEPIA). Compared with yolk sac tumors, dysgerminomas highly express PD-L1 and are associated with high levels of tumor infiltrating lymphocytes (TILs) and stem cell markers. In addition, compared with PD-L1-negative yolk sac tissue, dysgerminomas/seminomas with high PD-L1 expression are associated with more genetic alterations and a better prognosis. Our findings will contribute to the knowledge about the potential benefits of ovarian cancer immunotherapy in specific subsets of germ cell tumor patients and the risk factors for resistance mediated by tumor microenvironment cells.
Collapse
Affiliation(s)
- Kholoud Alwosaibai
- Biomedical Research Department, Research Center, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia.
| | - Zainab Ibrahim Alruwaii
- Department of Pathology and Lab Medicine, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Miral Mashhour
- Department of Pathology and Lab Medicine, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Fahad M Almsned
- Research Center, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
- School of Systems Biology, George Mason University, Fairfax, USA
| | - Reem Asraf
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Wadha Alrsheedy
- Biomedical Research Department, Research Center, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Ahmed Alessa
- Biomedical Research Department, Research Center, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Hani Almohanna
- Research Center, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Waleed Selwi
- Department of Medical Oncology, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| | - Faisal Azam
- Department of Medical Oncology, King Fahad Specialist Hospital, Eastern Health Cluster, Dammam, Saudi Arabia
| |
Collapse
|
17
|
Pan J, Yin W, Chen Y, Wang H, Wu W, Wang S, Li D, Ma Q. Sustained Response to Anti-PD-1 Therapy in Combination with Nab-Paclitaxel in Metastatic Testicular Germ Cell Tumor Harboring the KRAS-G12V Mutation: A Case Report. Urol Int 2024:1-9. [PMID: 39362200 DOI: 10.1159/000541588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/04/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION Cisplatin-based standardized therapy has been established for metastatic testicular germ cell tumors (TGCTs). However, the patient prognosis is considerably less favorable if the disease recurs following failure of first-line therapies. There is a need for novel treatment options for patients with recurrent or metastatic TGCTs, notably for those that are not sensitive to first-line chemotherapy. With the development of next-generation sequencing technologies, an increasing number of gene mutations has been identified in TGCTs. Previously published research studies have established a link between KRAS mutations and chemotherapy resistance, and have demonstrated that KRAS mutations are associated with inflammatory tumor microenvironment and tumor immunogenicity, leading to an improved response to inhibition of programmed death (PD-1) protein expression. Previous studies have reported that the tumor immune microenvironment of TGCT influences therapeutic efficacy. CASE PRESENTATION A 65-year-old metastatic patient with TGCT and a KRAS-12 valine-for-glycine gene mutation was described. This patient initially underwent inguinal orchiectomy and received two prior chemotherapeutic regimens. Following the rapid progression of the disease, the patient was treated with anti-PD-1 therapy and nab-paclitaxel chemotherapy, and his condition was successfully controlled by this combination treatment. CONCLUSION To the best of our knowledge, this is the first successful case of KRAS-mutation patient with TGCT who achieved partially and sustained disease remission by combining immune checkpoint inhibitors with chemotherapy. This case provides an excellent example for personalized treatment of metastatic TGCTs.
Collapse
Affiliation(s)
- Jinfeng Pan
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Comprehensive Genitourinary Cancer Center, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Weiqi Yin
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yingzhi Chen
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Hui Wang
- Department of Medical Oncology, Zhejiang University Mingzhou Hospital, Ningbo, China
| | - Wei Wu
- Department of Medical Oncology, Zhejiang University Mingzhou Hospital, Ningbo, China
| | - Suying Wang
- Department of Tissue Pathology, Ningbo Clinical Pathological Diagnosis Center, Ningbo, China
| | - Da Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University Medical School, Hangzhou, China
| | - Qi Ma
- Comprehensive Genitourinary Cancer Center, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Ningbo Clinical Research Center for Urological Disease, Ningbo, China
- Translational Research Laboratory for Urology, The Key Laboratory of Ningbo City, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Yi-Huan Genitourinary Cancer Group, Ningbo, China
| |
Collapse
|
18
|
Xu L, Liddell B, Nesic K, Geissler F, Ashwood L, Wakefield M, Scott C, Waddell N, Kondrashova O. High-level tumour methylation of BRCA1 and RAD51C is required for homologous recombination deficiency in solid cancers. NAR Cancer 2024; 6:zcae033. [PMID: 39055334 PMCID: PMC11270467 DOI: 10.1093/narcan/zcae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
In ovarian and breast cancer, promoter methylation of BRCA1 or RAD51C is a promising biomarker for PARP inhibitor response, as high levels lead to homologous recombination deficiency (HRD). Yet the extent and role of such methylation in other cancers is not clear. This study comprehensively investigated promoter methylation of eight homologous recombination repair genes across 23 solid cancer types. Here, we showed that BRCA1 methylated cancers were associated with reduced gene expression, loss of heterozygosity (LOH), TP53 mutations and genomic features of HRD. We identified BRCA1 methylation in 3% of the copy-number high subtype of endometrial cancer, and as a rare event in six other cancer types, including lung squamous cell, pancreatic, bladder and stomach cancer. RAD51C promoter methylation was widespread across multiple cancer types, but HRD features were only observed for cases which contained high-level tumour methylation and LOH of RAD51C. While RAD51C methylation was frequent in stomach adenocarcinoma (6%) and low-grade glioma (2.5%), it was mostly detected at a low tumour level, suggestive of heterozygous methylation, and was associated with CpG island methylator phenotype. Our findings indicate that high-level tumour methylation of BRCA1 and RAD51C should be explored as a PARP inhibitor biomarker across multiple cancers.
Collapse
Affiliation(s)
- Lijun Xu
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- The University of Queensland, Brisbane, QLD, Australia
| | - Brett Liddell
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- The University of Queensland, Brisbane, QLD, Australia
| | - Ksenija Nesic
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Franziska Geissler
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Lauren M Ashwood
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- The University of Queensland, Brisbane, QLD, Australia
| | - Matthew J Wakefield
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia
| | - Clare L Scott
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia
| | - Nicola Waddell
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- The University of Queensland, Brisbane, QLD, Australia
| | - Olga Kondrashova
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- The University of Queensland, Brisbane, QLD, Australia
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| |
Collapse
|
19
|
Soto-Heras S, Reinacher L, Wang B, Oh JE, Bunnell M, Park CJ, Hess RA, Ko CJ. Cryptorchidism and testicular cancer in the dog: unresolved questions and challenges in translating insights from human studies†. Biol Reprod 2024; 111:269-291. [PMID: 38738783 DOI: 10.1093/biolre/ioae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024] Open
Abstract
Cryptorchidism, the failure of one or both testes to descend into the scrotum, and testicular cancer show a strong correlation in both dogs and humans. Yet, long-standing medical debates persist about whether the location of undescended testes directly causes testicular cancer in humans or if both conditions stem from a common origin. Although testicular cancer is a prevalent disease in dogs, even less is known about its cause and correlation with testicular descent in this species. This review investigates the relation between these two disorders in dogs, drawing insights from human studies, and examines key biomarkers identified thus far. In addition, it explores potential causal links, including the impact of temperature on maturing testicular cells and a potential shared genetic origin. Notably, this literature review reveals significant differences between men and dogs in reproductive development, histological and molecular features of testicular tumors, and the prevalence of specific tumor types, such as Sertoli cell tumors in cryptorchid dogs and germ cell tumors in humans. These disparities caution against using dogs as models for human testicular cancer research and underscore the limitations when drawing comparisons between species. The paper concludes by suggesting specific research initiatives to enhance our understanding of the complex interplay between cryptorchidism and testicular cancer in dogs.
Collapse
Affiliation(s)
- Sandra Soto-Heras
- Epivara, Inc., 2109 S. Oak Street, Suite 100A, Champaign, IL 61820, USA
| | - Lindsey Reinacher
- Epivara, Inc., 2109 S. Oak Street, Suite 100A, Champaign, IL 61820, USA
| | - Bensen Wang
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Ji Eun Oh
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Mary Bunnell
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Chan Jin Park
- Epivara, Inc., 2109 S. Oak Street, Suite 100A, Champaign, IL 61820, USA
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Rex A Hess
- Epivara, Inc., 2109 S. Oak Street, Suite 100A, Champaign, IL 61820, USA
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - CheMyong Jay Ko
- Epivara, Inc., 2109 S. Oak Street, Suite 100A, Champaign, IL 61820, USA
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| |
Collapse
|
20
|
Baky FJ, Matulewicz RS, Feldman DR, Hamilton RJ, Bagrodia A. MicroRNA for Prediction of Teratoma and Viable Germ Cell Tumor after Chemotherapy. Urol Clin North Am 2024; 51:387-394. [PMID: 38925741 DOI: 10.1016/j.ucl.2024.03.007] [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: 06/28/2024]
Abstract
MicroRNAs (miRNAs) are emerging as highly sensitive and specific markers for testicular germ cell tumors (GCTs) across the spectrum of disease. However, their utility in specific clinical scenarios requires further study. Here, we review the current evidence for miRNAs as tumor markers for the evaluation of treatment response in patients undergoing chemotherapy for the treatment of advanced testicular GCT.
Collapse
Affiliation(s)
- Fady J Baky
- Department of Urology, Memorial Sloan Kettering Cancer Center, 430 East 67th Street, New York, NY 10065, USA
| | - Richard S Matulewicz
- Department of Urology, Memorial Sloan Kettering Cancer Center, 430 East 67th Street, New York, NY 10065, USA
| | - Darren R Feldman
- Germ Cell Cancer, Department of Medicine, Memorial Sloan Kettering Cancer Center, 430 East 67th Street, New York, NY 10065, USA
| | - Robert J Hamilton
- Division of Urology, Princess Margaret Hospital, 3-130, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Aditya Bagrodia
- Department of Urology, UC San Diego Health, 9400 Campus Point Drive, Suite 1-200, La Jolla, CA 92037, USA.
| |
Collapse
|
21
|
Kugaevskaya EV, Timoshenko OS, Gureeva TA, Radko SP, Lisitsa AV. MicroRNAs as promising diagnostic and prognostic markers for the human genitourinary cancer. BIOMEDITSINSKAIA KHIMIIA 2024; 70:191-205. [PMID: 39239894 DOI: 10.18097/pbmc20247004191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Genitourinary cancer (GUC) represents more than one fifth of all human cancers. This makes the development of approaches to its early diagnosis an important task of modern biomedicine. Circulating microRNAs, short (17-25 nucleotides) non-coding RNA molecules found in human biological fluids and performing a regulatory role in the cell, are considered as promising diagnostic and prognostic biomarkers of cancers, including GUC. In this review we have considered the current state of research aimed at assessing microRNAs as biomarkers of such human GUC types as malignant tumors of the bladder, kidney, prostate, testicles, ovaries, and cervix. A special attention has been paid to studies devoted to the identification of microRNAs in urine as a surrogate "liquid biopsy" that may provide the simplest and cheapest approach to mass non-invasive screening of human GUC. The use of microRNA panels instead of single types of microRNA generally leads to higher sensitivity and specificity of the developed diagnostic tests. However, to date, work on the microRNAs assessment as biomarkers of human GUC is still of a research nature, and the further introduction of diagnostic tests based on microRNAs into practice requires successful clinical trials.
Collapse
Affiliation(s)
| | | | - T A Gureeva
- Institute of Biomedical Chemistry, Moscow, Russia
| | - S P Radko
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A V Lisitsa
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
22
|
Puljak L, Bojcic R, Cikes Botic M, Odak Z, Riva N, Gold VR, Tabone Y, Squizzato A, Calleja-Agius J. Trials of interventions for ovarian and testicular germ cell tumors registered in ClinicalTrials.gov: A cross-sectional study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024:108545. [PMID: 39068044 DOI: 10.1016/j.ejso.2024.108545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION There is unmet need in the treatment of ovarian and testicular germ cell tumors (GCTs). This study analyzed registered trials of interventions for GCTs. MATERIALS AND METHODS We included trials of interventions for GCTs registered on ClinicalTrials.gov by July 29, 2022. We analyzed their interventions, outcome measures and study design. RESULTS We included 142 trials registrations; 42 (30 %) for ovarian GCTs, 50 (35 %) for testicular GCTs, and 50 (35 %) trials for both. The majority of the trials were completed (56 %) and did not have results available (75 %). Most trials were in Phase 2. Information about the study design were not reported for many analyzed trials. Most trials had a single-group assignment (44 %) and were open-label (68 %). The median planned number of enrolled participants was 43. Most registrations used medicine(s) (87 %), either as a single type of intervention or in combination. The most commonly used type of medicine was chemotherapy (54 %). Primary outcome was not reported in 23 % of registrations, and secondary outcomes were not reported in 35 % of registrations. Overall survival was used in 6 % of registrations as a primary outcome and in 31 % of registrations as a secondary outcome. CONCLUSIONS Few trials on GCTs were registered on ClinicalTrials.gov, and their number was declining in recent times. Most registrations did not report relevant information about the study design, or results if completed. More effort is needed to foster trials on GCTs, as well as to optimize the management of the registrations and foster the publication of research results.
Collapse
Affiliation(s)
- Livia Puljak
- Center for Evidence-Based Medicine and Health Care, Catholic University of Croatia, Zagreb, Croatia.
| | | | - Monika Cikes Botic
- Department of Gynecology and Obstetrics, University Hospital of Split, Split, Croatia.
| | - Zdravko Odak
- Department of Gynecology and Obstetrics, University Hospital of Split, Split, Croatia.
| | - Nicoletta Riva
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, Malta.
| | - Vienna R Gold
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, Msida, Malta.
| | - Ylenia Tabone
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, Msida, Malta.
| | | | - Jean Calleja-Agius
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, Msida, Malta.
| |
Collapse
|
23
|
Tan J, Wu J, Yuan R, Li W, Li L, Huang H, Lu Y. A case report of recurrent testicular germ cell tumor in a patient with a history of primary pulmonary germ cell tumor and a review of the literature. Front Oncol 2024; 14:1361380. [PMID: 39045558 PMCID: PMC11263088 DOI: 10.3389/fonc.2024.1361380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/07/2024] [Indexed: 07/25/2024] Open
Abstract
Background Compared to testicular germ cell tumors, the incidence of extragonadal germ cell tumors (EGCTs) is relatively low. While the lungs are a common site for metastasis of malignant germ cell tumors, primary pulmonary germ cell tumors are extremely rare. Objective To enhance the understanding of the diagnosis and treatment of germ cell tumors, particularly extragonadal germ cell tumors (EGCTs). Methods A Case Report of Recurrent Testicular Germ Cell Tumor in a Patient with Primary Pulmonary Germ Cell Tumor and a Review of the Literature. Clinical data The patient was initially diagnosed with primary pulmonary germ cell tumor and received standard treatment. Five years later, the patient developed a recurrent testicular germ cell tumor. The pathological results from the two surgeries were different, indicating embryonal carcinoma in the first instance and seminoma in the second. Conclusion For cases with a high suspicion of extragonadal germ cell tumors (EGCTs), early pathological biopsy is essential to confirm the histological subtype and to guide the selection of the most appropriate and sensitive treatment regimen.
Collapse
Affiliation(s)
- Jian Tan
- Department of Urology, Zhongshan People's Hospital, Zhongshan, China
- First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Jinfeng Wu
- Department of Urology, Zhongshan People's Hospital, Zhongshan, China
- First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Runqiang Yuan
- First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Wei Li
- First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Linfeng Li
- Department of Urology, Zhongshan People's Hospital, Zhongshan, China
| | - Hongxing Huang
- Department of Urology, Zhongshan People's Hospital, Zhongshan, China
- First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Yangbai Lu
- Department of Urology, Zhongshan People's Hospital, Zhongshan, China
| |
Collapse
|
24
|
Surucu A, de Biase D, Ricci C, di Sciascio L, Collins K, Idrees MT, Ebare K, Fiorentino M, Bridge JA, Ulbright TM, Acosta AM. Beta-Catenin Alterations in Postchemotherapy Yolk Sac Tumor, Postpubertal-Type With Enteroblastic Features. Mod Pathol 2024; 37:100513. [PMID: 38763421 DOI: 10.1016/j.modpat.2024.100513] [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: 01/30/2024] [Revised: 04/05/2024] [Accepted: 05/11/2024] [Indexed: 05/21/2024]
Abstract
Postchemotherapy postpubertal-type yolk sac tumors (YST) with glandular and solid phenotypes are aggressive and commonly resistant to systemic chemotherapy. These neoplasms show morphologic features that significantly overlap with those of somatic carcinomas with "enteroblastic" or "fetal" phenotype (the preferred terminology depends on the site of origin). They often present as late or very late recurrences, and their diagnosis is challenging because they frequently affect patients in an age group at risk for carcinomas of somatic origin. Recently, we incidentally identified examples of postchemotherapy glandular and solid YST with "enteroblastic" phenotypes and nuclear expression of beta-catenin, prompting us to further evaluate the prevalence of this phenomenon. We found nuclear expression of beta-catenin in 10 (29%) of 34 such tumors. A subset of cases with nuclear beta-catenin expression was further analyzed with a DNA sequencing panel (n = 6) and fluorescence in situ hybridization for isochromosome 12p [i(12p); n = 5]. Sequencing identified exon 3 CTNNB1 variants in 3 (50%) of 6 analyzed cases, and fluorescence in situ hybridization was positive for i(12p) in 5 of 5 cases. In conclusion, a significant subset of postchemotherapy YST with glandular or solid architecture and "enteroblastic" phenotype demonstrates beta-catenin alterations, suggesting that activation of Wnt signaling may play a role in the progression of these neoplasms. Moreover, nuclear beta-catenin expression in these tumors represents a potential diagnostic pitfall given that carcinomas of true somatic origin with overlapping morphology may also be positive for this marker.
Collapse
Affiliation(s)
- Ahmet Surucu
- Department of Pathology, Indiana University, Indianapolis, Indiana
| | - Dario de Biase
- Department of Pathology, Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna, Italy
| | - Costantino Ricci
- DIAP-Dipartimento InterAziendale di Anatomia Patologica di Bologna, Maggiore Hospital-AUSL Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Luisa di Sciascio
- DIAP-Dipartimento InterAziendale di Anatomia Patologica di Bologna, Maggiore Hospital-AUSL Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Katrina Collins
- Department of Pathology, Indiana University, Indianapolis, Indiana
| | | | - Kingsley Ebare
- Department of Pathology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Michelangelo Fiorentino
- DIAP-Dipartimento InterAziendale di Anatomia Patologica di Bologna, Maggiore Hospital-AUSL Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Julia A Bridge
- Department of Pathology, University of Nebraska Medical Center, Omaha, Nebraska; Cytogenetics and Fluorescence In Situ Hybridization, ProPath Laboratories, Dallas, Texas
| | | | - Andres M Acosta
- Department of Pathology, Indiana University, Indianapolis, Indiana.
| |
Collapse
|
25
|
Cheng K, Seita Y, Whelan EC, Yokomizo R, Hwang YS, Rotolo A, Krantz ID, Ginsberg JP, Kolon TF, Lal P, Luo X, Pierorazio PM, Linn RL, Ryeom S, Sasaki K. Defining the cellular origin of seminoma by transcriptional and epigenetic mapping to the normal human germline. Cell Rep 2024; 43:114323. [PMID: 38861385 DOI: 10.1016/j.celrep.2024.114323] [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: 03/11/2024] [Revised: 04/26/2024] [Accepted: 05/21/2024] [Indexed: 06/13/2024] Open
Abstract
Aberrant male germline development can lead to the formation of seminoma, a testicular germ cell tumor. Seminomas are biologically similar to primordial germ cells (PGCs) and many bear an isochromosome 12p [i(12p)] with two additional copies of the short arm of chromosome 12. By mapping seminoma transcriptomes and open chromatin landscape onto a normal human male germline trajectory, we find that seminoma resembles premigratory/migratory PGCs; however, it exhibits enhanced germline and pluripotency programs and upregulation of genes involved in apoptosis, angiogenesis, and MAPK/ERK pathways. Using pluripotent stem cell-derived PGCs from Pallister-Killian syndrome patients mosaic for i(12p), we model seminoma and identify gene dosage effects that may contribute to transformation. As murine seminoma models do not exist, our analyses provide critical insights into genetic, cellular, and signaling programs driving seminoma transformation, and the in vitro platform developed herein permits evaluation of additional signals required for seminoma tumorigenesis.
Collapse
Affiliation(s)
- Keren Cheng
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Yasunari Seita
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Eoin C Whelan
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Ryo Yokomizo
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Young Sun Hwang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Antonia Rotolo
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Ian D Krantz
- Division of Human Genetics, The Roberts Individualized Medical Genetics Center, The Children's Hospital of Philadelphia, 3500 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Jill P Ginsberg
- Department of Pediatrics, The Children's Hospital of Philadelphia, 3500 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Thomas F Kolon
- Division of Urology, The Children's Hospital of Philadelphia, 3500 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Priti Lal
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Xunda Luo
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Presbyterian Medical Center, 51 North 39th Street, Philadelphia, PA 19104, USA
| | - Phillip M Pierorazio
- Division of Urology, University of Pennsylvania Presbyterian Medical Center, 3737 Market St. 4th Floor, Philadelphia, PA 19104, USA
| | - Rebecca L Linn
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, Philadelphia, PA 19104, USA; Department of Pathology, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Sandra Ryeom
- Department of Surgery, Columbia University Irving Medical Center, 630 W. 168th Street, P&S 17-409, New York, NY 10032, USA
| | - Kotaro Sasaki
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
26
|
Islam R, Heyer J, Figura M, Wang X, Nie X, Nathaniel B, Indumathy S, Hartmann K, Pleuger C, Fijak M, Kliesch S, Dittmar F, Pilatz A, Wagenlehner F, Hedger M, Loveland B, Hotaling JH, Guo J, Loveland KL, Schuppe HC, Fietz D. T cells in testicular germ cell tumors: new evidence of fundamental contributions by rare subsets. Br J Cancer 2024; 130:1893-1903. [PMID: 38649788 PMCID: PMC11183042 DOI: 10.1038/s41416-024-02669-9] [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: 10/03/2023] [Accepted: 03/21/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Immune cell infiltration is heterogeneous but common in testicular germ cell tumors (TGCT) and pre-invasive germ cell neoplasia in situ (GCNIS). Tumor-infiltrating T cells including regulatory T (Treg) and follicular helper T (Tfh) cells are found in other cancer entities, but their contributions to TGCT are unknown. METHODS Human testis specimens from independent patient cohorts were analyzed using immunohistochemistry, flow cytometry and single-cell RNA sequencing (scRNA-seq) with special emphasis on delineating T cell subtypes. RESULTS Profound changes in immune cell composition within TGCT, shifting from macrophages in normal testes to T cells plus B and dendritic cells in TGCT, were documented. In most samples (96%), the CD4+ T cell frequency exceeded that of CD8+ cells, with decreasing numbers from central to peripheral tumor areas, and to tumor-free, contralateral testes. T cells including Treg and Tfh were most abundant in seminoma compared to mixed tumors and embryonal carcinoma. CONCLUSION Despite considerable heterogeneity between patients, T cell subtypes form a key part of the TGCT microenvironment. The novel finding of rare Treg and Tfh cells in human testis suggests their involvement in TGCT pathobiology, with implications for understanding tumor progression, to assess patients' prognosis, and as putative targets for personalized immunotherapy.
Collapse
Affiliation(s)
- Rashidul Islam
- Dept. of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Jannis Heyer
- Dept. of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
- Dept. of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
| | - Miriam Figura
- Dept. of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
- Dept. of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
| | - Xiaoyan Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
| | - Xichen Nie
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Benedict Nathaniel
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Sivanjah Indumathy
- Dept. of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Katja Hartmann
- Dept. of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany
| | - Christiane Pleuger
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University, Giessen, Germany
- Institute of Anatomy and Cell Biology, Justus Liebig University, Giessen, Germany
| | - Monika Fijak
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University, Giessen, Germany
- Institute of Anatomy and Cell Biology, Justus Liebig University, Giessen, Germany
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, University of Muenster, Muenster, Germany
| | - Florian Dittmar
- Dept. of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
| | - Adrian Pilatz
- Dept. of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University, Giessen, Germany
| | - Florian Wagenlehner
- Dept. of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University, Giessen, Germany
| | - Mark Hedger
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | | | - James H Hotaling
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Jingtao Guo
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kate L Loveland
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Hans-Christian Schuppe
- Dept. of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University, Giessen, Germany
| | - Daniela Fietz
- Dept. of Veterinary Anatomy, Histology and Embryology, Justus Liebig University, Giessen, Germany.
- Hessian Centre of Reproductive Medicine, Justus-Liebig-University, Giessen, Germany.
| |
Collapse
|
27
|
Lobo J, Tavares NT, Jerónimo C, Henrique R, Dvindenko E, Cornejo KM, Berney DM, Ulbright TM, Gupta S, Acosta AM. Analysis of MicroRNA-371-373 supports that a subset of spermatocytic tumors demonstrates biologic features similar to those of GCNIS-derived germ cell tumors. Hum Pathol 2024; 148:66-71. [PMID: 38782099 DOI: 10.1016/j.humpath.2024.05.005] [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: 04/03/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Spermatocytic tumors are rare testicular tumors occurring predominantly in older men. Most show a classical tripartite morphology (different from seminoma) and are benign. However, well-documented cases of malignant spermatocytic tumors exist. Our previous work showed that a subset of spermatocytic tumors exhibiting TP53 mutations, DNA methylation profiles closer to seminomas, and/or gains in chromosome 12p exhibited aggressive characteristics, including sarcomatoid transformation and metastatic dissemination. The microRNA-371-373 cluster is a promising biomarker which is upregulated in non-teratoma germ cell tumors with malignant behavior. In this work we analyze microRNAs-371-373 b y quantitative real-time polymerase chain reaction in 18 spermatocytic tumors representative of the whole clinical spectrum, including 6 with aggressive features (sarcomatoid transformation, metastases, or gains in chromosome 12p). The levels of microRNAs-371-373 were significantly higher in non-teratoma germ cell tumors compared to spermatocytic tumors, overall (p < 0.0001). Importantly, levels of microRNA-371-373 were higher in spermatocytic tumors with aggressive features compared to non-aggressive neoplasms. The highest levels were observed in one tumor showing isochromosome 12p. These results further support our previous findings that a subset of spermatocytic tumors are intermediate between so-called type II and type III germ cell tumors and that embryonic microRNAs play a role in aggressive behavior in spermatocytic tumors. Accordingly, this subset of tumors may behave aggressively and require close follow up. In the future, this opens an opportunity for microRNA testing in serum of spermatocytic tumor patients for risk stratification purposes.
Collapse
Affiliation(s)
- João Lobo
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (P.CCC) & RISE@CI-IPOP (Health Research Network), R. Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Department of Pathology and Molecular Immunology, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-513, Porto, Portugal
| | - Nuno Tiago Tavares
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (P.CCC) & RISE@CI-IPOP (Health Research Network), R. Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Doctoral Programme in Biomedical Sciences, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-513, Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (P.CCC) & RISE@CI-IPOP (Health Research Network), R. Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Department of Pathology and Molecular Immunology, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-513, Porto, Portugal
| | - Rui Henrique
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (P.CCC) & RISE@CI-IPOP (Health Research Network), R. Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal; Department of Pathology and Molecular Immunology, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-513, Porto, Portugal
| | - Eugenia Dvindenko
- Department of Pathology, Instituto Português de Oncologia de Lisboa Francisco Gentil, EPE, Lisbon, Portugal
| | - Kristine M Cornejo
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel M Berney
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Sounak Gupta
- Department of Pathology, Mayo Clinic, Rochester, MN, USA
| | - Andres M Acosta
- Department of Pathology Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
28
|
Labropoulou VT, Manou D, Ravazoula P, Alzahrani FM, Kalofonos HP, Theocharis AD. Expression of CD44 is associated with aggressiveness in seminomas. Mol Biol Rep 2024; 51:693. [PMID: 38796656 PMCID: PMC11127849 DOI: 10.1007/s11033-024-09638-8] [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: 11/27/2023] [Accepted: 05/13/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Testicular germ cell tumors (TGCTs) exhibit diverse biological and pathological features and are divided in two main types, seminomas and nonseminomatous germ cell tumors (NSGCTs). CD44 is a cell surface receptor, which is highly expressed in malignancies and is implicated in tumorigenesis affecting cell-matrix interactions and cell signaling. METHODS AND RESULTS Here, we examined the expression of CD44 in tumor cell lines and in patients' material. We found that CD44 is over-expressed in TGCTs compared to normal tissues. Immunohistochemical staining in 71 tissue specimens demonstrated increased expression of CD44 in some patients, whereas CD44 was absent in normal tissue. In seminomas, a high percentage of tumor and stromal cells showed cytoplasmic and/or cell surface staining for CD44 as well as increased staining for CD44 in the tumor stroma was found in some cases. The increased expression of CD44 either in tumor cells or in stromal components was associated with tumor size, nodal metastasis, vascular/lymphatic invasion, and disease stage only in seminomas. The increased stromal expression of CD44 in TGCTs was positively associated with angiogenesis. CONCLUSIONS CD44 may exhibit diverse biological functions in seminomas and NSGCTs. The expression of CD44 in tumor cells as well as in tumor stroma fosters an aggressive phenotype in seminomas and should be considered in disease treatment.
Collapse
Affiliation(s)
- Vasiliki T Labropoulou
- Department of Internal Medicine, Division of Hematology, University of Patras Medical School, Patras, Greece.
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Panagiota Ravazoula
- Department of Pathology, University Hospital of Patras, Patras, 26504, Greece
| | - Fatimah Mohammed Alzahrani
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Haralabos P Kalofonos
- Clinical Oncology Laboratory, Division of Oncology, Department of Medicine, University of Patras, Rio, 26504, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia.
| |
Collapse
|
29
|
Ding R, Li X, Liu Z. Overview of Novel Biomarkers for Management of Postchemotherapy Residual Masses in Testicular Cancer. Eur Urol Focus 2024; 10:380-382. [PMID: 38724341 DOI: 10.1016/j.euf.2024.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 09/01/2024]
Abstract
The diagnosis of postchemotherapy residual masses in testicular cancer must be based on the integration of clinical, imaging, and serology tests. Further validation is needed for novel biomarkers.
Collapse
Affiliation(s)
- Renjie Ding
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiangdong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhuowei Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
30
|
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
|
31
|
Rajpert-De Meyts E, Goriely A, Almstrup K. New analysis of atypical spermatocytic tumours reveals extensive heterogeneity and plasticity of germ cell tumours †. J Pathol 2024; 263:1-4. [PMID: 38362619 DOI: 10.1002/path.6262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024]
Abstract
Testicular germ cell tumours (TGCTs) derived from immature (type I) and pluripotent germ cell neoplasia in situ (GCNIS, type II) are characterised by remarkable phenotypic heterogeneity and plasticity. In contrast, the rare spermatocytic tumour (SpT, type III), derived from mature spermatogonia, is considered a homogenous and benign tumour but may occasionally present as an anaplastic or an aggressive sarcomatoid tumour. While various oncogenic processes had been proposed, the precise mechanism driving malignant progression remained elusive until the molecular characterisation of a series of atypical SpTs described in a recent issue of The Journal of Pathology. The emerging picture suggests the presence of two distinct trajectories for SpTs, involving either RAS/mitogen-activated protein kinase pathway mutations or a ploidy shift with secondary TP53 mutations and/or gain of chromosome 12p, the latter known as pathognomonic for type II GCNIS-derived TGCTs. Here, we discuss the implications of these findings, seen from the perspective of germ cell biology and the unique features of different TGCTs. The evolving phenotype of SpTs, induced by genomic and epigenetic changes, illustrates that the concept of plasticity applies to all germ cell tumours, making them inherently heterogenous and capable of significant transformation during progression. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Ewa Rajpert-De Meyts
- Department of Growth & Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Anne Goriely
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Kristian Almstrup
- Department of Growth & Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
32
|
Lee SM, Loo CE, Prasasya RD, Bartolomei MS, Kohli RM, Zhou W. Low-input and single-cell methods for Infinium DNA methylation BeadChips. Nucleic Acids Res 2024; 52:e38. [PMID: 38407446 PMCID: PMC11040145 DOI: 10.1093/nar/gkae127] [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: 09/11/2023] [Revised: 01/29/2024] [Accepted: 02/10/2024] [Indexed: 02/27/2024] Open
Abstract
The Infinium BeadChip is the most widely used DNA methylome assay technology for population-scale epigenome profiling. However, the standard workflow requires over 200 ng of input DNA, hindering its application to small cell-number samples, such as primordial germ cells. We developed experimental and analysis workflows to extend this technology to suboptimal input DNA conditions, including ultra-low input down to single cells. DNA preamplification significantly enhanced detection rates to over 50% in five-cell samples and ∼25% in single cells. Enzymatic conversion also substantially improved data quality. Computationally, we developed a method to model the background signal's influence on the DNA methylation level readings. The modified detection P-value calculation achieved higher sensitivities for low-input datasets and was validated in over 100 000 public diverse methylome profiles. We employed the optimized workflow to query the demethylation dynamics in mouse primordial germ cells available at low cell numbers. Our data revealed nuanced chromatin states, sex disparities, and the role of DNA methylation in transposable element regulation during germ cell development. Collectively, we present comprehensive experimental and computational solutions to extend this widely used methylation assay technology to applications with limited DNA.
Collapse
Affiliation(s)
- Sol Moe Lee
- Center for Computational and Genomic Medicine, The Children's Hospital of Philadelphia, PA 19104, USA
| | - Christian E Loo
- Graduate Group in Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rexxi D Prasasya
- Department of Cell and Developmental Biology, Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Marisa S Bartolomei
- Department of Cell and Developmental Biology, Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Rahul M Kohli
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wanding Zhou
- Center for Computational and Genomic Medicine, The Children's Hospital of Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
33
|
Clarke DJB, Marino GB, Deng EZ, Xie Z, Evangelista JE, Ma'ayan A. Rummagene: massive mining of gene sets from supporting materials of biomedical research publications. Commun Biol 2024; 7:482. [PMID: 38643247 PMCID: PMC11032387 DOI: 10.1038/s42003-024-06177-7] [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: 10/11/2023] [Accepted: 04/10/2024] [Indexed: 04/22/2024] Open
Abstract
Many biomedical research publications contain gene sets in their supporting tables, and these sets are currently not available for search and reuse. By crawling PubMed Central, the Rummagene server provides access to hundreds of thousands of such mammalian gene sets. So far, we scanned 5,448,589 articles to find 121,237 articles that contain 642,389 gene sets. These sets are served for enrichment analysis, free text, and table title search. Investigating statistical patterns within the Rummagene database, we demonstrate that Rummagene can be used for transcription factor and kinase enrichment analyses, and for gene function predictions. By combining gene set similarity with abstract similarity, Rummagene can find surprising relationships between biological processes, concepts, and named entities. Overall, Rummagene brings to surface the ability to search a massive collection of published biomedical datasets that are currently buried and inaccessible. The Rummagene web application is available at https://rummagene.com .
Collapse
Affiliation(s)
- Daniel J B Clarke
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Giacomo B Marino
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eden Z Deng
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Zhuorui Xie
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - John Erol Evangelista
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
34
|
Michaelis J, Himmelsbach R, Metzger P, Lassmann S, Börries M, Werner M, Miething C, Höfflin R, Illert AL, Duyster J, Becker H, Sigle A, Gratzke C, Grabbert M. Primary Results of Patients with Genitourinary Malignancies Presented at a Molecular Tumor Board. Urol Int 2024; 108:383-391. [PMID: 38626735 DOI: 10.1159/000538908] [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: 03/03/2024] [Accepted: 04/11/2024] [Indexed: 04/18/2024]
Abstract
INTRODUCTION Personalized medicine poses great opportunities and challenges. While the therapeutic landscape markedly expands, descriptions about status, clinical implementation and real-world benefits of precision oncology and molecular tumor boards (MTB) remain sparse, particularly in the field of genitourinary (GU) cancer. Hence, this study characterized urological MTB cases to better understand the potential role of MTB in uro-oncology. METHODS We analyzed patients with complete data sets being reviewed at an MTB from January 2019 to October 2022, focusing on results of molecular analysis and treatment recommendations. RESULTS We evaluated 102 patients with GU cancer with a mean patient age of 61.7 years. Prostate cancer (PCa) was the most frequent entity with 52.9% (54/102), followed by bladder cancer (18.6%, 19/102) and renal cell carcinoma (14.7%, 15/102). On average, case presentation at MTB took place 54.9 months after initial diagnosis and after 2.7 previous lines of therapy. During the study period, 49.0% (50/102) of patients deceased. Additional MTB-based treatment recommendations were achieved in a majority of 68.6% (70/102) of patients, with a recommendation for targeted therapy in 64.3% (45/70) of these patients. Only 6.7% (3/45) of patients - due to different reasons - received the recommended MTB-based therapy though, with 33% (1/3) of patients reaching disease control. Throughout the MTB study period, GU cancer case presentations and treatment recommendations increased, while the time interval between initial presentation and final therapy recommendation were decreasing over time. CONCLUSION Presentation of uro-oncological patients at the MTB is a highly valuable measure for clinical decision-making. Prospectively, earlier presentation of patients at the MTB and changing legislative issues regarding comprehensive molecular testing and targeted treatment approval might further improve patients' benefits from comprehensive molecular diagnostics.
Collapse
Affiliation(s)
- Jakob Michaelis
- Department of Urology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ruth Himmelsbach
- Department of Urology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Patrick Metzger
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Silke Lassmann
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Surgical Pathology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Börries
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Werner
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Surgical Pathology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cornelius Miething
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rouven Höfflin
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna L Illert
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Justus Duyster
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Heiko Becker
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - August Sigle
- Department of Urology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Gratzke
- Department of Urology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Markus Grabbert
- Department of Urology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Molecular Tumorboard Network (MTB) Freiburg, Freiburg, Germany AND German Cancer Consortium (DKTK), Partner Site Freiburg of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
35
|
Zhou W, Johnson BK, Morrison J, Beddows I, Eapen J, Katsman E, Semwal A, Habib W, Heo L, Laird P, Berman B, Triche T, Shen H. BISCUIT: an efficient, standards-compliant tool suite for simultaneous genetic and epigenetic inference in bulk and single-cell studies. Nucleic Acids Res 2024; 52:e32. [PMID: 38412294 PMCID: PMC11014253 DOI: 10.1093/nar/gkae097] [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: 05/22/2023] [Revised: 01/23/2024] [Accepted: 02/08/2024] [Indexed: 02/29/2024] Open
Abstract
Data from both bulk and single-cell whole-genome DNA methylation experiments are under-utilized in many ways. This is attributable to inefficient mapping of methylation sequencing reads, routinely discarded genetic information, and neglected read-level epigenetic and genetic linkage information. We introduce the BISulfite-seq Command line User Interface Toolkit (BISCUIT) and its companion R/Bioconductor package, biscuiteer, for simultaneous extraction of genetic and epigenetic information from bulk and single-cell DNA methylation sequencing. BISCUIT's performance, flexibility and standards-compliant output allow large, complex experimental designs to be characterized on clinical timescales. BISCUIT is particularly suited for processing data from single-cell DNA methylation assays, with its excellent scalability, efficiency, and ability to greatly enhance mappability, a key challenge for single-cell studies. We also introduce the epiBED format for single-molecule analysis of coupled epigenetic and genetic information, facilitating the study of cellular and tissue heterogeneity from DNA methylation sequencing.
Collapse
Affiliation(s)
- Wanding Zhou
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Benjamin K Johnson
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Jacob Morrison
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Ian Beddows
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - James Eapen
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Efrat Katsman
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Ayush Semwal
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Walid Abi Habib
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Lyong Heo
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Peter W Laird
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Benjamin P Berman
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Timothy J Triche
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Hui Shen
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
36
|
Hashemi Gheinani A, Kim J, You S, Adam RM. Bioinformatics in urology - molecular characterization of pathophysiology and response to treatment. Nat Rev Urol 2024; 21:214-242. [PMID: 37604982 DOI: 10.1038/s41585-023-00805-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2023] [Indexed: 08/23/2023]
Abstract
The application of bioinformatics has revolutionized the practice of medicine in the past 20 years. From early studies that uncovered subtypes of cancer to broad efforts spearheaded by the Cancer Genome Atlas initiative, the use of bioinformatics strategies to analyse high-dimensional data has provided unprecedented insights into the molecular basis of disease. In addition to the identification of disease subtypes - which enables risk stratification - informatics analysis has facilitated the identification of novel risk factors and drivers of disease, biomarkers of progression and treatment response, as well as possibilities for drug repurposing or repositioning; moreover, bioinformatics has guided research towards precision and personalized medicine. Implementation of specific computational approaches such as artificial intelligence, machine learning and molecular subtyping has yet to become widespread in urology clinical practice for reasons of cost, disruption of clinical workflow and need for prospective validation of informatics approaches in independent patient cohorts. Solving these challenges might accelerate routine integration of bioinformatics into clinical settings.
Collapse
Affiliation(s)
- Ali Hashemi Gheinani
- Department of Urology, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Urology, Inselspital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Jina Kim
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sungyong You
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rosalyn M Adam
- Department of Urology, Boston Children's Hospital, Boston, MA, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
37
|
Pyle LC, Kim J, Bradfield J, Damrauer SM, D'Andrea K, Einhorn LH, Godse R, Hakonarson H, Kanetsky PA, Kember RL, Jacobs LA, Maxwell KN, Rader DJ, Vaughn DJ, Weathers B, Wubbenhorst B, Regeneron Genetics Center Research Team, Cancer Genomics Research Laboratory, Greene MH, Nathanson KL, Stewart DR. Germline Exome Sequencing for Men with Testicular Germ Cell Tumor Reveals Coding Defects in Chromosomal Segregation and Protein-targeting Genes. Eur Urol 2024; 85:337-345. [PMID: 37246069 PMCID: PMC10676450 DOI: 10.1016/j.eururo.2023.05.008] [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: 11/30/2022] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 05/30/2023]
Abstract
BACKGROUND Testicular germ cell tumor (TGCT) is the most common cancer among young White men. TGCT is highly heritable, although there are no known high-penetrance predisposition genes. CHEK2 is associated with moderate TGCT risk. OBJECTIVE To identify coding genomic variants associated with predisposition to TGCT. DESIGN, SETTING, AND PARTICIPANTS The study involved 293 men with familial or bilateral (high risk; HR)-TGCT representing 228 unique families and 3157 cancer-free controls. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS We carried out exome sequencing and gene burden analysis to identify associations with TGCT risk. RESULTS AND LIMITATIONS Gene burden association identified several genes, including loss-of-function variants of NIN and QRSL1. We identified no statistically significant association with the sex- and germ-cell development pathways (hypergeometric overlap test: p = 0.65 for truncating variants, p = 0.47 for all variants) or evidence of associations with the regions previously identified via genome-wide association studies (GWAS). When considering all significant coding variants together with genes associated with TGCT on GWAS, there were associations with three major pathways: mitosis/cell cycle (Gene Ontology identity GO:1903047: observed/expected variant ratio [O/E] 6.17, false discovery rate [FDR] 1.53 × 10-11), co-translational protein targeting (GO:0006613: O/E 18.62, FDR 1.35 × 10-10), and sex differentiation (GO:0007548: O/E 5.25, FDR 1.90 × 10-4). CONCLUSIONS To the best of our knowledge, this study is the largest to date on men with HR-TGCT. As in previous studies, we identified associations with variants for several genes, suggesting multigenic heritability. We identified associations with co-translational protein targeting, and chromosomal segregation and sex determination, identified via GWAS. Our results suggest potentially druggable targets for TGCT prevention or treatment. PATIENT SUMMARY We searched for gene variations that increase the risk of testicular cancer and found numerous new specific variants that contribute to this risk. Our results support the idea that many gene variants inherited together contribute to the risk of testicular cancer.
Collapse
Affiliation(s)
- Louise C Pyle
- Rare Disease Institute, Center for Genetic Medicine, Children's National Hospital, Washington, DC, USA; Department of Precision Medicine, George Washington University, Washington, DC, USA; Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jung Kim
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | | | - Scott M Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kurt D'Andrea
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Rama Godse
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hakon Hakonarson
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Rachel L Kember
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Linda A Jacobs
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kara N Maxwell
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel J Rader
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David J Vaughn
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benita Weathers
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bradley Wubbenhorst
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Mark H Greene
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Katherine L Nathanson
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas R Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
| |
Collapse
|
38
|
Ran L, Liu Y, Jiang L. Iodine‑125 seeds combined with carboplatin in the treatment of retroperitoneal metastatic seminoma: A case report and literature review. Oncol Lett 2024; 27:156. [PMID: 38426154 PMCID: PMC10902756 DOI: 10.3892/ol.2024.14289] [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: 11/28/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Testicular seminoma is a relatively rare malignant tumor, with the most common site of recurrence and metastasis being the retroperitoneal lymph nodes. Since seminoma is highly sensitive to radiotherapy and chemotherapy, even if it metastasizes, its cure rate is still >95%. However, the long-term toxicity and side effects of radiotherapy and chemotherapy cannot be ignored. Iodine-125 seeds represent a low-energy radioactive source that kills tumor cells while protecting the surrounding normal tissues, and brachytherapy using iodine-125 seeds has been widely used for the treatment of various malignancies. In addition, carboplatin can be used as an alternative to cisplatin-based combination chemotherapy to reduce the incidence of pulmonary toxicity, neurological damage and renal toxicity. In the present study, a case in which iodine-125 seeds were implanted for the treatment of retroperitoneal metastatic seminoma is reported. The patient was diagnosed with postoperative recurrence of seminoma that metastasized to the retroperitoneal lymph nodes. Since the tumor was large and surrounded blood vessels, surgical intervention and external radiotherapy were not considered. Moreover, considering the potential long-term toxic side effects of standard chemotherapy, a treatment plan for the patient using iodine-125 seed implantation combined with carboplatin (AUC7) therapy was finally formulated. No disease recurrence or toxic reactions occurred during the 3-year follow-up after treatment. The present case therefore demonstrated the antitumor efficacy and reduced toxicity of iodine-125 seeds combined with carboplatin for treating seminoma.
Collapse
Affiliation(s)
- Linhao Ran
- Department of Radiology, People's Hospital of Chongqing Banan District, Chongqing 401320, P.R. China
| | - Ying Liu
- Department of Radiology, People's Hospital of Chongqing Banan District, Chongqing 401320, P.R. China
| | - Li Jiang
- Department of Ultrasound, People's Hospital of Chongqing Banan District, Chongqing 401320, P.R. China
| |
Collapse
|
39
|
Seales CL, Puri D, Yodkhunnatham N, Pandit K, Yuen K, Murray S, Smitham J, Lafin JT, Bagrodia A. Advancing GCT Management: A Review of miR-371a-3p and Other miRNAs in Comparison to Traditional Serum Tumor Markers. Cancers (Basel) 2024; 16:1379. [PMID: 38611057 PMCID: PMC11010994 DOI: 10.3390/cancers16071379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 03/29/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
MicroRNAs, short non-protein coding RNAs, are overexpressed in GCTs. Circulating levels of germ cell tumor (GCT)-associated miRNAs, such as miR-371a-3p, can be utilized as efficient and cost-effective alternatives in diagnosing and managing patients presenting with GCTs. This quality of miRNAs has demonstrated favorable performance characteristics as a reliable blood-based biomarker with high diagnostic accuracy compared to current serum tumor markers (STMs), including α-fetoprotein (AFP), beta human chorionic gonadotropin (β-hCG), and lactate dehydrogenase (LDH). The conventional STMs exhibit limited specificity and sensitivity. Potential clinical implications of miRNAs include impact on de-escalating or intensifying treatment, detecting recurrence at earlier stages, and lessening the necessity of cross-sectional imaging or invasive tissue biopsy for non-teratomatous GCTs. Here, we also highlight the outstanding issues that must be addressed prior to clinical implementation. Standards for measuring circulating miRNAs and determining ideal cutoff values are essential for integration into current clinical guidelines.
Collapse
Affiliation(s)
- Crystal L. Seales
- Morehouse School of Medicine, Atlanta, GA 30310, USA;
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (D.P.); (N.Y.); (K.P.); (K.Y.); (J.S.)
| | - Dhruv Puri
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (D.P.); (N.Y.); (K.P.); (K.Y.); (J.S.)
| | - Nuphat Yodkhunnatham
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (D.P.); (N.Y.); (K.P.); (K.Y.); (J.S.)
| | - Kshitij Pandit
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (D.P.); (N.Y.); (K.P.); (K.Y.); (J.S.)
| | - Kit Yuen
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (D.P.); (N.Y.); (K.P.); (K.Y.); (J.S.)
| | - Sarah Murray
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA;
| | - Jane Smitham
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (D.P.); (N.Y.); (K.P.); (K.Y.); (J.S.)
| | - John T. Lafin
- Department of Urology, University Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Aditya Bagrodia
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (D.P.); (N.Y.); (K.P.); (K.Y.); (J.S.)
- Department of Urology, University Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| |
Collapse
|
40
|
Satake D, Natsumeda M, Satomi K, Tada M, Sato T, Okubo N, Kawabe K, Takahashi H, Tsukamoto Y, Okada M, Sano M, Iwabuchi H, Shibata N, Imamura M, Imai C, Takami H, Ichimura K, Nishikawa R, Umezu H, Kakita A, Oishi M. Successful Multimodal Treatment of Intracranial Growing Teratoma Syndrome with Malignant Features. Curr Oncol 2024; 31:1831-1838. [PMID: 38668041 PMCID: PMC11049495 DOI: 10.3390/curroncol31040138] [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: 02/16/2024] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Molecular analysis of the growing teratoma syndrome has not been extensively studied. Here, we report a 14-year-old boy with a growing mass during treatment for a mixed germ cell tumor of the pineal region. Tumor markers were negative; thus, growing teratoma syndrome was suspected. A radical resection via the occipital transtentorial approach was performed, and histopathological examination revealed a teratoma with malignant features. Methylation classifier analysis confirmed the diagnosis of teratoma, and DMRT1 loss and 12p gain were identified by copy number variation analysis, potentially elucidating the cause of growth and malignant transformation of the teratoma. The patient remains in remission after intense chemoradiation treatment as a high-risk germ cell tumor.
Collapse
Affiliation(s)
- Daiken Satake
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (D.S.); (T.S.); (N.O.); (K.K.); (H.T.); (Y.T.); (M.O.); (M.S.); (M.O.)
| | - Manabu Natsumeda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (D.S.); (T.S.); (N.O.); (K.K.); (H.T.); (Y.T.); (M.O.); (M.S.); (M.O.)
- Advanced Treatment of Neurological Diseases Branch, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Kaishi Satomi
- Department of Pathology, Kyorin University Faculty of Medicine, Tokyo 181-8611, Japan;
| | - Mari Tada
- Department of Pathology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.T.); (A.K.)
| | - Taro Sato
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (D.S.); (T.S.); (N.O.); (K.K.); (H.T.); (Y.T.); (M.O.); (M.S.); (M.O.)
| | - Noritaka Okubo
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (D.S.); (T.S.); (N.O.); (K.K.); (H.T.); (Y.T.); (M.O.); (M.S.); (M.O.)
| | - Keita Kawabe
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (D.S.); (T.S.); (N.O.); (K.K.); (H.T.); (Y.T.); (M.O.); (M.S.); (M.O.)
| | - Haruhiko Takahashi
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (D.S.); (T.S.); (N.O.); (K.K.); (H.T.); (Y.T.); (M.O.); (M.S.); (M.O.)
| | - Yoshihiro Tsukamoto
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (D.S.); (T.S.); (N.O.); (K.K.); (H.T.); (Y.T.); (M.O.); (M.S.); (M.O.)
| | - Masayasu Okada
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (D.S.); (T.S.); (N.O.); (K.K.); (H.T.); (Y.T.); (M.O.); (M.S.); (M.O.)
| | - Masakazu Sano
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (D.S.); (T.S.); (N.O.); (K.K.); (H.T.); (Y.T.); (M.O.); (M.S.); (M.O.)
| | - Haruko Iwabuchi
- Department of Pediatrics, Niigata University Medical and Dental Hospital, Niigata 951-8520, Japan; (H.I.); (N.S.); (C.I.)
| | - Nao Shibata
- Department of Pediatrics, Niigata University Medical and Dental Hospital, Niigata 951-8520, Japan; (H.I.); (N.S.); (C.I.)
| | - Masaru Imamura
- Department of Pediatrics, Niigata University Medical and Dental Hospital, Niigata 951-8520, Japan; (H.I.); (N.S.); (C.I.)
| | - Chihaya Imai
- Department of Pediatrics, Niigata University Medical and Dental Hospital, Niigata 951-8520, Japan; (H.I.); (N.S.); (C.I.)
- Department of Pediatrics, Toyama University, Toyama 930-0194, Japan
| | - Hirokazu Takami
- Department of Neurosurgery, The University of Tokyo Hospital, Tokyo 113-8655, Japan;
| | - Koichi Ichimura
- Department of Brain Disease Translational Research, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan;
| | - Ryo Nishikawa
- Department of Neurosurgery/Neuro-Oncology, Saitama Medical University International Medical Center, Saitama 350-1298, Japan;
| | - Hajime Umezu
- Division of Pathology, Niigata University Medical and Dental Hospital, Niigata University, Niigata 951-8520, Japan;
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (M.T.); (A.K.)
| | - Makoto Oishi
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; (D.S.); (T.S.); (N.O.); (K.K.); (H.T.); (Y.T.); (M.O.); (M.S.); (M.O.)
| |
Collapse
|
41
|
McHugh DJ, Gleeson JP, Feldman DR. Testicular cancer in 2023: Current status and recent progress. CA Cancer J Clin 2024; 74:167-186. [PMID: 37947355 DOI: 10.3322/caac.21819] [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: 07/20/2023] [Revised: 09/25/2023] [Accepted: 09/29/2023] [Indexed: 11/12/2023] Open
Abstract
Testicular germ cell tumor (GCT) is the most common solid tumor in adolescent and young adult men. Progress in the management of GCT has been made in the last 50 years, with a substantial improvement in cure rates for advanced disease, from 25% in the 1970s to nearly 80%. However, relapsed or platinum-refractory disease occurs in a proportion, 20% of whom will die from disease progression. This article reviews the current evidence-based treatments for extracranial GCT, the acute and chronic toxic effects that may result, and highlights contemporary advances and progress in the field.
Collapse
Affiliation(s)
- Deaglan J McHugh
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medicine, New York, New York, USA
| | - Jack P Gleeson
- Cancer Research, College of Medicine and Health, University College Cork, Cork, Ireland
- Medical Oncology Department, Cork University Hospital, Cork, Ireland
| | - Darren R Feldman
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
42
|
Yodkhunnatham N, Pandit K, Puri D, Yuen KL, Bagrodia A. MicroRNAs in Testicular Germ Cell Tumors: The Teratoma Challenge. Int J Mol Sci 2024; 25:2156. [PMID: 38396829 PMCID: PMC10889716 DOI: 10.3390/ijms25042156] [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: 01/03/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Testicular germ cell tumors (TGCTs) are relatively common in young men, making accurate diagnosis and prognosis assessment essential. MicroRNAs (miRNAs), including microRNA-371a-3p (miR-371a-3p), have shown promise as biomarkers for TGCTs. This review discusses the recent advancements in the use of miRNA biomarkers in TGCTs, with a focus on the challenges surrounding the noninvasive detection of teratomas. Circulating miR-371a-3p, which is expressed in undifferentiated TGCTs but not in teratomas, is a promising biomarker for TGCTs. Its detection in serum, plasma, and, potentially, cystic fluid could be useful for TGCT diagnosis, surveillance, and monitoring of therapeutic response. Other miRNAs, such as miR-375-3p and miR-375-5p, have been investigated to differentiate between TGCT subtypes (teratoma, necrosis/fibrosis, and viable tumors), which can aid in treatment decisions. However, a reliable marker for teratoma has yet to be identified. The clinical applications of miRNA biomarkers could spare patients from unnecessary surgeries and allow for more personalized therapeutic approaches. Particularly in patients with residual masses larger than 1 cm following chemotherapy, it is critical to differentiate between viable tumors, teratomas, and necrosis/fibrosis. Teratomas, which mimic somatic tissues, present a challenge in differentiation and require a comprehensive diagnostic approach. The combination of miR-371 and miR-375 shows potential in enhancing diagnostic precision, aiding in distinguishing between teratomas, viable tumors, and necrosis. The implementation of miRNA biomarkers in TGCT care could improve patient outcomes, reduce overtreatment, and facilitate personalized therapeutic strategies. However, a reliable marker for teratoma is still lacking. Future research should focus on the clinical validation and standardization of these biomarkers to fully realize their potential.
Collapse
Affiliation(s)
- Nuphat Yodkhunnatham
- Department of Urology, University of California San Diego School of Medicine, La Jolla, CA 92093, USA; (N.Y.); (K.P.); (D.P.); (K.L.Y.)
| | - Kshitij Pandit
- Department of Urology, University of California San Diego School of Medicine, La Jolla, CA 92093, USA; (N.Y.); (K.P.); (D.P.); (K.L.Y.)
| | - Dhruv Puri
- Department of Urology, University of California San Diego School of Medicine, La Jolla, CA 92093, USA; (N.Y.); (K.P.); (D.P.); (K.L.Y.)
| | - Kit L. Yuen
- Department of Urology, University of California San Diego School of Medicine, La Jolla, CA 92093, USA; (N.Y.); (K.P.); (D.P.); (K.L.Y.)
| | - Aditya Bagrodia
- Department of Urology, University of California San Diego School of Medicine, La Jolla, CA 92093, USA; (N.Y.); (K.P.); (D.P.); (K.L.Y.)
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
43
|
Saliyeva S, Boranbayeva R, Bulegenova M, Beloussov V. Application of microRNAs in the diagnosis and monitoring of pediatric germ cell tumors: Kazakh experience. Pediatr Hematol Oncol 2024; 41:121-134. [PMID: 37898912 DOI: 10.1080/08880018.2023.2267607] [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: 02/27/2023] [Accepted: 10/02/2023] [Indexed: 10/31/2023]
Abstract
GCT is characterized by specific biochemical markers expression, such as human chorionic gonadotropin (hCG) and alpha-fetoprotein (AFP), which are the main tools in the diagnosis and monitoring of GCT treatment. They are expressed in 15-20% of cases of seminoma and in 60-80% of cases of non-seminoma. MicroRNA profiling allows to identify a number of microRNAs that are superior to classical serum tumor markers in the diagnosis of primary tumors, as well as in subsequent monitoring and prediction of recurrence. We analyzed the expression of 9 microRNAs (microRNA clusters 302/367 and 371-373, microRNA375) in the blood serum of 20 children with extracranial GCT at different stages of therapy and showed their usefulness and informativeness in early detection of events. Taking into consideration the high sensitivity and specificity, serum microRNAs 367,371,372,373,302d are of great interest for clinical use in malignant GCT. Significant expression of miR 375-3p was not detected either in malignant GCT or in teratomas.
Collapse
Affiliation(s)
- Symbat Saliyeva
- Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
- Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Riza Boranbayeva
- Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
- Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Minira Bulegenova
- Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
- Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | | |
Collapse
|
44
|
Du Y, Liu L, Zou B, Chen Z, Chen Q, Deng R, Yang P. Prognostic Differences Between Surveillance and Active Treatment After Initial Orchiectomy in Patients With Stage I Mixed Germ Cell Tumors of the Testis: A Propensity Score Matching Analysis. J Surg Res 2024; 294:26-36. [PMID: 37857140 DOI: 10.1016/j.jss.2023.09.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023]
Abstract
INTRODUCTION The prognosis and optimal treatment approach for stage I mixed germ cell cancers of the testis are not well-established. This study aimed to assess contemporary treatment rates and their correlation with the cancer-specific mortality (CSM) and other-cause mortality (OCM) in patients with stage I testicular mixed germ cell tumors (TMGCT) who underwent orchiectomy, comparing surveillance with active treatment, including chemotherapy (CHT) and retroperitoneal lymph node dissection (RPLND). METHODS Retrospective analysis of clinical data from stage I TMGCT patients who underwent orchiectomy was conducted using the Surveillance, Epidemiology, and End Results database from 2004 to 2019. The annual percentage change (APC) in the use of surveillance, postoperative CHT, and RPLND was examined. Propensity score matching (PSM) and cumulative incidence, analyses were employed to compare differences in CSM and OCM between surveillance and active treatment, as well as between CHT and RPLND. Multivariate competing-risks regression models were utilized to investigate independent factors affecting CSM and OCM among stage I TMGCT patients. RESULTS The study included 5743 individuals with stage I TMGCT that underwent surveillance (61.6%), CHT(27.2%), or RPLND (11.2%). Among them, 82 deaths were attributed to TMGCT, and 82 deaths resulted from other causes. Surveillance rates increased over time (APC: 0.635%, P = 0.008), as did CHT rates (APC: 0.863%, P < 0.001), while RPLND rates declined (APC: -0.96%, P < 0.001). After PSM, multivariate competing-risks regression analysis showed that, active treatment, compared to surveillance, was not an independent factor for CSM and OCM. In contrast, when compared to CHT, RPLND was an independent factor associated with lower CSM (hazard ratio = 0.247, 95% confidence interval: 0.08-0.761; P = 0.015), but not OCM (hazard ratio = 0.946, 95% confidence interval: 0.377-2.37; P = 0.91). CONCLUSIONS Surveillance and CHT rates have increased over time for patients with stage I TMGCT following initial orchiectomy, while RPLND utilization has decreased. There was no significant difference in CSM between surveillance and active treatment groups, but RPLND demonstrated significantly lower CSM than CHT in active treatment. Our findings suggest that the usage of RPLND in patients with stage I TMGCT should be reconsidered.
Collapse
Affiliation(s)
- Yong Du
- Department of Pediatric Surgery, Suining Central Hospital, Suining, Sichuan Province, China
| | - Lianghua Liu
- Department of Pathology, Biological Specimen Laboratory, Suining Central Hospital, Suining, Sichuan Province, China
| | - Bing Zou
- Department of Pediatric Surgery, Suining Central Hospital, Suining, Sichuan Province, China
| | - Zhili Chen
- Department of Pediatric Surgery, Suining Central Hospital, Suining, Sichuan Province, China
| | - Qiang Chen
- Department of Pediatric Surgery, Suining Central Hospital, Suining, Sichuan Province, China
| | - Rui Deng
- Department of Pediatric Surgery, Suining Central Hospital, Suining, Sichuan Province, China
| | - Ping Yang
- Department of Pediatric Surgery, Suining Central Hospital, Suining, Sichuan Province, China.
| |
Collapse
|
45
|
Satomi K, Ichimura K, Shibahara J. Decoding the DNA methylome of central nervous system tumors: An emerging modality for integrated diagnosis. Pathol Int 2024; 74:51-67. [PMID: 38224248 DOI: 10.1111/pin.13402] [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: 11/06/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 01/16/2024]
Abstract
The definitive diagnosis and classification of individual cancers are crucial for patient care and cancer research. To achieve a robust diagnosis of central nervous system (CNS) tumors, a genotype-phenotype integrated diagnostic approach was introduced in recent versions of the World Health Organization classification, followed by the incorporation of a genome-wide DNA methylome-based classification. Microarray-based platforms are widely used to obtain DNA methylome data, and the German Cancer Research Center (Deutsches Krebsforschungszentrum [DKFZ]) has a webtool for a DNA methylation-based classifier (DKFZ classifier). Integration of DNA methylome will further enhance the precision of CNS tumor classification, especially in diagnostically challenging cases. However, in the clinical application of DNA methylome-based classification, challenges related to data interpretation persist, in addition to technical caveats, regulations, and limited accessibility. Dimensionality reduction (DMR) can complement integrated diagnosis by visualizing a profile and comparing it with other known samples. Therefore, DNA methylome-based classification is a highly useful research tool for auxiliary analysis in challenging diagnostic and rare disease cases, and for establishing novel tumor concepts. Decoding the DNA methylome, especially by DMR in addition to DKFZ classifier, emphasizes the capability of grasping the fundamental biological principles that provide new perspectives on CNS tumors.
Collapse
Affiliation(s)
- Kaishi Satomi
- Department of Pathology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Koichi Ichimura
- Department of Brain Disease Translational Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Junji Shibahara
- Department of Pathology, Kyorin University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
46
|
Cao J, Liu Z, Yuan J, Luo Y, Wang J, Liu J, Bo H, Guo J. Subgrouping testicular germ cell tumors based on immunotherapy and chemotherapy associated lncRNAs. Heliyon 2024; 10:e24320. [PMID: 38298718 PMCID: PMC10827771 DOI: 10.1016/j.heliyon.2024.e24320] [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: 05/08/2023] [Revised: 12/01/2023] [Accepted: 01/07/2024] [Indexed: 02/02/2024] Open
Abstract
Testicular germ cell tumors (TGCT) are the most common reproductive system malignancies in men aged 15-44 years, accounting for 95 % of all testicular tumors. Our previous studies have been shown that long non-coding RNAs (lncRNAs), such as LINC00313, TTTY14 and RFPL3S, were associated with development of TGCT. Subgrouping TGCT according to differential expressed lncRNAs and immunological characteristics is helpful to comprehensively describe the characteristics of TGCT and implement precise treatment. In this study, the TGCT transcriptome data in The Cancer Genome Atlas Program (TCGA) database was used to perform consensus clustering analysis to construct a prognostic model for TGCT. TGCT was divided into 3 subtypes C1, C2, and C3 based on the differentially expressed lncRNAs. C1 subtype was sensitive to chemotherapy drugs, while the C2 subtype was not sensitive to chemotherapy drugs, and C3 subtype may benefit from immunotherapy. We defined the C1 subtype as epidermal progression subtype, the C2 subtype as mesenchymal progression subtype, and the C3 subtype as T cell activation subtype. Subgrouping based on differentially expressed genes (DEGs) and immunological characteristics is helpful for the precise treatment of TGCT.
Collapse
Affiliation(s)
- Jian Cao
- Hunan Cancer Hospital, Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine of Central South University, Changsha, 410013, Hunan, China
| | - Zhizhong Liu
- Hunan Cancer Hospital, Department of Urology, The Affiliated Cancer Hospital of Xiangya School of Medicine of Central South University, Changsha, 410013, Hunan, China
| | - Junbin Yuan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Yanwei Luo
- Department of Blood Transfusion, the Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Jinrong Wang
- Department of Urology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jianye Liu
- Department of Urology, The Third Xiangya Hospital of Central South University, No.138, Tongzipo Road, Changsha, 410013, Hunan, China
| | - Hao Bo
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, Hunan, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410078, Hunan, China
| | - Jie Guo
- National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, Changsha, Hunan, China
- China National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- International Science and Technology Innovation Cooperation Base for Early Clinical Trials of Biological Agents in Hunan Province, Changsha, Hunan, China
| |
Collapse
|
47
|
Alonso-Crisostomo L, Trendell J, Ferraresso M, Bailey S, Ward D, Scurlock ZGL, Wenlock SC, Bastos CAP, Jugdaohsingh R, Faria NJ, Enright AJ, Scarpini CG, Coleman N, Murray MJ. Testicular germ cell tumour cells release microRNA-containing extracellular vesicles that induce phenotypic and genotypic changes in cells of the tumour microenvironment. Int J Cancer 2024; 154:372-388. [PMID: 37632231 DOI: 10.1002/ijc.34697] [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: 01/02/2023] [Revised: 06/29/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023]
Abstract
Malignant germ-cell-tumours (GCTs) are characterised by microRNA (miRNA/miR-) dysregulation, with universal over-expression of miR-371~373 and miR-302/367 clusters regardless of patient age, tumour site, or subtype (seminoma/yolk-sac-tumour/embryonal carcinoma). These miRNAs are released into the bloodstream, presumed within extracellular-vesicles (EVs) and represent promising biomarkers. Here, we comprehensively examined the role of EVs, and their miRNA cargo, on (fibroblast/endothelial/macrophage) cells representative of the testicular GCT (TGCT) tumour microenvironment (TME). Small RNA next-generation-sequencing was performed on 34 samples, comprising representative malignant GCT cell lines/EVs and controls (testis fibroblast [Hs1.Tes] cell-line/EVs and testis/ovary samples). TME cells received TGCT co-culture, TGCT-derived EVs, and a miRNA overexpression system (miR-371a-OE) to assess functional relevance. TGCT cells secreted EVs into culture media. MiR-371~373 and miR-302/367 cluster miRNAs were overexpressed in all TGCT cells/subtypes compared with control cells and were highly abundant in TGCT-derived EVs, with miR-371a-3p/miR-371a-5p the most abundant. TGCT co-culture resulted in increased levels of miRNAs from the miR-371~373 and miR-302/367 clusters in TME (fibroblast) cells. Next, fluorescent labelling demonstrated TGCT-derived EVs were internalised by all TME (fibroblast/endothelial/macrophage) cells. TME (fibroblast/endothelial) cell treatment with EVs derived from different TGCT subtypes resulted in increased miR-371~373 and miR-302/367 miRNA levels, and other generic (eg, miR-205-5p/miR-148-3p) and subtype-specific (seminoma, eg, miR-203a-3p; yolk-sac-tumour, eg, miR-375-3p) miRNAs. MiR-371a-OE in TME cells resulted in increased collagen contraction (fibroblasts) and angiogenesis (endothelial cells), via direct mRNA downregulation and alteration of relevant pathways. TGCT cells communicate with nontumour stromal TME cells through release of EVs enriched in oncogenic miRNAs, potentially contributing to tumour progression.
Collapse
Affiliation(s)
| | | | | | - Shivani Bailey
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Dawn Ward
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | | | - Carlos A P Bastos
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Ravin Jugdaohsingh
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Nuno J Faria
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Anton J Enright
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | - Nicholas Coleman
- Department of Pathology, University of Cambridge, Cambridge, UK
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Matthew J Murray
- Department of Pathology, University of Cambridge, Cambridge, UK
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
48
|
Tavares NT, Lobo J, Bagrodia A. MicroRNAs for detecting occult genitourinary cancer. Curr Opin Urol 2024; 34:20-26. [PMID: 37916954 DOI: 10.1097/mou.0000000000001137] [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: 11/03/2023]
Abstract
PURPOSE OF REVIEW Genitourinary (GU) malignancies are a real burden in global health worldwide. Each model has its own clinical challenges, and the early screening and/or detection of occult cancer in follow-up is transversal to all of them. MicroRNAs (miRNAs) have been proposed as minimally invasive liquid biopsy cancer biomarkers, due to their stability and low degradation. RECENT FINDINGS The different GU tumor models are in different stages concerning miRNAs as biomarkers for cancer detection. Testicular germ cell tumors (TGCTs) already have a specific defined target, miR-371a-3p, that has shown high sensitivity and specificity in different clinical settings, and is now in final stages of preanalytical testing before entering the clinic. The other GU malignancies are in a different stage, with many liquid biopsy studies (both in urine and plasma/serum) being currently performed, but there is not an agreeable miRNA or set of miRNAs that is ready to follow the footsteps of miR-371a-3p in TGCTs. SUMMARY Further studies with proper molecular characterization of miRNA profiles of GU malignancies and standardization of sampling, biobanking and formal analysis may aid in the advance and choosing of specific target sets to be used for occult cancer detection.
Collapse
Affiliation(s)
- Nuno Tiago Tavares
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre Raquel Seruca (Porto.CCC)
- Doctoral Programme in Biomedical Sciences, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP)
| | - João Lobo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre Raquel Seruca (Porto.CCC)
- Department of Pathology, Portuguese Oncology Institute of Porto/Porto Comprehensive Cancer Centre Raquel Seruca (Porto.CCC)
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal
| | - Aditya Bagrodia
- Department of Urology, University of California - San Diego Health, San Diego, California
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
49
|
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
|
50
|
Lopez-Beltran A. Testicular cancer: new developments, molecular pathology, and current research keynote. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:204-207. [PMID: 37975918 DOI: 10.1007/s00292-023-01264-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
Germ cell tumors (GCTs) are now considered a curable cancer, with a > 95% cure rate in all patients and about 90% cure rate in patients with metastatic disease. The success of physicians in curing the disease is underpinned by multidisciplinary advances. Of relevance in this regard are the nowadays-applied homogeneous terminology based on pathologically better characterized testicular neoplasms and the development of a widely used risk stratification model for metastatic disease introduced by the International Germ Cell Cancer Collaborative Group in 1997 and updated in 2021. Non-pulmonary visceral metastases, high levels of the serum tumor markers alpha-fetoprotein (AFP) and human chorionic gonadotropin (HCG), and primary mediastinal non-seminoma are currently identified as determinants of poor prognosis. In addition, the presence of distinct microRNA profiles between seminomas and non-seminoma GCTs has opened up important perspectives in terms of noninvasive biomarkers that can be used in diagnosis and treatment monitoring.
Collapse
Affiliation(s)
- Antonio Lopez-Beltran
- Department of Morphological Sciences, Cordoba University Medical School, Avda. Menendez-Pidal S/N, 14004, Cordoba, Spain.
| |
Collapse
|