1
|
Lobo A, Collins K, Kaushal S, Acosta AM, Akgul M, Adhya AK, Al-Ahmadie HA, Al-Obaidy KI, Amin A, Amin MB, Aron M, Balzer BL, Biswal R, Mohanty S, Browning L, Chakrabarti I, Cima L, Cimadamore A, Desai S, Dhillon J, Deshwal A, Diego GG, Diwaker P, Galea LA, Magi-Galluzzi C, Giannico GA, Gupta NS, Haider A, Hirsch MS, Iczkowski KA, Arora S, Jain E, Jain D, Jha S, Kandukuri S, Kao CS, Kryvenko ON, Kumar RM, Kumari N, Kunju LP, Kuthi L, Lobo J, Lopez JI, Luthringer DJ, Maclean F, Manini C, Mannan R, Martos MG, Mehra R, Menon S, Mishra P, Moch H, Montironi R, Baisakh MR, Netto GJ, Nigam LK, Osunkoya AO, Pagliuca F, Paner GP, Panizo A, Parwani AV, Picken MM, Prendeville S, Przybycin CG, Purkait S, Queipo FJ, Rao BV, Rao P, Reuter VE, Sancheti S, Sangoi AR, Sardana R, Satturwar S, Shah RB, Sharma S, Dixit M, Verma M, Sirohi D, Smith SC, Soni S, Sundaram S, Swain M, Tretiakova M, Trpkov K, MuñizUnamunzaga G, Zhou M, Williamson SR, Lopez-Beltran A, Cheng L, Mohanty SK. Advances, recognition, and interpretation of molecular heterogeneity among conventional and subtype histology of urothelial carcinoma (UC): a survey among urologic pathologists and comprehensive review of the literature. Histopathology 2024; 85:748-759. [PMID: 39075659 DOI: 10.1111/his.15287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/31/2024]
Abstract
AIMS Urothelial carcinoma (UC) demonstrates significant molecular and histologic heterogeneity. The WHO 2022 classification has hinted at adding molecular signatures to the morphologic diagnosis. As morphology and associated molecular repertoire may potentially translate to choices of and response to therapy and relapse rate, broader acceptability of recognizing these key features among uropathologists is needed. This prompted an international survey to ascertain the practice patterns in classical/subtype UC among uropathologists across the globe. METHODS AND RESULTS A survey instrument was shared among 98 uropathologists using SurveyMonkey software. Anonymized respondent data were analysed. The response rate was 85%. A majority were in concordance with the profiles of luminal (93%) and basal (82%) types. Opinion on the FGFR3 testing platform was variable. While 95% concurred that TERT promoter mutation is the key driver in UC, 72% had the opinion that APOBEC mutagenesis is the main signature in muscle invasive bladder cancer (MIBC). Uropathologists have divergent opinions on MIBC and ERCC2 mutations. Among the participants, 94% would quantify aggressive micropapillary and sarcomatoid histology, while 88% would reevaluate another transurethral resection of the bladder tumour specimen in nonmuscle invasive tumour with micropapillary, small cell, or sarcomatoid histology. A leading number agreed to specific molecular signatures of micropapillary (93%), plasmacytoid (97%), and small cell (86%) subtypes. Ninety-six percent of participants agreed that a small-cell component portends a more aggressive course and should be treated with neoadjuvant chemotherapy and 63% would perform HER2/neu testing only on oncologist's request in advanced tumours. Ninety percent agreed that microsatellite instability testing, although not a standard protocol, should be considered in young patients with upper tract UC. Eighty-six percent agreed that UC with high tumour mutational burden would be a better candidate for immunotherapy. CONCLUSION In the era of precision medicine, enhanced understanding of molecular heterogeneity of UC will contribute to better therapeutic options, novel biomarker discovery, innovative management protocols, and outcomes. Our survey provides a broad perspective of pathologists' perceptions and experience regarding incorporation of histomolecular approaches to "personalize" therapy. Due to variable clinical adoption, there is a need for additional data using uniform study criteria. This will drive generation of best practice guidelines in this area for widespread and consistent clinical utility.
Collapse
Affiliation(s)
- Anandi Lobo
- Department of Pathology, Kapoor Centre of Urology and Pathology, Raipur, India
| | - Katrina Collins
- Department of Pathology, Indiana University Health, Indiana, USA
| | - Seema Kaushal
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Andres M Acosta
- Department of Pathology, Indiana University Health, Indiana, USA
| | - Mahmut Akgul
- Department of Pathology, Albany Medical Center, Albany, USA
| | - Amit K Adhya
- Department of Pathology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Hikmat A Al-Ahmadie
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | | | - Ali Amin
- Department of Pathology, Alpert Medical School of Brown University, Providence, USA
| | - Mahul B Amin
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, USA
| | - Manju Aron
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, USA
| | - Bonnie L Balzer
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Rupanita Biswal
- Department of Pathology, Bagchi Sri Shankara Cancer Hospital, Bhubaneswar, India
| | - Subashish Mohanty
- Department of Pathology, SUM Ultimate Medicare Hospital, Bhubaneswar, India
| | - Lisa Browning
- Department of Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Indranil Chakrabarti
- Department of Pathology, All India Institute of Medical Sciences, Kalyani, India
| | - Luca Cima
- Department of Pathology, Santa Chiara Hospital of Trento, Trento, Italy
| | - Alessia Cimadamore
- Department of Pathology, Molecular Medicine and Cell Therapy Foundation, c/o Polytechnic University of the Marche Region, Ancona, Italy
| | - Sangeeta Desai
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | | | | | - Guillermo G Diego
- Department of Pathology, University Gregorio Marañon Hospital, Madrid, Spain
| | - Preeti Diwaker
- Department of Pathology, University College of Medical Sciences, New Delhi, India
| | - Laurence A Galea
- Department of Pathology, Melbourne Pathology, Melbourne, Australia
| | | | | | - Nilesh S Gupta
- Department of Pathology, Henry Ford Health System, Detroit, USA
| | - Aiman Haider
- Department of Pathology, University College London Hospitals NHS Foundation Trust, London, UK
| | | | | | - Samriti Arora
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Ekta Jain
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Deepika Jain
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Shilpy Jha
- Department of Pathology, Advanced Medical and Research Institute, Bhubaneswar, India
| | - Shivani Kandukuri
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, USA
| | - Chia-Sui Kao
- Department of Pathology, Cleveland Clinic, Cleveland, USA
| | - Oleksandr N Kryvenko
- Department of Pathology, University of Miami Miller School of Medicine, Miami, USA
| | - Ramani M Kumar
- Department of Pathology, Dane Diagnostics, Palakkad, India
| | - Niraj Kumari
- Department of Pathology, All India Institute of Medical Sciences, Raebareli, India
| | - Lakshmi P Kunju
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Levente Kuthi
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - João Lobo
- Department of Pathology, Portuguese Oncology Institute - Porto, Porto, Portugal
| | - Jose I Lopez
- Department of Pathology, Cruces University Hospital, Barakaldo, Spain
| | | | - Fiona Maclean
- Department of Pathology, Douglass Hanly Moir Pathology, Sydney, Australia
| | - Claudia Manini
- Department of Pathology, University of Turin, Turin, Italy
| | - Rahul Mannan
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - María G Martos
- Department of Pathology, University Gregorio Marañon Hospital, Madrid, Spain
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Santosh Menon
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Pritinanda Mishra
- Department of Pathology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Holger Moch
- Department of Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Rodolfo Montironi
- Department of Pathology, Molecular Medicine and Cell Therapy Foundation, c/o Polytechnic University of the Marche Region, Ancona, Italy
| | - Manas R Baisakh
- Department of Pathology, Prolife Diagnostics, Bhubaneswar, India
| | - George J Netto
- Department of Pathology, University of Pennsylvania, Philadelphia, USA
| | - Lovelesh K Nigam
- Department of Pathology, Institute of Kidney Diseases and Research Center, Ahmedabad, India
| | - Adeboye O Osunkoya
- Department of Pathology, Emory University School of Medicine, Atlanta, USA
| | - Francesca Pagliuca
- Department of Pathology, Università degliStudidella Campania Luigi Vanvitelli, Caserta, Italy
| | - Gladell P Paner
- Department of Pathology, University of Chicago, Chicago, USA
| | - Angel Panizo
- Department of Pathology, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Anil V Parwani
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Maria M Picken
- Department of Pathology, Loyola University Medical Center, Hines, USA
| | - Susan Prendeville
- Department of Pathology, University Health Network, University of Toronto, Toronto, Canada
| | | | - Suvendu Purkait
- Department of Pathology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Francisco J Queipo
- Department of Pathology, Hospital Universitario de A Coruna, A Coruna, Spain
| | - B Vishal Rao
- Department of Pathology, Basavatarakam Indo-American Cancer Hospital and Research Institute, Hyderabad, India
| | - Priya Rao
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Victor E Reuter
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Sankalp Sancheti
- Department of Pathology, Homi Bhabha Cancer Hospital, Punjab, India
| | - Ankur R Sangoi
- Department of Pathology, Stanford University, Stanford, USA
| | - Rohan Sardana
- Department of Pathology, Sardana Laboratories, Jalandhar, India
| | - Swati Satturwar
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, USA
| | - Rajal B Shah
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, USA
| | - Shivani Sharma
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Mallika Dixit
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Monica Verma
- Department of Pathology, CORE Diagnostics, Gurgaon, India
| | - Deepika Sirohi
- Department of Pathology, University of California, San Francisco, USA
| | - Steven C Smith
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, USA
| | - Shailesh Soni
- Department of Pathology, Muljibhai Patel Urological Hospital, Nadiad, India
| | - Sandhya Sundaram
- Department of Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | | | | | - Kiril Trpkov
- Department of Pathology, University of Calgary, Calgary, Canada
| | | | - Ming Zhou
- Department of Pathology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | - Antonio Lopez-Beltran
- Department of Pathology, Unit of Anatomical Pathology, Faculty of Medicine, Cordoba University, Cordoba, Spain
| | - Liang Cheng
- Department of Pathology, Alpert Medical School of Brown University, Providence, USA
| | - Sambit K Mohanty
- Department of Pathology, CORE Diagnostics, Gurgaon, India
- Department of Pathology, Advanced Medical and Research Institute, Bhubaneswar, India
| |
Collapse
|
2
|
Hellgren LS, Stenman A, Jatta K, Condello V, Larsson C, Zedenius J, Juhlin CC. Catching the Silent Culprits: TERT Promoter Mutation Screening of Minimally Invasive Follicular and Oncocytic Thyroid Carcinoma in Clinical Practice. Endocr Pathol 2024:10.1007/s12022-024-09828-x. [PMID: 39363120 DOI: 10.1007/s12022-024-09828-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
De-escalation of thyroid cancer treatment is crucial to prevent overtreatment of indolent disease, but it remains important to identify clinically aggressive cases. TERT promoter mutations are molecular events frequently associated with high-risk thyroid tumors with poor outcomes and may identify cases at risk of dissemination. In various international guidelines, small minimally invasive follicular thyroid carcinoma and oncocytic thyroid carcinoma (miFTC/miOTC) are classified as low-risk lesions and are not recommended adjuvant treatment. Our study aimed to explore the association between size-based risk assessment and TERT promoter mutations. Between 2019 and May 2024, 84 miFTCs/miOTCs diagnosed at our department underwent digital droplet PCR analysis targeting TERT promoter mutational hotspots C228T and C250T in clinical routine. TERT promoter mutations were found in 10 out of 84 cases (11.9%). Mutated cases were pT1 (n = 1), pT2 (n = 3), or pT3 (n = 6). Patients with mutated tumors were older compared to patients with wild-type tumors (median age of 71 years vs. 57 years, p = 0.041). There were no significant differences regarding patient sex, tumor size, Ki-67 labeling index, or the presence of distant metastases. Notably, 30% of mutations displayed variant allele frequencies < 10%, possibly suggesting subclonal events. To conclude, TERT promoter mutations in miFTCs and miOTCs were associated with higher patient age and were often suspected to be subclonal. However, they did not affect clinical outcomes, possibly due to short follow-up. Reflex testing for this genetic alteration in miFTCs and miOTCs could be justified regardless of tumor size, though the clinical benefit remains uncertain.
Collapse
Affiliation(s)
- L Samuel Hellgren
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden.
| | - Adam Stenman
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Breast, Endocrine Tumors, and Sarcoma, Karolinska University Hospital, Stockholm, Sweden
| | - Kenbugul Jatta
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Vincenzo Condello
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Catharina Larsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Jan Zedenius
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Breast, Endocrine Tumors, and Sarcoma, Karolinska University Hospital, Stockholm, Sweden
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
3
|
Huang FF, Di XF, Bai MH. Analysis of urine cell-free DNA in bladder cancer diagnosis by emerging bioactive technologies and materials. Front Bioeng Biotechnol 2024; 12:1458362. [PMID: 39295845 PMCID: PMC11408225 DOI: 10.3389/fbioe.2024.1458362] [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: 07/02/2024] [Accepted: 08/23/2024] [Indexed: 09/21/2024] Open
Abstract
Urinary cell-free DNA (UcfDNA) is gaining recognition as an important biomarker for diagnosing bladder cancer. UcfDNA contains tumor derived DNA sequences, making it a viable candidate for non-invasive early detection, diagnosis, and surveillance of bladder cancer. The quantification and qualification of UcfDNA have demonstrated high sensitivity and specificity in the molecular characterization of bladder cancer. However, precise analysis of UcfDNA for clinical bladder cancer diagnosis remains challenging. This review summarizes the history of UcfDNA discovery, its biological properties, and the quantitative and qualitative evaluations of UcfDNA for its clinical significance and utility in bladder cancer patients, emphasizing the critical role of UcfDNA in bladder cancer diagnosis. Emerging bioactive technologies and materials currently offer promising tools for multiple UcfDNA analysis, aiming to achieve more precise and efficient capture of UcfDNA, thereby significantly enhancing diagnostic accuracy. This review also highlights breakthroughs in detection technologies and substrates with the potential to revolutionize bladder cancer diagnosis in clinic.
Collapse
Affiliation(s)
- Fei-Fei Huang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xiao-Fei Di
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Mo-Han Bai
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Hernandez-Prera JC, Wenig BM. RAS-Mutant Follicular Thyroid Tumors: A Continuous Challenge for Pathologists. Endocr Pathol 2024; 35:167-184. [PMID: 38888731 DOI: 10.1007/s12022-024-09812-5] [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] [Accepted: 05/20/2024] [Indexed: 06/20/2024]
Abstract
The classification of thyroid nodules, particularly those with a follicular growth pattern, has significantly evolved. These tumors, enriched with RAS or RAS-like mutations, remain challenging for pathologists due to variables such as nuclear atypia, invasion, mitotic activity, and tumor necrosis. This review addresses the histological correlates of benign, low-risk, and malignant RAS-mutant thyroid tumors, as well as some difficult-to-classify follicular nodules with worrisome features. One prototypical RAS-mutant nodule is non-invasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP). The assessment of nuclear characteristics in encapsulated/well-demarcated non-invasive RAS-mutant follicular-patterned tumors helps distinguish between follicular thyroid adenoma (FTA) and NIFTP. Despite this straightforward concept, questions about the degree of nuclear atypia necessary for the diagnosis of NIFTP are common in clinical practice. The nomenclature of follicular nodules lacking clear invasive features with increased mitotic activity, tumor necrosis, and/or high-risk mutations (e.g., TERT promoter or TP53) remains debated. Invasion, particularly angioinvasion, is the current hallmark of malignancy in RAS-mutant follicular-patterned neoplasms, with follicular thyroid carcinoma (FTC) as the model. Assessing the tumor interface is critical, though full capsule evaluation can be challenging. Multiple levels and NRASQ61R-specific immunohistochemistry can aid in identifying invasion. Controversies around vascular invasion persist, with ancillary stains like CD31, ERG, and CD61 aiding in its evaluation. Moreover, the review highlights that invasive encapsulated follicular variant papillary thyroid carcinoma (IEFVPTC) is closely associated with FTC, suggesting the need for better nomenclature. The concept of "high-grade" differentiated carcinomas, applicable to FTC or IEFVPTC with necrosis and/or high mitotic activity, is also discussed.
Collapse
Affiliation(s)
- Juan C Hernandez-Prera
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| | - Bruce M Wenig
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| |
Collapse
|
5
|
Kovács Á, Sükösd F, Kuthi L, Boros IM, Vedelek B. Novel method for detecting frequent TERT promoter hot spot mutations in bladder cancer samples. Clin Exp Med 2024; 24:192. [PMID: 39141194 PMCID: PMC11324672 DOI: 10.1007/s10238-024-01464-3] [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: 05/10/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024]
Abstract
Telomerase reverse transcriptase promoter (TERTp) mutations are frequently targeted tumor markers, however, they reside in regions with high GC content, which poses challenges when examined with simple molecular techniques or even with next-generation sequencing (NGS). In bladder cancer (BC), TERTp mutations are particularly frequent, however, none of the available tools have demonstrated efficacy in detecting TERTp mutations via a simple noninvasive technique. Therefore, we developed a novel PCR-based method for the detection of the two most common TERTp mutations and demonstrated its use for the analysis of BC samples. The developed SHARD-PCR TERTp mutation detection technique requires PCR and restriction digestion steps that are easily implementable even in less well-equipped laboratories. Cell lines with known mutational status were utilized for method development. Matching urine and tumor tissue samples from BC patients were analyzed, and the results were validated by next-generation sequencing. Analysis of eighteen urine and corresponding tumor tissue samples by SHARD-PCR revealed perfect matches in sample pairs, which paralleled the corresponding NGS results: fourteen samples exhibited mutations at the -124 position, two samples showed mutations at the -146 position, and no mutations were detected in two samples. Our study serves as a proof-of-concept and is limited by its small sample size, nonetheless, it demonstrates that SHARD-PCR is a simple, economic and highly reliable method for detecting TERTp mutations, which are common in different cancer types. For bladder cancer, SHARD-PCR can be performed with the use of noninvasive samples and could replace or complement currently used techniques.
Collapse
Affiliation(s)
- Ákos Kovács
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Farkas Sükösd
- Department of Pathology, University of Szeged, Szeged, Hungary
| | - Levente Kuthi
- Department of Pathology, University of Szeged, Szeged, Hungary
| | - Imre M Boros
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary.
- Hungarian Research Network Biological Research Center, Institute of Biochemistry, Szeged, Hungary.
| | - Balázs Vedelek
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary.
- Hungarian Research Network Biological Research Center, Institute of Genetics, Szeged, Hungary.
| |
Collapse
|
6
|
Tsuruoka M, Ninomiya M, Inoue J, Iwata T, Sano A, Sato K, Onuki M, Sawahashi S, Masamune A. Changes in Mutations of Cell-Free DNA and Liver Tumor Tissue in Patients with Advanced Hepatocellular Carcinoma before and after Introduction of Lenvatinib. Oncology 2024:1-12. [PMID: 39047713 DOI: 10.1159/000540438] [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: 03/10/2024] [Accepted: 07/07/2024] [Indexed: 07/27/2024]
Abstract
INTRODUCTION Cell-free DNA (cfDNA) is expected to contribute to the decision for treatment and prediction of effects with minimally invasion. We investigated the correlation between gene mutations before and after lenvatinib (LEN) treatment and its effectiveness, in order to find advanced hepatocellular carcinoma (HCC) patients who would benefit greatly from the therapy. METHODS We analyzed cfDNA before and 6-8 weeks after the start of treatment in 20 advanced HCC patients who started LEN. A next-generation sequencer was used for CTNNB1 and TP53. Concerning TERT promoter, -124C>T and -146C>T mutations are researched using digital PCR. In addition, we examined liver tumor biopsy tissues by the same method. Computerized tomography evaluation was performed at 6-8 weeks and 3-4 months to assess the efficacy. RESULTS Frequencies of TERT promoter, CTNNB1, and TP53 mutations in pretreatment cfDNA were 45%, 65%, and 65%, but 53%, 41%, and 47% in HCC tissues, respectively. There were no clear correlations between these gene mutations and the disease-suppressing effect or progression-free survival. Overall, there were many cases showing a decrease in mutations after LEN treatment. Integrating the reduction of CTNNB1 and TP53 genetic mutations increased the potential for disease suppression. CONCLUSION This study suggests that analysis of cfDNA in advanced HCC patients may be useful for identifying LEN responders and determining therapeutic efficacy. Furthermore, it has potential for selecting responders for other molecular-targeted drugs.
Collapse
Affiliation(s)
- Mio Tsuruoka
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan,
| | - Masashi Ninomiya
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jun Inoue
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoaki Iwata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akitoshi Sano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kosuke Sato
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masazumi Onuki
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoko Sawahashi
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
7
|
Devins KM, Ordulu Z, Mendoza RP, Croce S, Haridas R, Wanjari P, Pinto A, Oliva E, Bennett JA. Uterine Inflammatory Myofibroblastic Tumors: p16 as a Surrogate for CDKN2A Deletion and Predictor of Aggressive Behavior. Am J Surg Pathol 2024; 48:813-824. [PMID: 38630911 DOI: 10.1097/pas.0000000000002220] [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: 04/19/2024]
Abstract
Uterine inflammatory myofibroblastic tumors (IMTs) are rare mesenchymal neoplasms of uncertain malignant potential. Aside from the recently described risk stratification score, which has not been validated by other studies, and rare reports of aberrant p16 expression in malignant tumors, there are no criteria to reliably predict behavior. Herein, we evaluated the clinicopathologic features and p16 expression patterns in 30 IMTs, with genomic profiling performed in a subset (13 malignant, 3 benign). Fifteen patients had malignant IMTs, defined by extrauterine disease at diagnosis (n=5) or recurrence (n=10; median: 24 mo). Patients ranged from 8 to 65 (median: 51) years and tumors from 6 to 22 (median: 12.5) cm. In primary tumors (n=13), infiltrative borders were noted in 10, moderate/severe cytologic atypia in 9, tumor cell necrosis in 7, and lymphovascular invasion in 6, while mitoses ranged from 0 to 21 (median: 7) per 10 high-power fields. In contrast, 15 patients with benign IMTs ranged from 28 to 65 (median: 44) years, with follow-up of 18 to 114 (median: 41) months. Tumors ranged from 1.9 to 8.5 (median: 5.5) cm, 2 demonstrated infiltrative borders, and 1 had moderate cytologic atypia. No other high-risk histologic features were observed. Application of the previously described clinicopathologic risk stratification score in all primary IMTs with complete data (n=18) classified 8 as high-risk (all malignant), 8 as intermediate-risk (3 malignant, 5 benign), and 2 as low-risk (benign). p16 was aberrant in all malignant IMTs, with <1% expression noted in 10, overexpression (>90%) in 4, and subclonal loss in 1; all benign tumors had patchy staining (20% to 80%; median 50%). Molecular analysis detected CDKN2A deletions in 8 of 9 tumors with <1% p16 expression, while the other harbored a TERT promoter mutation. TERT promoter mutations were also identified in 2 of 3 IMTs with p16 overexpression. Neither of these alterations was detected in the 3 sequenced benign IMTs. Thus, we recommend performing p16 on all uterine IMTs, which, combined with the risk stratification score, is a promising and cost-effective tool for predicting CDKN2A status and outcome in these patients. It may be particularly useful for tumors with incomplete information for risk stratification (ie, morcellated tumors) and for further stratifying intermediate-risk IMTs when sequencing is unavailable.
Collapse
Affiliation(s)
- Kyle M Devins
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Zehra Ordulu
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Rachelle P Mendoza
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY
| | - Sabrina Croce
- Department of Biopathology, Institut Bergonie, Bordeaux, France
| | | | | | - Andre Pinto
- Department of Pathology, University of Miami, Miami, FL
| | - Esther Oliva
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | |
Collapse
|
8
|
Jin W, Xia Y, Thela SR, Liu Y, Chen L. In silico generation and augmentation of regulatory variants from massively parallel reporter assay using conditional variational autoencoder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600715. [PMID: 38979263 PMCID: PMC11230389 DOI: 10.1101/2024.06.25.600715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Predicting the functional consequences of genetic variants in non-coding regions is a challenging problem. Massively parallel reporter assays (MPRAs), which are an in vitro high-throughput method, can simultaneously test thousands of variants by evaluating the existence of allele specific regulatory activity. Nevertheless, the identified labelled variants by MPRAs, which shows differential allelic regulatory effects on the gene expression are usually limited to the scale of hundreds, limiting their potential to be used as the training set for achieving a robust genome-wide prediction. To address the limitation, we propose a deep generative model, MpraVAE, to in silico generate and augment the training sample size of labelled variants. By benchmarking on several MPRA datasets, we demonstrate that MpraVAE significantly improves the prediction performance for MPRA regulatory variants compared to the baseline method, conventional data augmentation approaches as well as existing variant scoring methods. Taking autoimmune diseases as one example, we apply MpraVAE to perform a genome-wide prediction of regulatory variants and find that predicted regulatory variants are more enriched than background variants in enhancers, active histone marks, open chromatin regions in immune-related cell types, and chromatin states associated with promoter, enhancer activity and binding sites of cMyC and Pol II that regulate gene expression. Importantly, predicted regulatory variants are found to link immune-related genes by leveraging chromatin loop and accessible chromatin, demonstrating the importance of MpraVAE in genetic and gene discovery for complex traits.
Collapse
Affiliation(s)
- Weijia Jin
- Department of Biostatistics, University of Florida, Gainesville, FL, 32603, USA
| | - Yi Xia
- Department of Biostatistics, University of Florida, Gainesville, FL, 32603, USA
| | - Sai Ritesh Thela
- Department of Biostatistics, University of Florida, Gainesville, FL, 32603, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Li Chen
- Department of Biostatistics, University of Florida, Gainesville, FL, 32603, USA
| |
Collapse
|
9
|
Gao R, Lou N, Li L, Xie T, Xing P, Tang L, Yao J, Han X, Shi Y. Mutational variant allele frequency profile as a biomarker of response to immune checkpoint blockade in non-small cell lung Cancer. J Transl Med 2024; 22:576. [PMID: 38890738 PMCID: PMC11184775 DOI: 10.1186/s12967-024-05400-7] [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: 04/06/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
INTRODUCTION Identifying new biomarkers for predicting immune checkpoint inhibitors (ICIs) response in non-small cell lung cancer (NSCLC) is crucial. We aimed to assess the variant allele frequency (VAF)-related profile as a novel biomarker for NSCLC personalized therapy. METHODS We utilized genomic data of 915 NSCLC patients via cBioPortal and a local cohort of 23 patients for model construction and mutational analysis. Genomic, transcriptomic data from 952 TCGA NSCLC patients, and immunofluorescence (IF) assessment with the local cohort supported mechanism analysis. RESULTS Utilizing the random forest algorithm, a 15-gene VAF-related model was established, differentiating patients with durable clinical benefit (DCB) from no durable benefit (NDB). The model demonstrated robust performance, with ROC-AUC values of 0.905, 0.737, and 0.711 across training (n = 313), internal validation (n = 133), and external validation (n = 157) cohorts. Stratification by the model into high- and low-score groups correlated significantly with both progression-free survival (PFS) (training: P < 0.0001, internal validation: P < 0.0001, external validation: P = 0.0066) and overall survival (OS) (n = 341) (P < 0.0001). Notably, the stratification system was independent of PD-L1 (P < 0.0001) and TMB (P < 0.0001). High-score patients exhibited an increased DCB ratio and longer PFS across both PD-L1 and TMB subgroups. Additionally, the high-score group appeared influenced by tobacco exposure, with activated DNA damage response pathways. Whereas, immune/inflammation-related pathways were enriched in the low-score group. Tumor immune microenvironment analyses revealed higher proportions of exhausted/effector memory CD8 + T cells in the high-score group. CONCLUSIONS The mutational VAF profile is a promising biomarker for ICI therapy in NSCLC, with enhanced therapeutic stratification and management as a supplement to PD-L1 or TMB.
Collapse
Affiliation(s)
- Ruyun Gao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Ning Lou
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Lin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Tongji Xie
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Puyuan Xing
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Le Tang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Jiarui Yao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
10
|
Hernandez-Prera JC. Molecular Pathology of Thyroid Tumors: Old Problems and New Concepts. Clin Lab Med 2024; 44:305-324. [PMID: 38821646 DOI: 10.1016/j.cll.2023.08.006] [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: 06/02/2024]
Abstract
The molecular signatures of many thyroid tumors have been uncovered. These discoveries have translated into clinical practice and are changing diagnostic and tumor classification paradigms. Here, the findings of recent studies are presented with special emphasis on how molecular insights are impacting the understating of RAS mutant thyroid nodules, Hürthel cell neoplasms, and unusual thyroid tumors, such as hyalinizing trabecular tumor, secretory carcinoma of the thyroid, and sclerosing mucoepidermoid carcinoma with eosinophilia. In addition, the utility of detecting actionable molecular alterations by immunohistochemistry in advanced and aggressive thyroid cancer is also discussed.
Collapse
Affiliation(s)
- Juan C Hernandez-Prera
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, Florida 33612, USA.
| |
Collapse
|
11
|
Drobyshev A, Modestov A, Suntsova M, Poddubskaya E, Seryakov A, Moisseev A, Sorokin M, Tkachev V, Zakharova G, Simonov A, Zolotovskaia MA, Buzdin A. Pan-cancer experimental characteristic of human transcriptional patterns connected with telomerase reverse transcriptase ( TERT) gene expression status. Front Genet 2024; 15:1401100. [PMID: 38859942 PMCID: PMC11163056 DOI: 10.3389/fgene.2024.1401100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024] Open
Abstract
The TERT gene encodes the reverse transcriptase subunit of telomerase and is normally transcriptionally suppressed in differentiated human cells but reactivated in cancers where its expression is frequently associated with poor survival prognosis. Here we experimentally assessed the RNA sequencing expression patterns associated with TERT transcription in 1039 human cancer samples of 27 tumor types. We observed a bimodal distribution of TERT expression where ∼27% of cancer samples did not express TERT and the rest showed a bell-shaped distribution. Expression of TERT strongly correlated with 1443 human genes including 103 encoding transcriptional factor proteins. Comparison of TERT- positive and negative cancers showed the differential activation of 496 genes and 1975 molecular pathways. Therein, 32/38 (84%) of DNA repair pathways were hyperactivated in TERT+ cancers which was also connected with accelerated replication, transcription, translation, and cell cycle progression. In contrast, the level of 40 positive cell cycle regulator proteins and a set of epithelial-to-mesenchymal transition pathways was specific for the TERT- group suggesting different proliferation strategies for both groups of cancer. Our pilot study showed that the TERT+ group had ∼13% of cancers with C228T or C250T mutated TERT promoter. However, the presence of promoter mutations was not associated with greater TERT expression compared with other TERT+ cancers, suggesting parallel mechanisms of its transcriptional activation in cancers. In addition, we detected a decreased expression of L1 retrotransposons in the TERT+ group, and further decreased L1 expression in promoter mutated TERT+ cancers. TERT expression was correlated with 17 genes encoding molecular targets of cancer therapeutics and may relate to differential survival patterns of TERT- positive and negative cancers.
Collapse
Affiliation(s)
- Aleksey Drobyshev
- Endocrinology Research Center, Moscow, Russia
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexander Modestov
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maria Suntsova
- Endocrinology Research Center, Moscow, Russia
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Elena Poddubskaya
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
- Clinical Center Vitamed, Moscow, Russia
| | | | - Aleksey Moisseev
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Maksim Sorokin
- Endocrinology Research Center, Moscow, Russia
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Galina Zakharova
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Aleksander Simonov
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Marianna A. Zolotovskaia
- Endocrinology Research Center, Moscow, Russia
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
- Moscow Center for Advanced Studies 20, Moscow, Russia
| | - Anton Buzdin
- Endocrinology Research Center, Moscow, Russia
- Institute of Personalized Oncology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
- Moscow Center for Advanced Studies 20, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
12
|
Chen B, Weng Y, Li M, Bian Z, Tao Y, Zhou W, Lu H, He S, Liao R, Huang J, Wang Q, Xu M, Ge Y, Cao W, Lei M, Zhang Y. LINC02454-CCT complex interaction is essential for telomerase activity and cell proliferation in head and neck squamous cell carcinoma. Cancer Lett 2024; 588:216734. [PMID: 38401886 DOI: 10.1016/j.canlet.2024.216734] [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: 10/27/2023] [Revised: 02/03/2024] [Accepted: 02/13/2024] [Indexed: 02/26/2024]
Abstract
Telomerase activity is upregulated in head and neck squamous cell carcinoma (HNSCC), yet its regulatory mechanisms remain unclear. Here, we identified a cancer-specific lncRNA (LINC02454) associated with poor prognosis by using LncRNA chip of our HNSCC cohorts and external datasets. Through employing negative-stain transmission electron microscopy (NS-TEM), we discovered an interaction between LINC02454 and CCT complex which would augment telomerase activity for maintaining telomere homeostasis. Supporting this, in the telomerase repeat amplification protocol (TRAP) assay and quantitative fluorescence in situ hybridization (Q-FISH) analysis, LINC02454 depletion significantly reduced telomerase activity and shortened telomere length. Consistently, pathways related to telomerase, mitosis, and apoptosis were significantly impacted upon LINC02454 knockdown in RNAseq analysis. Functionally, LINC02454-deficient cells exhibited a more significant senescence phenotype in β-galactosidase staining, cell cycle, and apoptosis assays. We further confirmed the role of LINC02454 in HNSCC proliferation through a combination of in vitro and in vivo experiments. The therapeutic potential of targeting LINC02454 was verified by adenovirus-shRNA approach in HNSCC patient-derived xenograft (PDX) models. In summary, our findings provided valuable insights into the molecular mechanisms of HNSCC tumorigenesis and potential targets for future treatment modalities.
Collapse
Affiliation(s)
- Biying Chen
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China; Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Yue Weng
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mingyue Li
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhouliang Bian
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China; Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Ye Tao
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenkai Zhou
- Department of Oral Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Hong Lu
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shufang He
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Rijing Liao
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Huang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Wang
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China; Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Ming Xu
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China; Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Yunhui Ge
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei Cao
- Department of Oral Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| | - Ming Lei
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yanjie Zhang
- Department of Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China; Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China.
| |
Collapse
|
13
|
Xiao Z, Ying W, Xing Z, Zhihui L, Qiuyu Z, Caijiao H, Changlong L, Shi H, Deng L, Zhenwen C, Jianquan N, Xueyun H, Xiaoyan D. Unexpected mutations occurred in CRISPR/Cas9 edited Drosophila analyzed by deeply whole genomic sequencing. Heliyon 2024; 10:e29061. [PMID: 38596060 PMCID: PMC11002691 DOI: 10.1016/j.heliyon.2024.e29061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/11/2024] Open
Abstract
CRISPR/Cas9 possesses the most promising prospects as a gene-editing tool in post-genomic researches. It becomes an epoch-marking technique for the features of speed and convenience of genomic modification. However, it is still unclear whether CRISPR/Cas9 gene editing can cause irreversible damage to the genome. In this study, we successfully knocked out the WHITE gene in Drosophila, which governs eye color, utilizing CRISPR/Cas9 technology. Subsequently, we conducted high-throughput sequencing to assess the impact of this editing process on the stability of the entire genomic profile. The results revealed the presence of numerous unexpected mutations in the Drosophila genome, including 630 SNVs (Single Nucleotide Variants), 525 Indels (Insertion and Deletion) and 425 MSIs (microsatellite instability). Although the KO (knockout) specifically occurred on chromosome X, the majority of mutations were observed on chromosome 3, indicating that this effect is genome-wide and associated with the spatial structure between chromosomes, rather than being solely limited to the location of the KO gene. It is worth noting that most of the mutations occurred in the intergenic and intron regions, without exerting any significant on the function or healthy of the animal. In addition, the mutations downstream of the knockout gene well beyond the upstream. This study has found that gene editing can lead to unexpected mutations in the genome, but most of these mutations are harmless. This research has deepened our understanding of CRISPR/Cas9 and broadened its application prospects.
Collapse
Affiliation(s)
- Zhu Xiao
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Wu Ying
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Zhang Xing
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Li Zhihui
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Zhang Qiuyu
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Hu Caijiao
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Li Changlong
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Hanping Shi
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Li Deng
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Chen Zhenwen
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Ni Jianquan
- Gene Regulatory Laboratory, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Huo Xueyun
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Du Xiaoyan
- School of Basic Medical Sciences, Capital Medical University, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Prime SS, Darski P, Hunter KD, Cirillo N, Parkinson EK. A Review of the Repair of DNA Double Strand Breaks in the Development of Oral Cancer. Int J Mol Sci 2024; 25:4092. [PMID: 38612901 PMCID: PMC11012950 DOI: 10.3390/ijms25074092] [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] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
We explore the possibility that defects in genes associated with the response and repair of DNA double strand breaks predispose oral potentially malignant disorders (OPMD) to undergo malignant transformation to oral squamous cell carcinoma (OSCC). Defects in the homologous recombination/Fanconi anemia (HR/FA), but not in the non-homologous end joining, causes the DNA repair pathway to appear to be consistent with features of familial conditions that are predisposed to OSCC (FA, Bloom's syndrome, Ataxia Telangiectasia); this is true for OSCC that occurs in young patients, sometimes with little/no exposure to classical risk factors. Even in Dyskeratosis Congenita, a disorder of the telomerase complex that is also predisposed to OSCC, attempts at maintaining telomere length involve a pathway with shared HR genes. Defects in the HR/FA pathway therefore appear to be pivotal in conditions that are predisposed to OSCC. There is also some evidence that abnormalities in the HR/FA pathway are associated with malignant transformation of sporadic cases OPMD and OSCC. We provide data showing overexpression of HR/FA genes in a cell-cycle-dependent manner in a series of OPMD-derived immortal keratinocyte cell lines compared to their mortal counterparts. The observations in this study argue strongly for an important role of the HA/FA DNA repair pathway in the development of OSCC.
Collapse
Affiliation(s)
- Stephen S. Prime
- Centre for Immunology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK;
| | - Piotr Darski
- Liverpool Head and Neck Centre, Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (P.D.); (K.D.H.)
| | - Keith D. Hunter
- Liverpool Head and Neck Centre, Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK; (P.D.); (K.D.H.)
| | - Nicola Cirillo
- Melbourne Dental School, University of Melbourne, 720 Swanson Street, Carlton, Melbourne, VIC 3053, Australia;
- School of Dentistry, University of Jordan, Amman 11942, Jordan
| | - E. Kenneth Parkinson
- Centre for Immunology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK;
| |
Collapse
|
15
|
Appin CL, Hong C, Suwala AK, Hilz S, Mathur R, Solomon DA, Smirnov IV, Stevers NO, Shai A, Wang A, Berger MS, Chang SM, Phillips JJ, Costello JF. Whole tumor analysis reveals early origin of the TERT promoter mutation and intercellular heterogeneity in TERT expression. Neuro Oncol 2024; 26:640-652. [PMID: 38141254 PMCID: PMC10995505 DOI: 10.1093/neuonc/noad231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Indexed: 12/25/2023] Open
Abstract
BACKGROUND The TERT promoter mutation (TPM) is acquired in most IDH-wildtype glioblastomas (GBM) and IDH-mutant oligodendrogliomas (OD) enabling tumor cell immortality. Previous studies on TPM clonality show conflicting results. This study was performed to determine whether TPM is clonal on a tumor-wide scale. METHODS We investigated TPM clonality in relation to presumed early events in 19 IDH-wildtype GBM and 10 IDH-mutant OD using 3-dimensional comprehensive tumor sampling. We performed Sanger sequencing on 264 tumor samples and deep amplicon sequencing on 187 tumor samples. We obtained tumor purity and copy number estimates from whole exome sequencing. TERT expression was assessed by RNA-seq and RNAscope. RESULTS We detected TPM in 100% of tumor samples with quantifiable tumor purity (219 samples). Variant allele frequencies (VAF) of TPM correlate positively with chromosome 10 loss in GBM (R = 0.85), IDH1 mutation in OD (R = 0.87), and with tumor purity (R = 0.91 for GBM; R = 0.90 for OD). In comparison, oncogene amplification was tumor-wide for MDM4- and most EGFR-amplified cases but heterogeneous for MYCN and PDGFRA, and strikingly high in low-purity samples. TPM VAF was moderately correlated with TERT expression (R = 0.52 for GBM; R = 0.65 for OD). TERT expression was detected in a subset of cells, solely in TPM-positive samples, including samples equivocal for tumor. CONCLUSIONS On a tumor-wide scale, TPM is among the earliest events in glioma evolution. Intercellular heterogeneity of TERT expression, however, suggests dynamic regulation during tumor growth. TERT expression may be a tumor cell-specific biomarker.
Collapse
Affiliation(s)
- Christina L Appin
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Chibo Hong
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Abigail K Suwala
- Department of Neurological Surgery, University of California, San Francisco, California, USA
- Department of Neuropathology, University of Heidelberg, Institute of Pathology, Heidelberg, Germany
| | - Stephanie Hilz
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Radhika Mathur
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - David A Solomon
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Ivan V Smirnov
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Nicholas O Stevers
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Anny Shai
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Albert Wang
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Susan M Chang
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Joanna J Phillips
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Joseph F Costello
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| |
Collapse
|
16
|
Martins B, Bister A, Dohmen RGJ, Gouveia MA, Hueber R, Melzener L, Messmer T, Papadopoulos J, Pimenta J, Raina D, Schaeken L, Shirley S, Bouchet BP, Flack JE. Advances and Challenges in Cell Biology for Cultured Meat. Annu Rev Anim Biosci 2024; 12:345-368. [PMID: 37963400 DOI: 10.1146/annurev-animal-021022-055132] [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: 11/16/2023]
Abstract
Cultured meat is an emerging biotechnology that aims to produce meat from animal cell culture, rather than from the raising and slaughtering of livestock, on environmental and animal welfare grounds. The detailed understanding and accurate manipulation of cell biology are critical to the design of cultured meat bioprocesses. Recent years have seen significant interest in this field, with numerous scientific and commercial breakthroughs. Nevertheless, these technologies remain at a nascent stage, and myriad challenges remain, spanning the entire bioprocess. From a cell biological perspective, these include the identification of suitable starting cell types, tuning of proliferation and differentiation conditions, and optimization of cell-biomaterial interactions to create nutritious, enticing foods. Here, we discuss the key advances and outstanding challenges in cultured meat, with a particular focus on cell biology, and argue that solving the remaining bottlenecks in a cost-effective, scalable fashion will require coordinated, concerted scientific efforts. Success will also require solutions to nonscientific challenges, including regulatory approval, consumer acceptance, and market feasibility. However, if these can be overcome, cultured meat technologies can revolutionize our approach to food.
Collapse
Affiliation(s)
- Beatriz Martins
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Arthur Bister
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Richard G J Dohmen
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Maria Ana Gouveia
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Rui Hueber
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Lea Melzener
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Tobias Messmer
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Joanna Papadopoulos
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Joana Pimenta
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Dhruv Raina
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Lieke Schaeken
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Sara Shirley
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| | - Benjamin P Bouchet
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands;
| | - Joshua E Flack
- Mosa Meat B.V., Maastricht, The Netherlands; , , , , , , , , , , , ,
| |
Collapse
|
17
|
Koca DS, Kolpakov V, Ihlow J, von Laffert M, Erb-Eigner K, Herbst H, Kriese K, Schweizer L, Bertelmann E. Prevalence of TERT Promoter Mutations in Orbital Solitary Fibrous Tumors. Curr Issues Mol Biol 2024; 46:1467-1484. [PMID: 38392213 PMCID: PMC10887834 DOI: 10.3390/cimb46020095] [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: 12/18/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
The orbital manifestation of a solitary fibrous tumor (SFT) is exceptionally rare and poses specific challenges in diagnosis and treatment. Its rather exceptional behavior among all SFTs comprises a high tendency towards local recurrence, but it rarely culminates in metastatic disease. This raises the question of prognostic factors in orbital SFTs (oSFTs). Telomerase reverse transcriptase (TERT)-promoter mutations have previously been linked to an unfavorable prognosis in SFTs of other locations. We analyzed the prevalence of TERT promoter mutations of SFTs in the orbital compartment. We performed a retrospective, descriptive clinico-histopathological analysis of nine cases of oSFTs between the years of 2017 and 2021. A TERT promoter mutation was present in one case, which was classified with intermediate metastatic risk. Local recurrence or progress occurred in six cases after primary resection; no distant metastases were reported. Multimodal imaging repeatedly showed particular morphologic patterns, including tubular vascular structures and ADC reduction. The prevalence of the TERT promoter mutation in oSFT was 11%, which is similar to the prevalence of extra-meningeal SFTs of the head and neck and lower than that in other extra-meningeal compartments. In the present study, the TERT promoter mutation in oSFT manifested in a case with an unfavorable prognosis, comprising aggressive local tumor growth, local recurrence, and eye loss.
Collapse
Affiliation(s)
- David Sinan Koca
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Vladimir Kolpakov
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Jana Ihlow
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Institute of Health Charité Clinician Scientist Program, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin Institute of Health Biomedical Innovation Academy, Anna-Louisa-Karsch-Str., 210178 Berlin, Germany
| | - Maximilian von Laffert
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Diagnostics, Institute of Pathology, Universitätsklinikum Leipzig AöR, Liebigstraße 26, 04103 Leipzig, Germany
| | - Katharina Erb-Eigner
- Department of Radiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Hermann Herbst
- Department of Pathology, Vivantes Hospital Neukölln, Vivantes Netzwerk für Gesundheit GmbH Berlin, Rudower Straße 48, 12351 Berlin, Germany
| | - Karen Kriese
- Department of Pathology, Vivantes Hospital Neukölln, Vivantes Netzwerk für Gesundheit GmbH Berlin, Rudower Straße 48, 12351 Berlin, Germany
| | - Leonille Schweizer
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Edinger Institute, Institute of Neurology, University of Frankfurt am Main, 60528 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt-Mainz, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
| | - Eckart Bertelmann
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
18
|
Liu M, Zhang Y, Jian Y, Gu L, Zhang D, Zhou H, Wang Y, Xu ZX. The regulations of telomerase reverse transcriptase (TERT) in cancer. Cell Death Dis 2024; 15:90. [PMID: 38278800 PMCID: PMC10817947 DOI: 10.1038/s41419-024-06454-7] [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: 09/22/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/28/2024]
Abstract
Abnormal activation of telomerase occurs in most cancer types, which facilitates escaping from cell senescence. As the key component of telomerase, telomerase reverse transcriptase (TERT) is regulated by various regulation pathways. TERT gene changing in its promoter and phosphorylation respectively leads to TERT ectopic expression at the transcription and protein levels. The co-interacting factors play an important role in the regulation of TERT in different cancer types. In this review, we focus on the regulators of TERT and these downstream functions in cancer regulation. Determining the specific regulatory mechanism will help to facilitate the development of a cancer treatment strategy that targets telomerase and cancer cell senescence. As the most important catalytic subunit component of telomerase, TERT is rapidly regulated by transcriptional factors and PTM-related activation. These changes directly influence TERT-related telomere maintenance by regulating telomerase activity in telomerase-positive cancer cells, telomerase assembly with telomere-binding proteins, and recruiting telomerase to the telomere. Besides, there are also non-canonical functions that are influenced by TERT, including the basic biological functions of cancer cells, such as proliferation, apoptosis, cell cycle regulation, initiating cell formation, EMT, and cell invasion. Other downstream effects are the results of the influence of transcriptional factors by TERT. Currently, some small molecular inhibitors of TERT and TERT vaccine are under research as a clinical therapeutic target. Purposeful work is in progress.
Collapse
Affiliation(s)
- Mingdi Liu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China
| | - Yuning Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China
| | - Yongping Jian
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China
| | - Liting Gu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China
| | - Dan Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China.
| | - Zhi-Xiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China.
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
19
|
Uziel O, Kanner AA, Beery E, Lev S, Lahav M, Horn‐Fichman S, Nof SH, Laviv Y, Yust‐Katz S, Amiel A, Shkara RA, Siddeeq M, Levy‐Barda A, Raanani P, Sela Y, Cohen Z, Siegal T. Is serum-derived exosomal hTERT transcript a marker of oncogenic activity in primary brain tumors? An exploratory study. Cancer Med 2024; 13:e6784. [PMID: 38155481 PMCID: PMC10823760 DOI: 10.1002/cam4.6784] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 08/01/2023] [Accepted: 10/03/2023] [Indexed: 12/30/2023] Open
Abstract
BACKGROUND In order to proliferate indefinitely, all tumors require a telomere maintenance mechanism. The expression of human telomerase reverse transcriptase (hTERT) enables telomere maintenance and provides cancer cells with limitless replicative potential. As such, it may serve as an attractive biomarker for oncogenic activity. This study explored whether a liquid biopsy that analyses blood derived exosomal hTERT transcript (e-hTERT-trans) may serve as such a biomarker in gliomas and meningiomas when compared to healthy controls. METHODS Exosomes were isolated from the pre-operative sera of patients' samples stored in the biobank of both Rabin and Sheba Medical Centers. The levels of e-hTERT-trans were measured in 81 healthy controls, 117 meningiomas, 17 low-grade gliomas, and 61 glioblastomas. Clinical parameters of the patients were collected retrospectively and compared to the levels of the e-hTERT-trans. RESULTS The upper normal limit of controls e-hTERT-trans was 1.85 relative quantitation (RQ). The rate of detection increased with rising tumor grade and correlated with tumor recurrence in meningiomas: mean RQ without recurrence (2.17 ± 11.7) versus with recurrence (3.59 ± 4.42; p = 0.002). In glioblastomas, preoperative measurements correlated with tumor volume and with the disease course on serial sampling. CONCLUSIONS We demonstrated for the first time that the expression of e-hTERT-trans transcript can be measured in the serum of primary brain tumors. This exosomal marker carries the potential to serve as a biomarker once used in conjunction with other clinical and radiological parameters. Future studies are required to investigate whether the sensitivity could be augmented and whether it can be implemented into routine patients care.
Collapse
Affiliation(s)
- Orit Uziel
- The Felsenstein Medical Research CenterPetah TikvaIsrael
- Institute of HematologyDavidoff Cancer Center, Rabin Medical CenterPetah TikvaIsrael
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
| | - Andrew A. Kanner
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
- Department of NeurosurgeryRabin Medical CenterPetah TikvaIsrael
| | - Einat Beery
- The Felsenstein Medical Research CenterPetah TikvaIsrael
| | - Sapir Lev
- Department of NeurosurgeryRabin Medical CenterPetah TikvaIsrael
| | - Meir Lahav
- The Felsenstein Medical Research CenterPetah TikvaIsrael
- Institute of HematologyDavidoff Cancer Center, Rabin Medical CenterPetah TikvaIsrael
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
| | - Suzana Horn‐Fichman
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
- Neuropathology, Department of PathologyRabin Medical CenterPetah TikvaIsrael
| | - Sagi Har Nof
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
- Department of NeurosurgeryRabin Medical CenterPetah TikvaIsrael
| | - Yuseph Laviv
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
- Department of NeurosurgeryRabin Medical CenterPetah TikvaIsrael
| | - S. Yust‐Katz
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
- Neurooncology UnitDavidoff Cancer Center, Rabin Medical CenterPetah TikvaIsrael
| | - Alexandra Amiel
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
- Neurooncology UnitDavidoff Cancer Center, Rabin Medical CenterPetah TikvaIsrael
| | | | - Mustafa Siddeeq
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
- Department of NeurosurgerySheba Medical CenterRamat‐GanIsrael
| | - Adva Levy‐Barda
- Biobank, Department of PathologyRabin Medical CenterPetah TikvaIsrael
| | - Pia Raanani
- The Felsenstein Medical Research CenterPetah TikvaIsrael
- Institute of HematologyDavidoff Cancer Center, Rabin Medical CenterPetah TikvaIsrael
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
| | - Yaron Sela
- The Center of Internet Psychology Reichman UniversityHerzliyaIsrael
| | - Zvi Cohen
- Sackler School of MedicineTel‐Aviv UniversityTel AvivIsrael
- Department of NeurosurgerySheba Medical CenterRamat‐GanIsrael
| | - Tali Siegal
- Neurooncology UnitDavidoff Cancer Center, Rabin Medical CenterPetah TikvaIsrael
- Hebrew University and Medical SchoolJerusalemIsrael
| |
Collapse
|
20
|
Wessman S, Fuentes BB, Severin-Karlsson J, Westbom-Fremer S, Nistér M, Kokaraki G, Petta TB, Haglund F, Carlson JW. FOXL2 Mutation Status in Sex Cord-stromal Tumors Cannot be Predicted by Morphology. Int J Gynecol Pathol 2024; 43:78-89. [PMID: 37255476 DOI: 10.1097/pgp.0000000000000953] [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: 06/01/2023]
Abstract
Granulosa-cell tumors (GCTs) are the most common type of malignant ovarian sex cord-stromal tumor (SCST). The histopathologic diagnosis of these tumors can be challenging. A recurrent somatic mutation of the forkhead box L2 (FOXL2) gene has been identified in adult GCT. In this retrospective single-center study of 44 SCST, a morphologic review together with analysis of FOXL2 C134W was evaluated in relation to tumor morphology. In addition, TERT promoter mutation testing was performed. Twelve of 36 cases got an altered diagnosis based on morphology alone. The overarching architectural growth pattern in 32/44 (72.7%) tumors was diffuse/solid with several tumors showing markedly heterogeneous architecture. In correlation to FOXL2 C134W mutation status, cytoplasmic color, and nuclear shape, differed between the FOXL2 C134W positive and FOXL2 C134 W negative groups, but these differences were not significant when comparing them separately. Nineteen of 44 cases underwent TERT promoter sequencing with a positive result in 3 cases; 2 adult GCTs and 1 cellular fibroma. Three patients developed a recurrence of which 2 were FOXL2 C134W positive adult GCTs and the third was an unclassified SCST. In conclusion, the morphologic and immunohistochemical diagnosis of different SCSTs is challenging and one cannot reliably identify FOXL2 mutation-positive tumors solely by morphologic features. Therefore, broad use of molecular analysis of the FOXL2 C134W mutation is suggested for SCSTs, and further studies are needed to evaluate the clinical outcome of these tumors as well as the diagnostic and prognostic implications of TERT promoter mutations.
Collapse
|
21
|
Urso C. Spitz Tumors and Melanoma in the Genomic Age: A Retrospective Look at Ackerman's Conundrum. Cancers (Basel) 2023; 15:5834. [PMID: 38136379 PMCID: PMC10741987 DOI: 10.3390/cancers15245834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
After 25 years, "Ackerman's conundrum", namely, the distinction of benign from malignant Spitz neoplasms, remains challenging. Genomic studies have shown that most Spitz tumors harbor tyrosine and serine/threonine kinase fusions, including ALK, ROS1, NTRK1, NTRK2, NTRK3, BRAF and MAP3K8, or some mutations, such as HRAS and MAP3K8. These chromosomal abnormalities act as drivers, initiating the oncogenetic process and conferring basic bio-morphological features. Most Spitz tumors show no additional genomic alterations or few ones; others harbor a variable number of mutations, capable of conferring characteristics related to clinical behavior, including CDKN2A deletion and TERT-p mutation. Since the accumulation of mutations is gradual and progressive, tumors appear to form a bio-morphologic spectrum, in which they show a progressive increase of clinical risk and histological atypia. In this context, a binary classification Spitz nevus-melanoma appears as no longer adequate, not corresponding to the real genomic substrate of lesions. A ternary classification Spitz nevus-Spitz melanocytoma-Spitz melanoma is more adherent to the real neoplastic pathway, but some cases with intermediate ambiguous features remain difficult to diagnose. A prognostic stratification of Spitz tumors, based on the morphologic and genomic characteristics, as a complement to the diagnosis, may contribute to better treatment plans for patients.
Collapse
Affiliation(s)
- Carmelo Urso
- Dermatopathology Study Center of Florence, I-50129 Florence, Italy
| |
Collapse
|
22
|
Zappe K, Pühringer K, Pflug S, Berger D, Weis S, Spiegl-Kreinecker S, Cichna-Markl M. Association of MGMT Promoter and Enhancer Methylation with Genetic Variants, Clinical Parameters, and Demographic Characteristics in Glioblastoma. Cancers (Basel) 2023; 15:5777. [PMID: 38136323 PMCID: PMC10742072 DOI: 10.3390/cancers15245777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
The response of glioblastoma (GBM) patients to the alkylating agent temozolomide (TMZ) vitally depends on the expression level of the repair protein O6-methylguanine-DNA methyltransferase (MGMT). Since MGMT is strongly regulated by promoter methylation, the methylation status of the MGMT promoter has emerged as a prognostic and predictive biomarker for GBM patients. By determining the methylation levels of the four enhancers located within or close to the MGMT gene, we recently found that enhancer methylation contributes to MGMT regulation. In this study, we investigated if methylation of the four enhancers is associated with SNP rs16906252, TERT promoter mutations C228T and C250T, TERT SNP rs2853669, proliferation index Ki-67, overall survival (OS), age, and sex of the patients. In general, associations with genetic variants, clinical parameters, and demographic characteristics were caused by a complex interplay of multiple CpGs in the MGMT promoter and of multiple CpGs in enhancer regions. The observed associations for intragenic enhancer 4, located in intron 2 of MGMT, differed from associations observed for the three intergenic enhancers. Some findings were restricted to subgroups of samples with either methylated or unmethylated MGMT promoters, underpinning the relevance of the MGMT promoter status in GBMs.
Collapse
Affiliation(s)
- Katja Zappe
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (K.Z.); (K.P.); (S.P.); (D.B.)
| | - Katharina Pühringer
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (K.Z.); (K.P.); (S.P.); (D.B.)
| | - Simon Pflug
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (K.Z.); (K.P.); (S.P.); (D.B.)
| | - Daniel Berger
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (K.Z.); (K.P.); (S.P.); (D.B.)
| | - Serge Weis
- Division of Neuropathology, Department of Pathology and Molecular Pathology, Kepler University Hospital GmbH, Johannes Kepler University, 4040 Linz, Austria;
| | - Sabine Spiegl-Kreinecker
- Department of Neurosurgery, Kepler University Hospital GmbH, Johannes Kepler University, 4040 Linz, Austria;
| | - Margit Cichna-Markl
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (K.Z.); (K.P.); (S.P.); (D.B.)
| |
Collapse
|
23
|
Praiss AM, Marra A, Zhou Q, Rios-Doria E, Momeni-Boroujeni A, Iasonos A, Selenica P, Brown DN, Aghajanian C, Abu-Rustum NR, Ellenson LH, Weigelt B. TERT promoter mutations and gene amplification in endometrial cancer. Gynecol Oncol 2023; 179:16-23. [PMID: 37890416 PMCID: PMC10841990 DOI: 10.1016/j.ygyno.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/06/2023] [Accepted: 10/14/2023] [Indexed: 10/29/2023]
Abstract
OBJECTIVE To assess the clinicopathologic, molecular profiles, and survival outcomes of patients with endometrial carcinomas (ECs) harboring telomerase reverse transcriptase (TERT) hotspot mutations or gene amplification. METHODS ECs harboring somatic TERT promoter hotspot mutations or gene amplification (TERT-altered) were identified from 1944 ECs that underwent clinical tumor-normal sequencing from 08/2016-12/2021. Clinicopathologic variables, somatic mutation profiles, and survival outcomes of TERT-alt and TERT-wild-type EC were assessed. RESULTS We identified 66 TERT-altered ECs (43 TERT-mutated and 23 TERT-amplified), representing 3% of the unselected ECs across histologic subtypes. Most TERT-altered ECs were of copy number (CN)-high/TP53abn molecular subtype (n = 40, 60%), followed by microsatellite-unstable (MSI-H) or CN-low/no specific molecular profile (NSMP)(n = 13, 20% each). TERT-amplified and TERT-mutated ECs were molecularly distinct, with TERT-amplified ECs being more genomically instable and more frequently harboring TP53 and PPP2R1A alterations (q < 0.1). Compared to TERT-wild-type ECs, TERT-altered ECs were more commonly of CN-H/TP53abn molecular subtype (31% vs 57%, p = 0.001), serous histology (10% vs 26%, p = 0.004), and were significantly enriched for TP53, CDKN2A/B, and DROSHA somatic genetic alterations (q < 0.1). Median progression-free survival was 18.7 months (95% CI 11.8-not estimable [NE]) for patients with TERT-altered EC and 80.9 months (65.8-NE) for patients with TERT-wild-type EC (HR 0.33, 95% CI 0.21-0.51, p < 0.001). Similarly, median overall survival was 46.7 months (95% CI 30-NE) for TERT-altered EC patients and not reached for TERT-wild-type EC patients (HR 0.24, 95% CI 0.13-0.44, p < 0.001). CONCLUSION TERT-altered ECs, although rare, are enriched for CN-high/TP53abn tumors, TP53, CDKN2A/B and DROSHA somatic mutations, and independently predict worse survival outcomes.
Collapse
Affiliation(s)
- Aaron M Praiss
- Gynecology Service, Departments of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Antonio Marra
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Qin Zhou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eric Rios-Doria
- Gynecology Service, Departments of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amir Momeni-Boroujeni
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexia Iasonos
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pier Selenica
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David N Brown
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Carol Aghajanian
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem R Abu-Rustum
- Gynecology Service, Departments of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lora H Ellenson
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
24
|
Chua BH, Zaal Anuar N, Ferry L, Domrane C, Wittek A, Mukundan VT, Jha S, Butter F, Tenen DG, Defossez PA, Kappei D. E4F1 and ZNF148 are transcriptional activators of the -57A > C and wild-type TERT promoter. Genome Res 2023; 33:1893-1905. [PMID: 37918959 PMCID: PMC10760450 DOI: 10.1101/gr.277724.123] [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: 01/20/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
Point mutations within the TERT promoter are the most recurrent somatic noncoding mutations identified across different cancer types, including glioblastoma, melanoma, hepatocellular carcinoma, and bladder cancer. They are most abundant at -146C > T and -124C > T, and rarer at -57A > C, with the latter originally described as a familial case, but subsequently shown also to occur somatically. All three mutations create de novo E26-specific (ETS) binding sites and result in activation of the TERT gene, allowing cancer cells to achieve replicative immortality. Here, we used a systematic proteomics screen to identify transcription factors preferentially binding to the -146C > T, -124C > T, and -57A > C mutations. Although we confirmed binding of multiple ETS factors to the mutant -146C > T and -124C > T sequences, we identified E4F1 as a -57A > C-specific binder and ZNF148 as a TERT wild-type (WT) promoter binder that showed reduced interaction with the -124C > T allele. Both proteins are activating transcription factors that bind specifically to the -57A > C and WT (at position 124) TERT promoter sequence in corresponding cell lines, and up-regulate TERT transcription and telomerase activity. Our work describes new regulators of TERT gene expression with possible roles in cancer.
Collapse
Affiliation(s)
- Boon Haow Chua
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117596 Singapore
| | - Nurkaiyisah Zaal Anuar
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore
| | - Laure Ferry
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, F-75013 Paris, France
| | - Cecilia Domrane
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, F-75013 Paris, France
| | - Anna Wittek
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore
| | - Vineeth T Mukundan
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore
| | - Sudhakar Jha
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117596 Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma 74078, USA
| | - Falk Butter
- Institute of Molecular Biology (IMB), 55128 Mainz, Germany
- Institute of Molecular Virology and Cell Biology (IMVZ), Friedrich Loeffler Institute, 17493 Greifswald, Germany
| | - Daniel G Tenen
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore
- Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | - Dennis Kappei
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore;
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117596 Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore
| |
Collapse
|
25
|
Udutha S, Taglang C, Batsios G, Gillespie AM, Tran M, Ronen SM, Ten Hoeve J, Graeber TG, Viswanath P. Telomerase reverse transcriptase induces targetable alterations in glutathione and nucleotide biosynthesis in glioblastomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.566937. [PMID: 38014170 PMCID: PMC10680720 DOI: 10.1101/2023.11.14.566937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Telomerase reverse transcriptase (TERT) is essential for glioblastoma (GBM) proliferation. Delineating metabolic vulnerabilities induced by TERT can lead to novel GBM therapies. We previously showed that TERT upregulates glutathione (GSH) pool size in GBMs. Here, we show that TERT acts via the FOXO1 transcription factor to upregulate expression of the catalytic subunit of glutamate-cysteine ligase (GCLC), the rate-limiting enzyme of de novo GSH synthesis. Inhibiting GCLC using siRNA or buthionine sulfoximine (BSO) reduces synthesis of 13 C-GSH from [U- 13 C]-glutamine and inhibits clonogenicity. However, GCLC inhibition does not induce cell death, an effect that is associated with elevated [U- 13 C]-glutamine metabolism to glutamate and pyrimidine nucleotide biosynthesis. Mechanistically, GCLC inhibition activates MYC and leads to compensatory upregulation of two key glutamine-utilizing enzymes i.e., glutaminase (GLS), which generates glutamate from glutamine, and CAD (carbamoyl-phosphate synthetase 2, aspartate transcarbamoylase, dihydroorotatase), the enzyme that converts glutamine to the pyrimidine nucleotide precursor dihydroorotate. We then examined the therapeutic potential of inhibiting GLS and CAD in combination with GCLC. 6-diazo-5-oxy-L-norleucin (DON) is a potent inhibitor of glutamine-utilizing enzymes including GLS and CAD. The combination of BSO and DON suppresses GSH and pyrimidine nucleotide biosynthesis and is synergistically lethal in GBM cells. Importantly, in vivo stable isotope tracing indicates that combined treatment with JHU-083 (a brain-penetrant prodrug of DON) and BSO abrogates synthesis of GSH and pyrimidine nucleotides from [U- 13 C]-glutamine and induces tumor shrinkage in mice bearing intracranial GBM xenografts. Collectively, our studies exploit a mechanistic understanding of TERT biology to identify synthetically lethal metabolic vulnerabilities in GBMs. SIGNIFICANCE Using in vivo stable isotope tracing, metabolomics, and loss-of-function studies, we demonstrate that TERT expression is associated with metabolic alterations that can be synergistically targeted for therapy in glioblastomas.
Collapse
|
26
|
Dadmanesh F, Li X, Leong M, Maluf H, Balzer B. The Genetic Landscape of Fibroepithelial Lesions of the Breast. Adv Anat Pathol 2023; 30:415-420. [PMID: 37539688 DOI: 10.1097/pap.0000000000000407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Fibroepithelial lesions of the breast encompass a broad spectrum of lesions from fibroadenomas and their variants to phyllodes tumors, including their clinical range of benign, borderline, and malignant. Classification of this spectrum of neoplasms has historically and currently been based purely on morphology, although the nomenclature has shifted over the years largely due to the significant histologic overlap that exists primarily within the cellular fibroadenomas to borderline malignant phyllodes tumor categories. A review of the current diagnostic challenge, proposed ancillary studied and their value in prognostic significance, is provided. This article highlights the most recent molecular and genetic findings as well as the limitations of the studies, in the context of practical and available applications for the diagnostician and managerial implications for the clinician.
Collapse
Affiliation(s)
- Farnaz Dadmanesh
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | | | | | | |
Collapse
|
27
|
Yan MP, Wee CE, Yen KP, Stevens A, Wai LK. G-quadruplex ligands as therapeutic agents against cancer, neurological disorders and viral infections. Future Med Chem 2023; 15:1987-2009. [PMID: 37933551 DOI: 10.4155/fmc-2023-0202] [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: 11/08/2023] Open
Abstract
G-quadruplexes (G4s) within the human genome have undergone extensive molecular investigation, with a strong focus on telomeres, gene promoters and repetitive regulatory sequences. G4s play central roles in regulating essential biological processes, including telomere maintenance, replication, transcription and translation. Targeting these molecular processes with G4-binding ligands holds substantial therapeutic potential in anticancer treatments and has also shown promise in treating neurological, skeletal and muscular disorders. The presence of G4s in bacterial and viral genomes also suggests that G4-binding ligands could be a critical tool in fighting infections. This review provides an overview of the progress and applications of G4-binding ligands, their proposed mechanisms of action, challenges faced and prospects for their utilization in anticancer treatments, neurological disorders and antiviral activities.
Collapse
Affiliation(s)
- Mock Phooi Yan
- Centre for Drug & Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| | - Chua Eng Wee
- Centre for Drug & Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| | - Khor Poh Yen
- Faculty Pharmacy & Health Sciences, Universiti Kuala Lumpur, Royal College of Medicine Perak, 3, Jalan Greentown, Ipoh, Perak, 30450, Malaysia
| | - Aaron Stevens
- Department of Pathology & Molecular Medicine, University of Otago, Wellington, 6021, New Zealand
| | - Lam Kok Wai
- Centre for Drug & Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| |
Collapse
|
28
|
Sergi MC, Filoni E, Triggiano G, Cazzato G, Internò V, Porta C, Tucci M. Mucosal Melanoma: Epidemiology, Clinical Features, and Treatment. Curr Oncol Rep 2023; 25:1247-1258. [PMID: 37773078 PMCID: PMC10640506 DOI: 10.1007/s11912-023-01453-x] [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] [Accepted: 08/21/2023] [Indexed: 09/30/2023]
Abstract
PURPOSE OF REVIEW Summarize the writings published in the last years on the management and novel therapies of mucosal melanoma (MM). RECENT FINDINGS New research has demonstrated a difference between MM and cutaneous melanoma (CM) in their genomic and molecular landscapes, explaining the response's heterogeneity. Immunotherapy and targeted therapy have limited benefit, but novel therapies are rapidly expanding. MM is aggressive cancer occurring in gastrointestinal, respiratory, or urogenital mucosa; whose incidence is greater in the Asian population. The etiology and pathogenesis remain unclear since UV exposure is not a proven risk factor as in cutaneous melanoma. In contrast to CM, lesions on the mucosal surface are less likely to be recognized early; therefore, the disease is diagnosed in an advanced stage. Clinical manifestations, such as bleeding or pain, can help to detect this tumor, although the prognosis remains unfavorable with an overall 5-year survival rate of less than 20%. The mutational landscape of MM includes mutations of BRAF and NRAS, as well as mutations in the c-KIT/CD117 gene (in 50% of patients), thus limiting therapeutic interventions to immunotherapy. However, clinical studies show less responsiveness to immunotherapy compared to CM, therefore novel therapeutic strategies targeting new molecules are needed to improve the survival of patients with MM.
Collapse
Affiliation(s)
- Maria Chiara Sergi
- Department of Interdisciplinary Medicine, Oncology Unit, University of Bari "Aldo Moro", P.za Giulio Cesare, 11, 70124, Bari, Italy.
| | - Elisabetta Filoni
- Department of Interdisciplinary Medicine, Oncology Unit, University of Bari "Aldo Moro", P.za Giulio Cesare, 11, 70124, Bari, Italy
| | - Giacomo Triggiano
- Department of Interdisciplinary Medicine, Oncology Unit, University of Bari "Aldo Moro", P.za Giulio Cesare, 11, 70124, Bari, Italy
| | - Gerardo Cazzato
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari "Aldo Moro", 70124, Bari, Italy
| | | | - Camillo Porta
- Department of Interdisciplinary Medicine, Oncology Unit, University of Bari "Aldo Moro", P.za Giulio Cesare, 11, 70124, Bari, Italy
| | - Marco Tucci
- Department of Interdisciplinary Medicine, Oncology Unit, University of Bari "Aldo Moro", P.za Giulio Cesare, 11, 70124, Bari, Italy
| |
Collapse
|
29
|
Burioli EAV, Hammel M, Vignal E, Vidal-Dupiol J, Mitta G, Thomas F, Bierne N, Destoumieux-Garzón D, Charrière GM. Transcriptomics of mussel transmissible cancer MtrBTN2 suggests accumulation of multiple cancer traits and oncogenic pathways shared among bilaterians. Open Biol 2023; 13:230259. [PMID: 37816387 PMCID: PMC10564563 DOI: 10.1098/rsob.230259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/12/2023] [Indexed: 10/12/2023] Open
Abstract
Transmissible cancer cell lines are rare biological entities giving rise to diseases at the crossroads of cancer and parasitic diseases. These malignant cells have acquired the amazing capacity to spread from host to host. They have been described only in dogs, Tasmanian devils and marine bivalves. The Mytilus trossulus bivalve transmissible neoplasia 2 (MtrBTN2) lineage has even acquired the capacity to spread inter-specifically between marine mussels of the Mytilus edulis complex worldwide. To identify the oncogenic processes underpinning the biology of these atypical cancers we performed transcriptomics of MtrBTN2 cells. Differential expression, enrichment, protein-protein interaction network, and targeted analyses were used. Overall, our results suggest the accumulation of multiple cancerous traits that may be linked to the long-term evolution of MtrBTN2. We also highlight that vertebrate and lophotrochozoan cancers could share a large panel of common drivers, which supports the hypothesis of an ancient origin of oncogenic processes in bilaterians.
Collapse
Affiliation(s)
- E A V Burioli
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| | - M Hammel
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
- ISEM, Univ Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - E Vignal
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| | - J Vidal-Dupiol
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| | - G Mitta
- IFREMER, UMR 241 Écosystèmes Insulaires Océaniens, Labex Corail, Centre Ifremer du Pacifique, Tahiti, Polynésie française
| | - F Thomas
- CREEC/CANECEV (CREES), MIVEGEC, Unité Mixte de Recherches, IRD 224-CNRS 5290-Université de Montpellier, Montpellier, France
| | - N Bierne
- ISEM, Univ Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - D Destoumieux-Garzón
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| | - G M Charrière
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| |
Collapse
|
30
|
Yu W, Mei Y, Lu Z, Zhou L, Jia F, Chen S, Wang Z. The causal relationship between genetically determined telomere length and meningiomas risk. Front Neurol 2023; 14:1178404. [PMID: 37693759 PMCID: PMC10484632 DOI: 10.3389/fneur.2023.1178404] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Background Studies have shown that longer leukocyte telomere length (LTL) is significantly associated with increased risk of meningioma. However, there is limited evidence concerning the causal association of LTL with benign and malignant meningiomas or with the location of benign tumors. Methods We used three LTL datasets from different sources, designated by name and sample size as LTL-78592, LTL-9190, and LTL-472174. The linkage disequilibrium score (LDSC) was used to explore the association between LTL and meningioma. We utilized two-sample bidirectional Mendelian randomization (TSMR) to evaluate whether LTL is causally related to meningioma risk. We adjusted for confounders by conducting multivariable Mendelian randomization (MVMR). Results In the LTL-78592, longer LTL was significantly associated with increased risk of malignant [odds ratio (OR) = 5.14, p = 1.04 × 10-5], benign (OR = 4.81, p < 0.05), benign cerebral (OR = 5.36, p < 0.05), and benign unspecified meningioma (OR = 8.26, p < 0.05). The same results were obtained for the LTL-9190. In the LTL-472174, longer LTL was significantly associated with increased risk of malignant (OR = 4.94, p < 0.05), benign (OR = 3.14, p < 0.05), and benign cerebral meningioma (OR = 3.59, p < 0.05). Similar results were obtained in the MVMR. In contrast, only benign cerebral meningioma displayed a possible association with longer LTL (OR = 1.01, p < 0.05). No heterogeneity or horizontal pleiotropy was detected. Conclusion In brief, genetically predicted longer LTL may increase the risk of benign, malignant, and benign cerebral meningiomas, regardless of the LTL measure, in European populations.
Collapse
Affiliation(s)
- Weijie Yu
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen Key Laboratory of Brain Center, Xiamen, China
| | - Yunyun Mei
- Department of Neurosurgery, Fudan University Shanghai Cancer Center (Xiamen Hospital), Xiamen, China
| | - Zhenwei Lu
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen Key Laboratory of Brain Center, Xiamen, China
| | - Liwei Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen Key Laboratory of Brain Center, Xiamen, China
| | - Fang Jia
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen Key Laboratory of Brain Center, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, China
| | - Sifang Chen
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen Key Laboratory of Brain Center, Xiamen, China
| | - Zhanxiang Wang
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen Key Laboratory of Brain Center, Xiamen, China
| |
Collapse
|
31
|
Wu X, Huang G, Li W, Chen Y. Ethnicity-specific association between TERT rs2736100 (A > C) polymorphism and lung cancer risk: a comprehensive meta-analysis. Sci Rep 2023; 13:13271. [PMID: 37582820 PMCID: PMC10427644 DOI: 10.1038/s41598-023-40504-y] [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: 01/11/2023] [Accepted: 08/11/2023] [Indexed: 08/17/2023] Open
Abstract
The rs2736100 (A > C) polymorphism of the second intron of Telomerase reverse transcriptase (TERT) has been confirmed to be closely associated with the risk of Lung cancer (LC), but there is still no unified conclusion on the results of its association with LC. This study included Genome-wide association studies (GWAS) and case-control studies reported so far on this association between TERT rs2736100 polymorphism and LC to clarify such a correlation with LC and the differences in it between different ethnicities and different types of LC. Relevant literatures published before May 7, 2022 on 'TERT rs2736100 polymorphism and LC susceptibility' in PubMed, EMbase, CENTRAL, MEDLINE databases were searched through the Internet, and data were extracted. Statistical analysis of data was performed in Revman5.3 software, including drawing forest diagrams, drawing funnel diagrams and so on. Sensitivity and publication bias analysis were performed in Stata 12.0 software. The C allele of TERT rs2736100 was associated with the risk of LC (Overall population: [OR] = 1.21, 95%CI [1.17, 1.25]; Caucasians: [OR] = 1.11, 95%CI [1.06, 1.17]; Asians: [OR] = 1.26, 95%CI [1.21, 1.30]), and Asians had a higher risk of LC than Caucasians (C vs. A: Caucasians: [OR] = 1.11 /Asians: [OR]) = 1.26). The other gene models also showed similar results. The results of stratified analysis of LC patients showed that the C allele was associated with the risk of Non-small-cell lung carcinoma (NSCLC) and Lung adenocarcinoma (LUAD), and the risk of NSCLC and LUAD in Asians was higher than that in Caucasians. The C allele was associated with the risk of Lung squamous cell carcinoma (LUSC) and Small cell lung carcinoma(SCLC) in Asians but not in Caucasians. NSCLC patients ([OR] = 1.27) had a stronger correlation than SCLC patients ([OR] = 1.03), and LUAD patients ([OR] = 1.32) had a stronger correlation than LUSC patients ([OR] = 1.09).In addition, the C allele of TERT rs2736100 was associated with the risk of LC, NSCLC and LUAD in both smoking groups and non-smoking groups, and the risk of LC in non-smokers of different ethnic groups was higher than that in smokers. In the Asians, non-smoking women were more at risk of developing LUAD. The C allele of TERT rs2736100 is a risk factor for LC, NSCLC, and LUAD in different ethnic groups, and the Asian population is at a more common risk. The C allele is a risk factor for LUSC and SCLC in Asians but not in Caucasians. And smoking is not the most critical factor that causes variation in TERT rs2736100 to increase the risk of most LC (NSCLC, LUAD). Therefore, LC is a multi-etiological disease caused by a combination of genetic, environmental and lifestyle factors.
Collapse
Affiliation(s)
- Xiaozheng Wu
- Department of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 510025, China
| | - Gao Huang
- Department of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 510025, China
| | - Wen Li
- Department of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 510025, China
| | - Yunzhi Chen
- Department of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 510025, China.
| |
Collapse
|
32
|
Bertol BC, Massaro JD, Debortoli G, Santos ALP, de Araújo JNG, Giorgenon TMV, Costa e Silva M, de Figueiredo-Feitosa NL, Collares CVA, de Freitas LCC, Soares EG, Neder L, Silbiger VN, Calado RT, Maciel LMZ, Donadi EA. BRAF, TERT and HLA-G Status in the Papillary Thyroid Carcinoma: A Clinicopathological Association Study. Int J Mol Sci 2023; 24:12459. [PMID: 37569841 PMCID: PMC10419559 DOI: 10.3390/ijms241512459] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
As BRAF, TERT, HLA-G, and microRNAs have been individually associated with papillary thyroid carcinoma (PTC), we aimed to evaluate the individual and collaborative role of these markers in PTC in the same patient cohort. HLA-G and BRAF tumor expression was evaluated by immunohistochemistry. Using molecular methods, BRAFV600E and TERT promoter mutations were evaluated in thyroid fine needle aspirates. MicroRNA tumor profiling was investigated using massively parallel sequencing. We observed strong HLA-G (67.96%) while BRAF (62.43%) staining was observed in PTC specimens. BRAF overexpression was associated with poor response to therapy. The BRAFV600E (52.9%) and TERTC228T (13%) mutations were associated with extrathyroidal extension, advanced-age, and advanced-stage cancer. The TERT rs2853669 CC+TC genotypes (38%) were overrepresented in metastatic tumors. Nine modulated microRNAs targeting the BRAF, TERT, and/or HLA-G genes were observed in PTC and involved with cancer-related signaling pathways. The markers were individually associated with PTC features, emphasizing the synergistic effect of BRAFV600E and TERTC228T; however, their collaborative role on PTC outcome was not fully demonstrated. The differentially expressed miRNAs targeting the BRAF and/or HLA-G genes may explain their increased expression in the tumor milieu.
Collapse
Affiliation(s)
- Bruna C. Bertol
- Postgraduate Program of Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Juliana D. Massaro
- Division of Clinical Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil; (J.D.M.); (M.C.e.S.); (C.V.A.C.)
| | - Guilherme Debortoli
- Department of Anthropology, University of Toronto, Mississauga, ON L5L 1C6, Canada;
| | - André L. P. Santos
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil; (A.L.P.S.); (R.T.C.)
| | - Jéssica N. G. de Araújo
- Department of Clinical Analysis and Toxicology, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (J.N.G.d.A.); (V.N.S.)
| | - Tatiana M. V. Giorgenon
- Division of Endocrinology and Metabolism, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil; (T.M.V.G.); (N.L.d.F.-F.); (L.M.Z.M.)
| | - Matheus Costa e Silva
- Division of Clinical Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil; (J.D.M.); (M.C.e.S.); (C.V.A.C.)
| | - Nathalie L. de Figueiredo-Feitosa
- Division of Endocrinology and Metabolism, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil; (T.M.V.G.); (N.L.d.F.-F.); (L.M.Z.M.)
| | - Cristhianna V. A. Collares
- Division of Clinical Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil; (J.D.M.); (M.C.e.S.); (C.V.A.C.)
| | - Luiz Carlos C. de Freitas
- Department of Ophthalmology, Otorhinolaryngology and Head and Neck Surgery, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil;
| | - Edson G. Soares
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil; (E.G.S.); (L.N.)
| | - Luciano Neder
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil; (E.G.S.); (L.N.)
| | - Vivian N. Silbiger
- Department of Clinical Analysis and Toxicology, Federal University of Rio Grande do Norte, Natal 59012-570, Brazil; (J.N.G.d.A.); (V.N.S.)
| | - Rodrigo T. Calado
- Department of Medical Imaging, Hematology, and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil; (A.L.P.S.); (R.T.C.)
| | - Léa M. Z. Maciel
- Division of Endocrinology and Metabolism, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil; (T.M.V.G.); (N.L.d.F.-F.); (L.M.Z.M.)
| | - Eduardo A. Donadi
- Postgraduate Program of Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
- Division of Clinical Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil; (J.D.M.); (M.C.e.S.); (C.V.A.C.)
| |
Collapse
|
33
|
Kiewa J, Mortlock S, Meltzer-Brody S, Middeldorp C, Wray NR, Byrne EM. A Common Genetic Factor Underlies Genetic Risk for Gynaecological and Reproductive Disorders and Is Correlated with Risk to Depression. Neuroendocrinology 2023; 113:1059-1075. [PMID: 37544299 PMCID: PMC10614513 DOI: 10.1159/000533413] [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: 05/29/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023]
Abstract
INTRODUCTION Sex steroid hormone fluctuations may underlie both reproductive disorders and sex differences in lifetime depression prevalence. Previous studies report high comorbidity among reproductive disorders and between reproductive disorders and depression. This study sought to assess the multivariate genetic architecture of reproductive disorders and their loading onto a common genetic factor and investigated whether this latent factor shares a common genetic architecture with female depression, including perinatal depression (PND). METHOD Using UK Biobank and FinnGen data, genome-wide association meta-analyses were conducted for nine reproductive disorders, and genetic correlation between disorders was estimated. Genomic Structural Equation Modelling identified a latent genetic factor underlying disorders, accounting for their significant genetic correlations. SNPs significantly associated with both latent factor and depression were identified. RESULTS Excellent model fit existed between a latent factor underlying five reproductive disorders (χ2 (5) = 6.4; AIC = 26.4; CFI = 1.00; SRMR = 0.03) with high standardised loadings for menorrhagia (0.96, SE = 0.05); ovarian cysts (0.94, SE = 0.05); endometriosis (0.83, SE = 0.05); menopausal symptoms (0.77, SE = 0.10); and uterine fibroids (0.65, SE = 0.05). This latent factor was genetically correlated with PND (rG = 0.37, SE = 0.15, p = 1.4e-03), depression in females only (rG = 0.48, SE = 0.06, p = 7.2e-11), and depression in both males and females (MD) (rG = 0.35, SE = 0.03, p = 1.8e-30), with its top locus associated with FSHB/ARL14EP (rs11031006; p = 9.1e-33). SNPs intronic to ESR1, significantly associated with the latent factor, were also associated with PND, female depression, and MD. CONCLUSION A common genetic factor, correlated with depression, underlies risk of reproductive disorders, with implications for aetiology and treatment. Genetic variation in ESR1 is associated with reproductive disorders and depression, highlighting the importance of oestrogen signalling for both reproductive and mental health.
Collapse
Affiliation(s)
- Jacqueline Kiewa
- Child Health Research Centre, University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Sally Mortlock
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | | | - Christel Middeldorp
- Child Health Research Centre, University of Queensland, Brisbane, QLD, Australia
- Child and Youth Mental Health Service, Children’s Health Queensland Hospital and Health Service, Brisbane, QLD, Australia
- Department of Child and Adolescent Psychiatry and Psychology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Youth and Family and Department of Research, Arkin Institute for Mental Health, Amsterdam, The Netherlands
- Levvel, Academic Centre for Child and Adolescent Psychiatry, Amsterdam, The Netherlands
| | - Naomi R. Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Enda M. Byrne
- Child Health Research Centre, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
34
|
Li L, Fu Y, Zhang Y, Mao Y, Huang D, Yi X, Wang J, Tan Z, Jiang M, Chen BT. Magnetic resonance imaging findings of intracranial extraventricular ependymoma: A retrospective multi-center cohort study of 114 cases. Cancer Med 2023; 12:16195-16206. [PMID: 37376821 PMCID: PMC10469843 DOI: 10.1002/cam4.6279] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Intracranial extraventricular ependymoma (IEE) is an ependymoma located in the brain parenchyma outside the ventricles. IEE has overlapping clinical and imaging characteristics with glioblastoma multiforme (GBM) but different treatment strategy and prognosis. Therefore, an accurate preoperative diagnosis is necessary for optimizing therapy for IEE. METHODS A retrospective multicenter cohort of IEE and GBM was identified. MR imaging characteristics assessed with the Visually Accessible Rembrandt Images (VASARI) feature set and clinicopathological findings were recorded. Independent predictors for IEE were identified using multivariate logistic regression, which was used to construct a diagnostic score for differentiating IEE from GBM. RESULTS Compared to GBM, IEE tended to occur in younger patients. Multivariate logistic regression analysis identified seven independent predictors for IEE. Among them, 3 predictors including tumor necrosis rate (F7), age, and tumor-enhancing margin thickness (F11), demonstrated higher diagnostic performance with an Area Under Curve (AUC) of more than 70% in distinguishing IEE from GBM. The AUC was 0.85, 0.78, and 0.70, with sensitivity of 92.98%, 72.81%, and 96.49%, and specificity of 65.50%, 73.64%, and 43.41%, for F7, age, and F11, respectively. CONCLUSION We identified specific MR imaging features such as tumor necrosis and thickness of enhancing tumor margins that could help to differentiate IEE from GBM. Our study results should be helpful to assist in diagnosis and clinical management of this rare brain tumor.
Collapse
Affiliation(s)
- Liyan Li
- Department of RadiologyFirst Affiliated Hospital of Guangxi Medical UniversityNanningP. R. China
| | - Yan Fu
- Department of RadiologyXiangya Hospital, Central South UniversityChangshaP. R. China
| | - Yinping Zhang
- Department of RadiologyXiangya Hospital, Central South UniversityChangshaP. R. China
| | - Yipu Mao
- Department of RadiologyNanning First People's HospitalNanningP. R. China
| | - Deyou Huang
- Department of RadiologyAffiliated Hospital of Youjiang Medical University for NationalitiesBaiseP. R. China
| | - Xiaoping Yi
- Department of RadiologyXiangya Hospital, Central South UniversityChangshaP. R. China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyXiangya HospitalChangshaP. R. China
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaP. R. China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya HospitalCentral South UniversityChangshaP. R. China
- Hunan Engineering Research Center of Skin Health and DiseaseXiangya Hospital, Central South UniversityChangshaP. R. China
- Department of DermatologyXiangya Hospital, Central South UniversityChangshaP. R. China
| | - Jing Wang
- Department of NeurologyXiangya Hospital, Central South UniversityChangshaP. R. China
| | - Zeming Tan
- Department of NeurosurgeryXiangya Hospital, Central South UniversityChangshaP. R. China
| | - Muliang Jiang
- Department of RadiologyFirst Affiliated Hospital of Guangxi Medical UniversityNanningP. R. China
| | - Bihong T. Chen
- Department of Diagnostic RadiologyCity of Hope National Medical CenterDuarteCaliforniaUSA
| |
Collapse
|
35
|
Gregório C, Thakur S, Camara Rivero R, Márcia Dos Santos Machado S, Cuenin C, Carreira C, White V, Cree IA, Vukojevic K, Glavina Durdov M, Bersch Osvaldt A, Ashton-Prolla P, Herceg Z, Talukdar FR. Telomere length assessment and molecular characterization of TERT gene promoter in periampullary carcinomas. Gene 2023; 873:147460. [PMID: 37150235 DOI: 10.1016/j.gene.2023.147460] [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: 12/28/2022] [Revised: 04/14/2023] [Accepted: 05/01/2023] [Indexed: 05/09/2023]
Abstract
Genetic and epigenetic alterations of the telomere maintenance machinery like telomere length and telomerase reverse transcriptase (encoded by TERT gene) are reported in several human malignancies. However, there is limited knowledge on the status of the telomere machinery in periampullary carcinomas (PAC) which are rare and heterogeneous groups of cancers arising from different anatomic sites around the ampulla of Vater. In the current study, we investigated the relative telomere length (RTL) and the most frequent genetic and epigenetic alterations in the TERT promoter in PAC and compared it with tumor-adjacent nonpathological duodenum (NDu). We found shorter RTLs (1.27 vs 1.33, P = 0.01) and lower TERT protein expression (p = 0.04) in PAC tissues as compared to the NDu. Although we did not find any mutation at two reactivating hotspot mutation sites of the TERT promoter, we detected polymorphism in 45% (9/20) of the cases at rs2853669 (T > C). Also, we found a hypermethylated region in the TERT promoter of PACs consisting of four CpGs (cg10896616 with Δβ 7%; cg02545192 with Δβ 9%; cg03323598 with Δβ 19%; and cg07285213 with Δβ 15%). In conclusion, we identified shorter telomeres with DNA hypermethylation in the TERT promoter region and lower TERT protein expression in PAC tissues. These results could be used further to investigate molecular pathology and develop theranostics for PAC.
Collapse
Affiliation(s)
- Cleandra Gregório
- Departamento de Genética, Programa de Pós-graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Laboratório de Medicina Genômica, Centro de Pesquisa Experimental - Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Shefali Thakur
- International Agency for Research on Cancer, Lyon, France; Faculty of Science, Charles University, Prague, Czech Republic
| | - Raquel Camara Rivero
- Departamento de Patologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Serviço de Patologia- Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Simone Márcia Dos Santos Machado
- Grupo de Vias Biliares e Pâncreas - Cirurgia do Aparelho Digestivo, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cyrille Cuenin
- International Agency for Research on Cancer, Lyon, France
| | | | - Valerie White
- International Agency for Research on Cancer, Lyon, France
| | - Ian A Cree
- International Agency for Research on Cancer, Lyon, France
| | - Katarina Vukojevic
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Split, Croatia
| | | | - Alessandro Bersch Osvaldt
- Serviço de Patologia- Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-graduação em Medicina: Ciências Cirúrgicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Patricia Ashton-Prolla
- Departamento de Genética, Programa de Pós-graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Laboratório de Medicina Genômica, Centro de Pesquisa Experimental - Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Rio Grande do Sul, Brazil; Serviço de Patologia- Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Zdenko Herceg
- International Agency for Research on Cancer, Lyon, France
| | | |
Collapse
|
36
|
Kim S, Lee C, Kim H, Yoon SO. Genetic characteristics of advanced oral tongue squamous cell carcinoma in young patients. Oral Oncol 2023; 144:106466. [PMID: 37393663 DOI: 10.1016/j.oraloncology.2023.106466] [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: 03/21/2023] [Revised: 05/26/2023] [Accepted: 06/20/2023] [Indexed: 07/04/2023]
Abstract
OBJECTIVES We aimed to investigate genetic alterations in oral tongue squamous cell carcinoma (OTSCC) based on age and the clinical significance of these alterations in young OTSCC patients. MATERIALS AND METHODS We detected genetic alterations in 44 cases of advanced OTSCC through next-generation sequencing and analyzed and compared patients either younger or older than 45 years. Further analysis was conducted on a validation group of 96 OTSCC patients aged ≤ 45 years to examine the clinical and prognostic associations of TERT promoter (TERTp) mutations. RESULTS TP53 mutation was the most common genetic alteration in advanced OTSCC (88.6%), followed by TERTp mutation (59.1%), CDKN2A mutation (31.8%), FAT1 mutation (9.1%), NOTCH1 mutation (9.1%), EGFR amplification (18.2%), and CDKN2A homozygous deletion (4.5%). TERTp mutation was the only genetic alteration significantly enriched in young patients (81.3% in young versus 46.4% in older; P < 0.024). Within the validation group of young patients, TERTp mutation was identified in 30 cases (30/96, 31.3%) and tended to be related to both smoking and alcohol consumption (P = 0.072), higher stage (P = 0.002), more frequent perineural invasion (P = 0.094), and worse overall survival (P = 0.012) than wild type. CONCLUSION Our findings suggest that TERTp mutation is more frequent in young patients with advanced OTSCC and is associated with worse clinical outcomes. Therefore, TERTp mutation may serve as a prognostic biomarker for OTSCC in young patients. The findings of this study may help in developing personalized treatment strategies for OTSCC based on age and genetic alterations.
Collapse
Affiliation(s)
- Sehui Kim
- Department of Pathology, Yonsei University College of Medicine, Severance Hospital, Seoul, South Korea; Department of Pathology, Korea University Guro Hospital, Seoul, South Korea
| | - Chung Lee
- Department of Pathology, Yonsei University College of Medicine, Severance Hospital, Seoul, South Korea
| | - Hyangmi Kim
- Department of Pathology, Yonsei University College of Medicine, Severance Hospital, Seoul, South Korea
| | - Sun Och Yoon
- Department of Pathology, Yonsei University College of Medicine, Severance Hospital, Seoul, South Korea.
| |
Collapse
|
37
|
Giunco S, Padovan M, Angelini C, Cavallin F, Cerretti G, Morello M, Caccese M, Rizzo B, d'Avella D, Della Puppa A, Chioffi F, De Bonis P, Zagonel V, De Rossi A, Lombardi G. Prognostic role and interaction of TERT promoter status, telomere length and MGMT promoter methylation in newly diagnosed IDH wild-type glioblastoma patients. ESMO Open 2023; 8:101570. [PMID: 37230028 PMCID: PMC10265608 DOI: 10.1016/j.esmoop.2023.101570] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND The clinical relevance of promoter mutations and single nucleotide polymorphism rs2853669 of telomerase reverse transcriptase (TERT) and telomere length in patients with isocitrate dehydrogenase (IDH) wild-type glioblastoma (GBM) patients remains unclear. Moreover, some studies speculated that TERT promoter status might influence the prognostic role of O6-methylguanine DNA methyltransferase (MGMT) promoter methylation in newly diagnosed GBM. We carried out a large study to investigate their clinical impact and their interaction in newly diagnosed GBM patients. PATIENTS AND METHODS We included 273 newly diagnosed IDH wild-type GBM patients who started treatment at Veneto Institute of Oncology IOV - IRCCS (Padua, Italy) from December 2016 to January 2020. TERT promoter mutations (-124 C>T and -146 C>T) and SNP rs2853669 (-245 T>C), relative telomere length (RTL) and MGMT methylation status were retrospectively assessed in this prospective cohort of patients. RESULTS Median overall survival (OS) of 273 newly diagnosed IDH wild-type GBM patients was 15 months. TERT promoter was mutated in 80.2% of patients, and most had the rs2853669 single nucleotide polymorphism as T/T genotype (46.2%). Median RTL was 1.57 (interquartile range 1.13-2.32). MGMT promoter was methylated in 53.4% of cases. At multivariable analysis, RTL and TERT promoter mutations were not associated with OS or progression-free survival (PFS). Notably, patients C carrier of rs2853669 (C/C+C/T genotypes) showed a better PFS compared with those with the T/T genotype (hazard ratio 0.69, P = 0.007). In terms of OS and PFS, all interactions between MGMT, TERT and RTL and between TERT and rs2853669 genotype were not statistically significant. CONCLUSIONS Our findings suggest the presence of the C variant allele at the rs2853669 of the TERT promoter as an attractive independent prognostic biomarker of disease progression in IDH wild-type GBM patients. RTL and TERT promoter mutational status were not correlated to survival regardless of MGMT methylation status.
Collapse
Affiliation(s)
- S Giunco
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - M Padovan
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - C Angelini
- Neurosurgery, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - F Cavallin
- Independent Statistician, Solagna, Italy
| | - G Cerretti
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - M Morello
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - M Caccese
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - B Rizzo
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - D d'Avella
- Department of Neuroscience, Neurosurgery, University of Padua, Padua, Italy
| | - A Della Puppa
- Department of Neurosurgery, Neuroscience, Psychology, Drug Area and Child Health (NEUROFARBA), University of Florence, Careggi University Hospital, Florence, Italy
| | - F Chioffi
- Neurosurgery, Azienda Ospedaliera- Università Padova, Padua, Italy
| | - P De Bonis
- Neurosurgery, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - V Zagonel
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - A De Rossi
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy; Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - G Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy.
| |
Collapse
|
38
|
Dono A, Moosvi AM, Goli PS, Bellman AC, Aung PP, Esquenazi Y, Ballester LY. TERT Immunohistochemistry as a Surrogate Marker for TERT Promoter Mutations in Infiltrating Gliomas. Appl Immunohistochem Mol Morphol 2023; 31:288-294. [PMID: 36952585 PMCID: PMC10393218 DOI: 10.1097/pai.0000000000001118] [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: 01/17/2022] [Accepted: 02/15/2023] [Indexed: 03/25/2023]
Abstract
Genomic alterations are critical for the diagnosis, prognostication, and treatment of patients with infiltrating gliomas. Telomerase reverse transcriptase promoter ( TERT p) mutations are among such crucial alterations. Although DNA sequencing is the preferred method for identifying TERT p mutations, it has limitations related to cost and accessibility. We tested telomerase reverse transcriptase (TERT) immunohistochemistry (IHC) as a surrogate for TERT p mutations in infiltrating gliomas. Thirty-one infiltrating gliomas were assessed by IHC using an anti-TERT Y182 antibody. IHC results were analyzed by a board-certified neuropathologist. Tumors were analyzed by targeted next-generation sequencing. A literature review of the use of TERT antibodies as a surrogate for TERT p mutations was performed. Eighteen gliomas harbored TERT p mutations. Overall, TERT IHC demonstrated a sensitivity of 61.1% and a specificity of 69.2% for identifying TERT p mutations. Among the 19 IDH1/IDH2 -wild-type gliomas, 16 (84%) harbored TERT p mutations, and TERT IHC had a sensitivity of 62.5% and a specificity of 33.3%. Among the 12 IDH1/IDH2 -mutant gliomas, 2 (17%) harbored TERT p mutations, and TERT IHC had a sensitivity of 50% and a specificity of 80%. TERT IHC had low positive and negative likelihood values in the identification of TERT p mutations. The literature review included 5 studies with 645 patients and 4 different TERT antibodies. The results consistently showed poor sensitivity and specificity of TERT IHC for identifying TERT p mutations. TERT IHC is a suboptimal surrogate marker for TERT p mutations in infiltrating gliomas. The need remains for cost-effective, efficient, and accessible alternatives to next-generation sequencing for the evaluation of TERT p mutations in gliomas.
Collapse
Affiliation(s)
- Antonio Dono
- Department of Pathology and Laboratory Medicine
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston
| | | | | | | | - Phyu P. Aung
- Department of Pathology, The University of Texas MD Anderson Cancer Center
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston
- Memorial Hermann Hospital—TMC, Houston, TX
| | - Leomar Y. Ballester
- Department of Pathology and Laboratory Medicine
- Department of Pathology, The University of Texas MD Anderson Cancer Center
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center
| |
Collapse
|
39
|
Mohanty SK, Lobo A, Mishra SK, Cheng L. Precision Medicine in Bladder Cancer: Present Challenges and Future Directions. J Pers Med 2023; 13:jpm13050756. [PMID: 37240925 DOI: 10.3390/jpm13050756] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Bladder cancer (BC) is characterized by significant histopathologic and molecular heterogeneity. The discovery of molecular pathways and knowledge of cellular mechanisms have grown exponentially and may allow for better disease classification, prognostication, and development of novel and more efficacious noninvasive detection and surveillance strategies, as well as selection of therapeutic targets, which can be used in BC, particularly in a neoadjuvant or adjuvant setting. This article outlines recent advances in the molecular pathology of BC with a better understanding and deeper focus on the development and deployment of promising biomarkers and therapeutic avenues that may soon make a transition into the domain of precision medicine and clinical management for patients with BC.
Collapse
Affiliation(s)
- Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute and CORE Diagnostics, Gurgaon 122016, India
| | - Anandi Lobo
- Department of Pathology and Laboratory Medicine, Kapoor Center for Pathology and Urology, Raipur 490042, India
| | - Sourav K Mishra
- Department of Medical Oncology, All India Institute of Medical Sciences, Bhubaneswar 750017, India
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Brown University Warren Alpert Medical School, Lifespan Academic Medical Center, and the Legorreta Cancer Center at Brown University, 593 Eddy Street, APC 12-105, Providence, RI 02903, USA
| |
Collapse
|
40
|
Lee K, Kim SI, Kim EE, Shim YM, Won JK, Park CK, Choi SH, Yun H, Lee H, Park SH. Genomic profiles of IDH-mutant gliomas: MYCN-amplified IDH-mutant astrocytoma had the worst prognosis. Sci Rep 2023; 13:6761. [PMID: 37185778 PMCID: PMC10130138 DOI: 10.1038/s41598-023-32153-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/23/2023] [Indexed: 05/17/2023] Open
Abstract
This study aimed to find any ambiguous genetic outlier for "oligodendroglioma, IDH-mutant and 1p/19q-codeleted (O_IDH_mut)" and "astrocytoma, IDH-mutant (A_IDH_mut)" and to redefine the genetic landscape and prognostic factors of IDH-mutant gliomas. Next-generation sequencing (NGS) using a brain tumor-targeted gene panel, methylation profiles, and clinicopathological features were analyzed for O_IDH_mut (n = 74) in 70 patients and for A_IDH_mut (n = 95) in 90 patients. 97.3% of O_IDH_mut and 98.9% of A_IDH_mut displayed a classic genomic landscape. Combined CIC (75.7%) and/or FUBP1 (45.9%) mutations were detected in 93.2% and MGMTp methylation in 95.9% of O_IDH_mut patients. In A_IDH_mut, TP53 mutations were found in 86.3% and combined ATRX (82.1%) and TERTp (6.3%) mutations in 88.4%. Although there were 3 confusing cases, NOS (not otherwise specified) category, based on genetic profiles, but they were clearly classified by combining histopathology and DKFZ methylation classifier algorithms. The patients with MYCN amplification and/or CDKN2A/2B homozygous deletion in the A_IDH_mut category had a worse prognosis than those without these gene alterations and MYCN-amplified A_IDH_mut showed the worst prognosis. However, there was no prognostic genetic marker in O_IDH_mut. In histopathologically or genetically ambiguous cases, methylation profiles can be used as an objective tool to avoid a diagnosis of NOS or NEC (not elsewhere classified), as well as for tumor classification. The authors have not encountered a case of true mixed oligoastrocytoma using an integrated diagnosis of histopathological, genetic and methylation profiles. MYCN amplification, in addition to CDKN2A/2B homozygous deletion, should be included in the genetic criteria for CNS WHO grade 4 A_IDH_mut.
Collapse
Affiliation(s)
- Kwanghoon Lee
- Department of Pathology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seong-Ik Kim
- Department of Pathology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Eric Eunshik Kim
- Department of Pathology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yu-Mi Shim
- Department of Pathology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jae-Kyung Won
- Department of Pathology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Kee Park
- Department of Neurosurgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung Hong Choi
- Department of Radiology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hongseok Yun
- Department of Genomic Medicine, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyunju Lee
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
- Artificial Intelligence Graduate School, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
41
|
Hanna C, Willman M, Cole D, Mehkri Y, Liu S, Willman J, Lucke-Wold B. Review of meningioma diagnosis and management. EGYPTIAN JOURNAL OF NEUROSURGERY 2023; 38:16. [PMID: 37124311 PMCID: PMC10138329 DOI: 10.1186/s41984-023-00195-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/14/2022] [Indexed: 05/02/2023] Open
Abstract
Meningiomas are the most common intracranial tumors in adult patients. Although the majority of meningiomas are diagnosed as benign, approximately 20% of cases are high-grade tumors that require significant clinical treatment. The gold standard for grading central nervous system tumors comes from the World Health Organization Classification of Tumors of the central nervous system. Treatment options also depend on the location, imaging, and histopathological features of the tumor. This review will cover diagnostic strategies for meningiomas, including 2021 updates to the World Health Organization's grading of meningiomas. Meningioma treatment plans are variable and highly dependent on tumor grading. This review will also update the reader on developments in the treatment of meningiomas, including surgery, radiation therapy and monoclonal antibody treatment.
Collapse
Affiliation(s)
- Chadwin Hanna
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Matthew Willman
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Dwayne Cole
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Yusuf Mehkri
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Sophie Liu
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Jonathan Willman
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | | |
Collapse
|
42
|
Cheng CK, Yung YL, Chan HY, Leung KT, Chan KYY, Leung AWK, Cheng FWT, Li CK, Wan TSK, Luo X, Pitts HA, Cheung JS, Chan NPH, Ng MHL. Deep genomic characterization highlights complexities and prognostic markers of pediatric acute myeloid leukemia. Commun Biol 2023; 6:356. [PMID: 37002311 PMCID: PMC10066286 DOI: 10.1038/s42003-023-04732-2] [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: 10/05/2022] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
Pediatric acute myeloid leukemia (AML) is an uncommon but aggressive hematological malignancy. The poor outcome is attributed to inadequate prognostic classification and limited treatment options. A thorough understanding on the genetic basis of pediatric AML is important for the development of effective approaches to improve outcomes. Here, by comprehensively profiling fusion genes as well as mutations and copy number changes of 141 myeloid-related genes in 147 pediatric AML patients with subsequent variant functional characterization, we unveil complex mutational patterns of biological relevance and disease mechanisms including MYC deregulation. Also, our findings highlight TP53 alterations as strong adverse prognostic markers in pediatric AML and suggest the core spindle checkpoint kinase BUB1B as a selective dependency in this aggressive subgroup. Collectively, our present study provides detailed genomic characterization revealing not only complexities and mechanistic insights into pediatric AML but also significant risk stratification and therapeutic strategies to tackle the disease.
Collapse
Affiliation(s)
- Chi-Keung Cheng
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuk-Lin Yung
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Hoi-Yun Chan
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Kam-Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Kathy Y Y Chan
- Department of Paediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Alex W K Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Frankie W T Cheng
- Department of Paediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi-Kong Li
- Department of Paediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Thomas S K Wan
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Xi Luo
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Herbert-Augustus Pitts
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Joyce S Cheung
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Natalie P H Chan
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Margaret H L Ng
- Blood Cancer Cytogenetics and Genomics Laboratory, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
43
|
Cheng L, Lopez-Beltran A, Wang M, Whaley RD, De Souza A, Au S, Ge R, Cimadamore A, Amin A, Golijanin B, MacLennan GT, Osunkoya AO, Montironi R, Zhang S. Frequent Telomerase Reverse Transcriptase (TERT) Promoter and Fibroblast Growth Factor Receptor 3 (FGFR3) Mutations Support the Precursor Nature of Papillary Urothelial Hyperplasia of the Urinary Bladder. Mod Pathol 2023; 36:100151. [PMID: 36906071 DOI: 10.1016/j.modpat.2023.100151] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/11/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023]
Abstract
The precursor nature of papillary urothelial hyperplasia of the urinary bladder is uncertain. In this study, we investigated TERT promoter and FGFR3 mutations in 82 patients with papillary urothelial hyperplasia lesions. Thirty eight patients presented with papillary urothelial hyperplasia and concurrent non-invasive papillary urothelial carcinoma and 44 patients presented with de novo papillary urothelial hyperplasia. The prevalence of TERT promoter and FGFR3 mutation are compared between de novo papillary urothelial hyperplasia and those with concurrent papillary urothelial carcinoma. Mutational concordance between papillary urothelial hyperplasia and concurrent carcinoma were also compared. The TERT promoter mutations were detected in 44% (36/82) of papillary urothelial hyperplasia including 23 (23/38, 61%) papillary urothelial hyperplasia with urothelial carcinoma, and 13 (13/44, 29%) de novo papillary urothelial hyperplasia. The overall concordance of TERT promoter mutation status between papillary urothelial hyperplasia and concurrent urothelial carcinoma was 76%. Overall FGFR3 mutation rate of papillary urothelial hyperplasia was 23% (19/82). FGFR3 mutations were detected in 11 patients with papillary urothelial hyperplasia and concurrent urothelial carcinoma (11/38, 29%), and 8 patients with de novo papillary urothelial hyperplasia (8/44, 18%). Identical FGFR3 mutation status was detected in both papillary urothelial hyperplasia and urothelial carcinoma component in all 11 patients with FGFR3 mutations. Our findings provide strong evidence of genetic association between papillary urothelial hyperplasia and urothelial carcinoma. High frequency of TERT promoter and FGFR3 mutations suggests the precursor role of papillary urothelial hyperplasia in urothelial carcinogenesis.
Collapse
Affiliation(s)
- Liang Cheng
- Department of Pathology and Laboratory Medicine, Brown University Warren Alpert Medical School, Lifespan Academic Medical Center, and the Legorreta Cancer Center at Brown University, Providence, RI, USA; Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Antonio Lopez-Beltran
- Department of Morphological Sciences, University of Cordoba Medical School, Cordoba, Spain
| | - Mingsheng Wang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rumeal D Whaley
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN, USA
| | - Andre De Souza
- Division of Hematology Oncology, Legorreta Cancer Center at Brown University, Lifespan Cancer Institute, Providence, RI, USA
| | - Sammy Au
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rongbin Ge
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | | | - Ali Amin
- Department of Pathology and Laboratory Medicine, Brown University Warren Alpert Medical School, Lifespan Academic Medical Center, and the Legorreta Cancer Center at Brown University, Providence, RI, USA
| | - Borivoj Golijanin
- Department of Pathology and Laboratory Medicine, Brown University Warren Alpert Medical School, Lifespan Academic Medical Center, and the Legorreta Cancer Center at Brown University, Providence, RI, USA
| | - Gregory T MacLennan
- Department of Pathology and Urology, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Adeboye O Osunkoya
- Departments of Pathology and Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rodolfo Montironi
- Molecular Medicine and Cell Therapy Foundation, Department of Clinical and Molecular Sciences, Polytechnic University of the Marche Region, Ancona, Italy
| | - Shaobo Zhang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
44
|
Cheng L, Zhang S, Wang M, Lopez-Beltran A. Biological and clinical perspectives of TERT promoter mutation detection on bladder cancer diagnosis and management. Hum Pathol 2023; 133:56-75. [PMID: 35700749 DOI: 10.1016/j.humpath.2022.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/05/2022] [Indexed: 02/08/2023]
Abstract
The telomerase reverse transcriptase (TERT) promoter mutations are associated with increased TERT mRNA and TERT protein levels, telomerase activity, and shorter but stable telomere length. TERT promoter mutation is the most common mutation that occurs in approximately 60-80% of patients with bladder cancer. The TERT promoter mutations occur in a wide spectrum of urothelial lesions, including benign urothelial proliferation and tumor-like conditions, benign urothelial tumors, premalignant and putative precursor lesions, urothelial carcinoma and its variants, and nonurothelial malignancies. The prevalence and incidence of TERT promoter mutations in a total of 7259 cases from the urinary tract were systematically reviewed. Different platforms of TERT promoter mutation detection were presented. In this review, we also discussed the significance and clinical implications of TERT promoter mutation detection in urothelial tumorigenesis, surveillance and early detection, diagnosis, differential diagnosis, prognosis, prediction of treatment responses, and clinical outcome. Identification of TERT promoter mutations from urine or plasma cell-free DNA (liquid biopsy) will facilitate bladder cancer screening program and optimal clinical management. A better understanding of TERT promoter mutation and its pathway would open new therapeutic avenues for patients with bladder cancer.
Collapse
Affiliation(s)
- Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University and Lifespan Academic Medical Center, Providence, RI, 02903, USA.
| | - Shaobo Zhang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mingsheng Wang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Antonio Lopez-Beltran
- Department of Morphological Sciences, University of Cordoba Medical School, Cordoba, E-14004, Spain
| |
Collapse
|
45
|
Yeh TJ, Luo CW, Du JS, Huang CT, Wang MH, Chuang TM, Gau YC, Cho SF, Liu YC, Hsiao HH, Chen LT, Pan MR, Wang HC, Moi SH. Deciphering the Functions of Telomerase Reverse Transcriptase in Head and Neck Cancer. Biomedicines 2023; 11:691. [PMID: 36979671 PMCID: PMC10044978 DOI: 10.3390/biomedicines11030691] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/30/2023] Open
Abstract
Head and neck cancers (HNCs) are among the ten leading malignancies worldwide. Despite significant progress in all therapeutic modalities, predictive biomarkers, and targeted therapies for HNCs are limited and the survival rate is unsatisfactory. The importance of telomere maintenance via telomerase reactivation in carcinogenesis has been demonstrated in recent decades. Several mechanisms could activate telomerase reverse transcriptase (TERT), the most common of which is promoter alternation. Two major hotspot TERT promoter mutations (C228T and C250T) have been reported in different malignancies such as melanoma, genitourinary cancers, CNS tumors, hepatocellular carcinoma, thyroid cancers, sarcomas, and HNCs. The frequencies of TERT promoter mutations vary widely across tumors and is quite high in HNCs (11.9-64.7%). These mutations have been reported to be more enriched in oral cavity SCCs and HPV-negative tumors. The association between TERT promoter mutations and poor survival has also been demonstrated. Till now, several therapeutic strategies targeting telomerase have been developed although only a few drugs have been used in clinical trials. Here, we briefly review and summarize our current understanding and evidence of TERT promoter mutations in HNC patients.
Collapse
Affiliation(s)
- Tsung-Jang Yeh
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chi-Wen Luo
- Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Cosmetic Science and Institute of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
| | - Jeng-Shiun Du
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chien-Tzu Huang
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Min-Hung Wang
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tzer-Ming Chuang
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yuh-Ching Gau
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shih-Feng Cho
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yi-Chang Liu
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hui-Hua Hsiao
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Li-Tzong Chen
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
| | - Mei-Ren Pan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Hui-Ching Wang
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Sin-Hua Moi
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
46
|
Carrasco Pro S, Hook H, Bray D, Berenzy D, Moyer D, Yin M, Labadorf AT, Tewhey R, Siggers T, Fuxman Bass JI. Widespread perturbation of ETS factor binding sites in cancer. Nat Commun 2023; 14:913. [PMID: 36808133 PMCID: PMC9938127 DOI: 10.1038/s41467-023-36535-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 02/03/2023] [Indexed: 02/19/2023] Open
Abstract
Although >90% of somatic mutations reside in non-coding regions, few have been reported as cancer drivers. To predict driver non-coding variants (NCVs), we present a transcription factor (TF)-aware burden test based on a model of coherent TF function in promoters. We apply this test to NCVs from the Pan-Cancer Analysis of Whole Genomes cohort and predict 2555 driver NCVs in the promoters of 813 genes across 20 cancer types. These genes are enriched in cancer-related gene ontologies, essential genes, and genes associated with cancer prognosis. We find that 765 candidate driver NCVs alter transcriptional activity, 510 lead to differential binding of TF-cofactor regulatory complexes, and that they primarily impact the binding of ETS factors. Finally, we show that different NCVs within a promoter often affect transcriptional activity through shared mechanisms. Our integrated computational and experimental approach shows that cancer NCVs are widespread and that ETS factors are commonly disrupted.
Collapse
Affiliation(s)
| | - Heather Hook
- Department of Biology, Boston University, Boston, MA, USA
| | - David Bray
- Bioinformatics Program, Boston University, Boston, MA, USA
| | | | - Devlin Moyer
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - Meimei Yin
- Department of Biology, Boston University, Boston, MA, USA
| | - Adam Thomas Labadorf
- Bioinformatics Hub, Boston University, Boston, MA, USA
- Boston University School of Medicine, Department of Neurology, Boston, MA, USA
| | | | - Trevor Siggers
- Bioinformatics Program, Boston University, Boston, MA, USA.
- Department of Biology, Boston University, Boston, MA, USA.
- Biological Design Center, Boston University, Boston, MA, USA.
| | - Juan Ignacio Fuxman Bass
- Bioinformatics Program, Boston University, Boston, MA, USA.
- Department of Biology, Boston University, Boston, MA, USA.
| |
Collapse
|
47
|
Jain M, Tivtikyan A, Kamalov D, Avdonin S, Rakhmatullin T, Pisarev E, Zvereva M, Samokhodskaya L, Kamalov A. Development of a Sensitive Digital Droplet PCR Screening Assay for the Detection of GPR126 Non-Coding Mutations in Bladder Cancer Urine Liquid Biopsies. Biomedicines 2023; 11:495. [PMID: 36831030 PMCID: PMC9953558 DOI: 10.3390/biomedicines11020495] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Recent whole-genome sequencing studies identified two novel recurrent mutations in the enhancer region of GPR126 in urothelial bladder cancer (UBC) tumor samples. This mutational hotspot is the second most common after the TERT promoter in UBC. The aim of the study was to develop a digital droplet PCR screening assay for the simultaneous detection of GPR126 mutations in a single tube. Its performance combined with TERT promoter mutation analysis was evaluated in urine of healthy volunteers (n = 50) and patients with cystitis (n = 22) and UBC (n = 70). The developed assay was validated using DNA constructs carrying the studied variants. None of the mutations were detected in control and cystitis group samples. GPR126 mutations were observed in the urine of 25/70 UBC patients (area under the ROC curve (AUC) of 0.679; mutant allele fraction (MAF) of 21.61 [8.30-44.52] %); TERT mutations-in 40/70 (AUC of 0.786; MAF = 28.29 [19.03-38.08] %); ≥1 mutation-in 47/70 (AUC of 0.836)). The simultaneous presence of GPR126 and TERT mutations was observed in 18/70 cases, with no difference in MAFs for the paired samples (31.96 [14.78-47.49] % vs. 27.13 [17.00-37.62] %, p = 0.349, respectively). The combined analysis of these common non-coding mutations in urine allows the sensitive and non-invasive detection of UBC.
Collapse
Affiliation(s)
- Mark Jain
- Medical Research and Educational Center, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Alexander Tivtikyan
- Medical Research and Educational Center, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - David Kamalov
- Medical Research and Educational Center, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Savva Avdonin
- Department of Fundamental Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Tagir Rakhmatullin
- Department of Fundamental Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Eduard Pisarev
- Department of Bioinformatics and Bioengineering, Lomonosov Moscow State University, 119991 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Maria Zvereva
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Larisa Samokhodskaya
- Medical Research and Educational Center, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Armais Kamalov
- Medical Research and Educational Center, Lomonosov Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
48
|
Fujiwara-Tani R, Takagi T, Mori S, Kishi S, Nishiguchi Y, Sasaki T, Ikeda M, Nagai K, Bhawal UK, Ohmori H, Fujii K, Kuniyasu H. Short Telomere Lesions with Dysplastic Metaplasia Histology May Represent Precancerous Lesions of Helicobacter pylori-Positive Gastric Mucosa. Int J Mol Sci 2023; 24:3182. [PMID: 36834592 PMCID: PMC9958872 DOI: 10.3390/ijms24043182] [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: 11/14/2022] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Gastric cancers are strongly associated with Helicobacter pylori infection, with intestinal metaplasia characterizing the background mucosa in most cases. However, only a subset of intestinal metaplasia cases proceed to carcinogenesis, and the characteristics of high-risk intestinal metaplasia that link it with gastric cancer are still unclear. We examined telomere reduction in five gastrectomy specimens using fluorescence in situ hybridization, and identified areas with localized telomere loss (outside of cancerous lesions), which were designated as short telomere lesions (STLs). Histological analyses indicated that STLs were characteristic of intestinal metaplasia accompanied by nuclear enlargement but lacking structural atypia, which we termed dysplastic metaplasia (DM). A review of gastric biopsy specimens from 587 H. pylori-positive patients revealed 32 cases of DM, 13 of which were classified as high-grade based on the degree of nuclear enlargement. All high-grade DM cases exhibited a telomere volume reduced to less than 60% of that of lymphocytes, increased stemness, and telomerase reverse transcriptase (TERT) expression. Two patients (15%) exhibited low levels of p53 nuclear retention. After a 10-year follow-up, 7 (54%) of the high-grade DM cases had progressed to gastric cancer. These results suggest that DM is characterized by telomere shortening, TERT expression, and stem cell proliferation, and high-grade DM is a high-grade intestinal metaplasia that likely represents a precancerous lesion of gastric cancer. High-grade DM is expected to effectively prevent progression to gastric cancer in H. pylori-positive patients.
Collapse
Grants
- 19K16564 Ministry of Education, Culture, Sports, Science and Technology
- 20K21659 Ministry of Education, Culture, Sports, Science and Technology
- 20K18007 Ministry of Education, Culture, Sports, Science and Technology
- 21K10143 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan
| | - Tadataka Takagi
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan
| | - Shiori Mori
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan
| | - Shingo Kishi
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan
| | - Yukiko Nishiguchi
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan
| | - Takamitsu Sasaki
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan
| | - Masayuki Ikeda
- Miyoshi Central Hospital, 10531 Higashi-Sakaya-cho, Miyoshi 728-8502, Japan
| | - Kenta Nagai
- Miyoshi Central Hospital, 10531 Higashi-Sakaya-cho, Miyoshi 728-8502, Japan
| | - Ujjal Kumar Bhawal
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Japan
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College, Chennai 600077, India
| | - Hitoshi Ohmori
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan
| | - Kiyomu Fujii
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan
| |
Collapse
|
49
|
Dasgupta P, Balasubramanyian V, de Groot JF, Majd NK. Preclinical Models of Low-Grade Gliomas. Cancers (Basel) 2023; 15:cancers15030596. [PMID: 36765553 PMCID: PMC9913857 DOI: 10.3390/cancers15030596] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/03/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
Diffuse infiltrating low-grade glioma (LGG) is classified as WHO grade 2 astrocytoma with isocitrate dehydrogenase (IDH) mutation and oligodendroglioma with IDH1 mutation and 1p/19q codeletion. Despite their better prognosis compared with glioblastoma, LGGs invariably recur, leading to disability and premature death. There is an unmet need to discover new therapeutics for LGG, which necessitates preclinical models that closely resemble the human disease. Basic scientific efforts in the field of neuro-oncology are mostly focused on high-grade glioma, due to the ease of maintaining rapidly growing cell cultures and highly reproducible murine tumors. Development of preclinical models of LGG, on the other hand, has been difficult due to the slow-growing nature of these tumors as well as challenges involved in recapitulating the widespread genomic and epigenomic effects of IDH mutation. The most recent WHO classification of CNS tumors emphasizes the importance of the role of IDH mutation in the classification of gliomas, yet there are relatively few IDH-mutant preclinical models available. Here, we review the in vitro and in vivo preclinical models of LGG and discuss the mechanistic challenges involved in generating such models and potential strategies to overcome these hurdles.
Collapse
Affiliation(s)
- Pushan Dasgupta
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
| | | | - John F. de Groot
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA 94143, USA
- Correspondence: (J.F.d.G.); (N.K.M.)
| | - Nazanin K. Majd
- Department of Neuro-Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (J.F.d.G.); (N.K.M.)
| |
Collapse
|
50
|
Effect of Having Concurrent Mutations on the Degree of Aggressiveness in Patients with Thyroid Cancer Positive for TERT Promoter Mutations. Cancers (Basel) 2023; 15:cancers15020413. [PMID: 36672362 PMCID: PMC9856260 DOI: 10.3390/cancers15020413] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/30/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
This study aimed to examine whether concurrent mutations with a TERT promoter mutation are associated with a greater likelihood of more aggressive disease than a TERT promoter mutation alone. The medical records of 1477 patients who underwent thyroid surgery at two tertiary hospitals between 2017 and 2022 were reviewed. Twenty-four patients had TERT promoter mutations based on molecular profile testing. Clinicodemographic data, mutational profiles, and histopathological features were assessed. Descriptive analysis, Fisher's exact test, and binary logistic regression were performed. Seven patients had single-gene TERT promoter mutations, and 17 had concurrent mutations, including BRAF V600E, HRAS, NRAS, PIK3CA, and EIF1AX. The overall prevalence of malignancy was 95.8%, of which 78.3% were aggressive thyroid cancers. There was a statistically significant association between concurrent mutations and disease aggressiveness. The odds of having aggressive disease were 10 times higher in patients with a TERT promoter mutation and a concurrent molecular alteration than in those with a TERT promoter mutation alone. This is an important finding for thyroid specialists to consider when counseling patients concerning risk stratification and management options.
Collapse
|