1
|
Zhang B, Zhou Q, Xue C, Zhang P, Ke X, Wang Y, Zhang Y, Deng L, Jing M, Han T, Zhou F, Dong W, Zhou J. Predicting telomerase reverse transcriptase promoter mutation status in glioblastoma by whole-tumor multi-sequence magnetic resonance texture analysis. Magn Reson Imaging 2025; 118:110360. [PMID: 39983804 DOI: 10.1016/j.mri.2025.110360] [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: 09/10/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
OBJECTIVE This study aimed to determine the feasibility of preoperative multi-sequence magnetic resonance texture analysis (MRTA) for predicting TERT promoter mutation status in IDH-wildtype glioblastoma (IDHwt GB). METHODS The clinical and imaging data of 111 patients with IDHwt GB at our hospital between November 2018 and June 2023 were retrospectively analyzed as the training set, and those of 23 patients with IDHwt GB between July 2023 and November 2023 were interpreted as the validation set. We used molecular sequencing results to classify the training set into TERT promoter mutation and wildtype groups. Textural features of the whole-tumor volume were extracted, including T2-weighted imaging (T2WI), T2-fluid-attenuated inversion recovery, apparent diffusion coefficient (ADC) map, and contrast-enhanced T1-weighted imaging (CE-T1). All textural features were obtained using open-source pyradiomics. After feature selection, logistic regression was used to build prediction models, and a nomogram was generated. Finally, the model was validated using validation cohort. RESULTS The CE-T1_Model (AUC 0.704) had a better predictive ability than the T2_Model (AUC 0.684) and ADC_Model (AUC 0.624). The MRI_Combined_Model (CE-T1, T2, and ADC texture features) (AUC 0.780) had a better predictive ability than the Clinical_Model (AUC 0.758). The Combined_Model (CE-T1, T2, ADC texture features, and clinical features) had the best predictive performance (AUC 0.871), with a sensitivity, specificity, and accuracy of 82.60 %, 83.30 %, and 80.18 %, respectively. The AUC, sensitivity, specificity, and accuracy in the validation cohort were 0.775, 86.70 %, 75.00 %, and 69.57 %, respectively. CONCLUSIONS Whole-tumor multi-sequence MRTA can be used as non-invasive quantitative parameters to assist in the preoperative clinical prediction of TERT promoter mutation status in IDHwt GB.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Second Clinical School, Lanzhou University, Lanzhou, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Second Clinical School, Lanzhou University, Lanzhou, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Second Clinical School, Lanzhou University, Lanzhou, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Peng Zhang
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Second Clinical School, Lanzhou University, Lanzhou, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Yige Wang
- Medical Department, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Yuting Zhang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Second Clinical School, Lanzhou University, Lanzhou, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Liangna Deng
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Second Clinical School, Lanzhou University, Lanzhou, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Mengyuan Jing
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Second Clinical School, Lanzhou University, Lanzhou, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Tao Han
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Second Clinical School, Lanzhou University, Lanzhou, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Fengyu Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Second Clinical School, Lanzhou University, Lanzhou, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Wenjie Dong
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Second Clinical School, Lanzhou University, Lanzhou, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Second Clinical School, Lanzhou University, Lanzhou, Gansu, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China.
| |
Collapse
|
2
|
Zhang X, Gao Y, Zhang S, Wang Y, Du Y, Hao S, Ni T. The Regulation of Cellular Senescence in Cancer. Biomolecules 2025; 15:448. [PMID: 40149983 PMCID: PMC11940315 DOI: 10.3390/biom15030448] [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: 01/31/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Cellular senescence is a stable state of cell cycle arrest caused by telomere shortening or various stresses. After senescence, cells cease dividing and exhibit many age-related characteristics. Unlike the halted proliferation of senescence cells, cancer cells are considered to have unlimited growth potential. When cells display senescence-related features, such as telomere loss or stem cell failure, they can inhibit tumor development. Therefore, inducing cells to enter a senescence state can serve as a barrier to tumor cell development. However, many recent studies have found that sustained senescence of tumor cells or normal cells under certain circumstances can exert environment-dependent effects of tumor promotion and inhibition by producing various cytokines. In this review, we first introduce the causes and characteristics of induced cellular senescence, analyze the senescence process of immune cells and cancer cells, and then discuss the dual regulatory role of cell senescence on tumor growth and senescence-induced therapies targeting cancer cells. Finally, we discuss the role of senescence in tumor progression and treatment opportunities, and propose further studies on cellular senescence and cancer therapy.
Collapse
Affiliation(s)
- Xianhong Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Yue Gao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Siyu Zhang
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Sciences, Ningxia University, Yinchuan 750021, China;
| | - Yixiong Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Yitian Du
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Shuailin Hao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| | - Ting Ni
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China; (X.Z.); (Y.G.); (Y.W.); (Y.D.)
| |
Collapse
|
3
|
Cao C, Gong W, Shuai Y, Rasouli S, Ge Q, Khan A, Dakic A, Putluri N, Shvets G, Zheng YL, Daneshdoust D, Mahyoob R, Li J, Liu X. Canonical and non-canonical functions of the non-coding RNA component (TERC) of telomerase complex. Cell Biosci 2025; 15:30. [PMID: 40025596 PMCID: PMC11871756 DOI: 10.1186/s13578-025-01367-0] [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: 12/23/2024] [Accepted: 02/11/2025] [Indexed: 03/04/2025] Open
Abstract
The telomerase complex consists of a protein component (TERT), which has reverse transcriptase activity, and an RNA component (TERC), which serves as a template for telomere synthesis. Evidence is rapidly accumulating regarding the non-canonical functions of these components in both normal or diseased cells. An oligonucleotide-based drug, the first telomerase inhibitor, secured FDA approval in June 2024. We recently summarized the non-canonical functions of TERT in viral infections and cancer. In this review, we expand on these non-canonical functions of TERC beyond telomere maintenance. Specifically, we explore TERC's roles in cellular aging and senescence, immune regulation, genetic diseases, human cancer, as well as involvement in viral infections and host interactions. Finally, we discuss a transcription product of telomere repeats, TERRA, and explore strategies for targeting TERC as a therapeutic approach.
Collapse
Affiliation(s)
- Chongwen Cao
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Weiyi Gong
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Biomedical Science Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Yuanlong Shuai
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Biomedical Science Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Sara Rasouli
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Biomedical Science Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Qianyun Ge
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Anam Khan
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Aleksandra Dakic
- Division of Neuroscience, National Institute of Aging, Bethesda, MD, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Gennady Shvets
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY, USA
| | - Yun-Ling Zheng
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Danyal Daneshdoust
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Rani Mahyoob
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Jenny Li
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Xuefeng Liu
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
- Departments of Pathology, Urology and Radiation Oncology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
4
|
Sanford SL, Badstübner M, Gerber M, Mannherz W, Lampl N, Dannenberg R, Hinchie A, Schaich MA, Myong S, Hedglin M, Agarwal S, Alder JK, Stone MD, Opresko PL. Chemotherapeutic 6-thio-2'-deoxyguanosine selectively targets and inhibits telomerase by inducing a non-productive telomere-bound telomerase complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636339. [PMID: 39975053 PMCID: PMC11838547 DOI: 10.1101/2025.02.05.636339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Most cancers upregulate the telomere lengthening enzyme telomerase to achieve unlimited cell division. How chemotherapeutic nucleoside 6-thio-2'-deoxyguanosine (6-thio-dG) targets telomerase to inhibit telomere maintenance in cancer cells and tumors was unclear. Here, we demonstrate that telomerase insertion of 6-thio-dGTP prevents synthesis of additional telomeric repeats but does not disrupt telomerase binding to telomeres. Specifically, 6-thio-dG inhibits telomere extension after telomerase translocates along its product DNA to reposition the template, inducing a non-productive complex rather than enzyme dissociation. Furthermore, we provide direct evidence that 6-thio-dG treatment inhibits telomere synthesis by telomerase in cancer cells. In agreement, telomerase-expressing cancer cells harboring critically short telomeres are more sensitive to 6-thio-dG and show a greater induction of telomere losses compared to cancer cells with long telomere reserves. Our studies reveal that telomere length and telomerase status determine 6-thio-dG sensitivity and uncover the molecular mechanism by which 6-thio-dG selectively inhibits telomerase synthesis of telomeric DNA.
Collapse
Affiliation(s)
| | - Mareike Badstübner
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA, USA
| | | | - William Mannherz
- Division of Hematology/Oncology and Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Biological and Biomedical Sciences Program, Harvard/MIT MD-PhD Program, Harvard Stem Cell Institute, Harvard Initiative for RNA Medicine, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Noah Lampl
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Biological and Biomedical Sciences Program, Harvard/MIT MD-PhD Program, Harvard Stem Cell Institute, Harvard Initiative for RNA Medicine, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Rachel Dannenberg
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Angela Hinchie
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew A Schaich
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sua Myong
- Program in Cell, Molecular, Developmental Biology and Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mark Hedglin
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Suneet Agarwal
- Division of Hematology/Oncology and Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Biological and Biomedical Sciences Program, Harvard/MIT MD-PhD Program, Harvard Stem Cell Institute, Harvard Initiative for RNA Medicine, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Jonathan K Alder
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael D. Stone
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Patricia L Opresko
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Murugan AK, Kannan S, Alzahrani AS. TERT promoter mutations in gliomas: Molecular roles in tumorigenesis, metastasis, diagnosis, prognosis, therapeutic targeting, and drug resistance. Biochim Biophys Acta Rev Cancer 2025; 1880:189243. [PMID: 39674418 DOI: 10.1016/j.bbcan.2024.189243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Telomerase reverse transcriptase (TERT), a critical player in cellular immortalization, has emerged as a focal point of investigation due to its frequent promoter mutations in various human malignancies. TERT promoter mutations exhibit a significant role in tumorigenesis, fostering unbridled cellular proliferation and survival. This comprehensive review delves into the landscape of TERT promoter mutations and their profound implications in cancer, particularly within the context of gliomas. This article meticulously examines the intricate interplay between TERT promoter mutations and the metastatic cascade, shedding light on their capacity to orchestrate invasive behavior in gliomas. Moreover, this review describes the recent trends in therapeutic targeting of the TERT and dissects the evolving landscape of drug resistance associated with TERT mutations, providing insights into potential therapeutic challenges. In addition, the diagnostic and prognostic implications of TERT promoter mutations in gliomas are scrutinized, unraveling their potential as robust biomarkers. It also discusses the recent advancements in molecular diagnostics, illustrating the promise of TERT mutations as diagnostic tools and prognostic indicators. This review collectively aims to contribute to a deeper understanding of TERT promoter mutations in gliomas, offering a foundation for future research endeavors and paving the way for innovative strategies in glioma management.
Collapse
Affiliation(s)
- Avaniyapuram Kannan Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia.
| | - Siddarth Kannan
- School of Medicine, University of Central Lancashire, Preston PR1 2HE, UK
| | - Ali S Alzahrani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
6
|
Gehle DB, Jablonowski CM, Pichavaram P, Singh S, Woolard MA, Morton CL, Billups CA, Davidoff AM, Yang J, Murphy AJ. Characterizing Sensitivity to Vincristine, Irinotecan, and Telomerase-targeted Therapy in Diffuse Anaplastic Wilms Tumor Patient-derived Xenografts ☆. J Pediatr Surg 2024:162122. [PMID: 39753413 DOI: 10.1016/j.jpedsurg.2024.162122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Patients with diffuse anaplastic Wilms tumor (DAWT) experience relatively poor oncologic outcomes. Previous work has described mechanisms of telomerase upregulation in DAWT, posing a potential therapeutic target. METHODS We assessed in vitro sensitivity to vincristine, irinotecan, and telomerase-targeting drug 6-thio-2'-deoxyguanosine (6 dG) in DAWT cell lines WiT49 and PDM115 and in spheroids derived from cell lines and four DAWT patient-derived xenografts (PDX). We also tested in vivo response to vincristine/irinotecan (VI), 6 dG, or combination in WTPDX. RESULTS Sensitivity to vincristine varied with EC50 between 0.13 and 44.92 nM in spheroids, with EC50 for SN-38 (irinotecan active metabolite) from 3.06 to 70.96 nM. All were resistant to 6 dG monotherapy with EC50 from 3.06 to 50+ μM. In KT-51, 10 μM 6 dG significantly slowed spheroid growth. 6 dG treatment increased DNA damage response markers pChk1 S345, p53 and γH2AX levels in KT-51, KT-53 and KT-60 spheroids. In WiT49 2D culture, treatment of sub-toxic doses of 6 dG did not induce apoptosis or cell cycle arrest and exhibited minimal synergistic capacity with VI; TERT overexpression did not increase 6 dG sensitivity. In vivo treatment of KT-51, KT-53, and KT-60 with VI exhibited variable responses from progressive disease to complete clinical responses, but 6 dG monotherapy resulted in no tumor responses and 6 dG addition to VI conferred no increased tumor suppression. CONCLUSIONS DAWT models are variably sensitive to VI but are resistant to 6 dG monotherapy or combination with VI. Future research will address limitations of preclinical WT model systems and assess additional targeted therapies for high-risk WT subtypes.
Collapse
Affiliation(s)
- Daniel B Gehle
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Carolyn M Jablonowski
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Shivendra Singh
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Mary A Woolard
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Christopher L Morton
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Catherine A Billups
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jun Yang
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Andrew J Murphy
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
| |
Collapse
|
7
|
Siteni S, Grichuk A, Shay JW. Telomerase in Cancer Therapeutics. Cold Spring Harb Perspect Biol 2024; 16:a041703. [PMID: 39349313 PMCID: PMC11610755 DOI: 10.1101/cshperspect.a041703] [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: 10/02/2024]
Abstract
While silent in normal differentiated human tissues, telomerase is reactivated in most human cancers. Thus, telomerase is an almost universal oncology target. This update describes preclinical and clinical advancements using a variety of approaches to target telomerase. These include direct telomerase inhibitors, G-quadruplex DNA-interacting ligands, telomerase-based vaccine platforms, telomerase promoter-driven attenuated viruses, and telomerase-mediated telomere targeting approaches. While imetelstat has been recently approved by the Food and Drug Administration (FDA), several other approaches are in late-stage clinical development. The pros and cons of the major approaches will be reviewed.
Collapse
Affiliation(s)
- Silvia Siteni
- University of Texas Southwestern Medical Center, Department of Cell Biology, Dallas, Texas 75390, USA
| | - Anthony Grichuk
- University of Texas Southwestern Medical Center, Department of Cell Biology, Dallas, Texas 75390, USA
| | - Jerry W Shay
- University of Texas Southwestern Medical Center, Department of Cell Biology, Dallas, Texas 75390, USA
| |
Collapse
|
8
|
Hicks WH, Gattie LC, Shami ME, Traylor JI, Davar D, Najjar YG, Richardson TE, McBrayer SK, Abdullah KG. Matched three-dimensional organoids and two-dimensional cell lines of melanoma brain metastases mirror response to targeted molecular therapy. Sci Rep 2024; 14:24843. [PMID: 39438602 PMCID: PMC11496643 DOI: 10.1038/s41598-024-76583-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Despite advances in the treatment paradigm for patients with metastatic melanoma, melanoma brain metastasis (MBM) continues to represent a significant treatment challenge. The study of MBM is limited, in part, by shortcomings in existing preclinical models. Surgically eXplanted Organoids (SXOs) are ex vivo, three-dimensional cultures prepared from primary tissue samples with minimal processing that recapitulate genotypic and phenotypic features of parent tumors without an artificial extracellular scaffold. MBM SXOs were created by a novel protocol incorporating techniques for establishing glioma and cutaneous melanoma organoids. A BRAFV600K-mutant and BRAF-wildtype MBM sample were collected directly from the operating room. Organoids were cultured in an optimized culture medium without an artificial extracellular scaffold. Concurrently, matched patient-derived cell lines were created. Organoid growth was observed within 3-4 weeks, and MBM SXOs retained histological features of the parent tissue, including pleomorphic epithelioid cells with abundant cytoplasm, large nuclei, focal melanin accumulation, and strong SOX10 positivity. After sufficient growth, organoids could be manually parcellated to increase the number of replicates. Matched SXOs and cell lines demonstrated sensitivity to BRAF and MEK inhibitors. Further study using SXOs may improve the translational relevance of preclinical studies and enable the study of the metastatic melanoma tumor microenvironment.
Collapse
Affiliation(s)
- William H Hicks
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop St, Pittsburgh, PA, 15213, USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Lauren C Gattie
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop St, Pittsburgh, PA, 15213, USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Mohamad El Shami
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop St, Pittsburgh, PA, 15213, USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Jeffrey I Traylor
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, 75235, USA
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, 5303 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Diwakar Davar
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Yana G Najjar
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Timothy E Richardson
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Ave, New York, NY, 10029, USA
| | - Samuel K McBrayer
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, 75235, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kalil G Abdullah
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop St, Pittsburgh, PA, 15213, USA.
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA.
- Department of Neurological Surgery, University of Pittsburgh Medical Center Hillman Cancer Center, 5150 Centre Avenue, Suite 430, Pittsburgh, PA, 15232, USA.
| |
Collapse
|
9
|
Ge Y, Peng Y, Xie R, Luo Y, Li Y, Chen G. Visible Light-Mediated Late-Stage Thioetherification of Mercaptopurine Derivatives. Chemistry 2024; 30:e202401774. [PMID: 38923704 DOI: 10.1002/chem.202401774] [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: 05/06/2024] [Revised: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024]
Abstract
We disclose herein a novel and general radical approach to alkylthiopurines, encompassing 4 types of thiopurines, as well as their corresponding ribosides. This strategy is achieved through visible light-mediated late-stage functionalization of the sulfur atoms of mercaptopurines. The in situ-generated disulfide was proposed as the pivotal neutral intermediate for this transformation. We present herein a novel photo-mediated homolytic C-S bond formation for the preparation of alkylthiopurines and alkylthiopurine nucleosides. Despite the presence of reactive sites for the Minisci reaction, chemoselective S-alkylation remained the predominant pathway. This method allows for the late-stage introduction of a broad spectrum of alkyl groups onto the sulfur atom of unprotective mercaptopurine derivatives, encompassing 2-, 6-, and 8-mercaptopurine rings. Organoborons serve as efficient and eco-friendly alkylating reagents, providing advantages in terms of readily availability, stability, and reduced toxicity. Further derivatization of the thioetherified nucleosides, together with anti-tumor assays, led to the discovery of potent anti-tumor agents with an IC50 value reaching 6.1 μM (Comp. 31 for Jurkat).
Collapse
Affiliation(s)
- Yuhua Ge
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, P. R.China
| | - Yijiang Peng
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, P. R.China
| | - Ruoqian Xie
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, P. R.China
| | - Yang Luo
- Shanghai Key Laboratory for Molecular Engineer of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yangyan Li
- Shanghai Key Laboratory for Molecular Engineer of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Gang Chen
- Shanghai Key Laboratory for Molecular Engineer of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
- Key Laboratory of Green and High-value Utilization of Salt Lake Resources, Qinghai Institute of Salt Lakes, Chinese Academy of Sciences, Xining, Qinghai, 810008, P. R. China
| |
Collapse
|
10
|
Boccardi V, Marano L. Aging, Cancer, and Inflammation: The Telomerase Connection. Int J Mol Sci 2024; 25:8542. [PMID: 39126110 PMCID: PMC11313618 DOI: 10.3390/ijms25158542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024] Open
Abstract
Understanding the complex dynamics of telomere biology is important in the strong link between aging and cancer. Telomeres, the protective caps at the end of chromosomes, are central players in this connection. While their gradual shortening due to replication limits tumors expansion by triggering DNA repair mechanisms, it also promotes oncogenic changes within chromosomes, thus sustaining tumorigenesis. The enzyme telomerase, responsible for maintaining telomere length, emerges as a central player in this context. Its expression in cancer cells facilitates the preservation of telomeres, allowing them to circumvent the growth-limiting effects of short telomeres. Interestingly, the influence of telomerase extends beyond telomere maintenance, as evidenced by its involvement in promoting cell growth through alternative pathways. In this context, inflammation accelerates telomere shortening, resulting in telomere dysfunction, while telomere elements also play a role in modulating the inflammatory response. The recognition of this interplay has promoted the development of novel therapeutic approaches centered around telomerase inhibition. This review provides a comprehensive overview of the field, emphasizing recent progress in knowledge and the implications in understanding of cancer biology.
Collapse
Affiliation(s)
- Virginia Boccardi
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Luigi Marano
- Department of Medicine, Academy of Applied Medical and Social Sciences—AMiSNS: Akademia Medycznych I Spolecznych Nauk Stosowanych, 82-300 Elbląg, Poland;
- Department of General Surgery and Surgical Oncology, “Saint Wojciech” Hospital, “Nicolaus Copernicus” Health Center, 80-462 Gdańsk, Poland
| |
Collapse
|
11
|
Liu M, Gu L, Zhang Y, Li Y, Zhang L, Xin Y, Wang Y, Xu ZX. LKB1 inhibits telomerase activity resulting in cellular senescence through histone lactylation in lung adenocarcinoma. Cancer Lett 2024; 595:217025. [PMID: 38844063 DOI: 10.1016/j.canlet.2024.217025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/15/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Despite the confirmed role of LKB1 in suppressing lung cancer progression, its precise effect on cellular senescence is unknown. The aim of this research was to clarify the role and mechanism of LKB1 in restraining telomerase activity in lung adenocarcinoma. The results showed that LKB1 induced cellular senescence and apoptosis either in vitro or in vivo. Overexpression of LKB1 in LKB1-deficient A549 cells led to the inhibition of telomerase activity and the induction of telomere dysfunction by regulating telomerase reverse transcriptase (TERT) expression in terms of transcription. As a transcription factor, Sp1 mediated TERT inhibition after LKB1 overexpression. LKB1 induced lactate production and inhibited histone H4 (Lys8) and H4 (Lys16) lactylation, which further altered Sp1-related transcriptional activity. The telomerase inhibitor BIBR1532 was beneficial for achieving the optimum curative effect of traditional chemotherapeutic drugs accompanied by the glycolysis inhibitor 2DG. These data reveal a new mechanism by which LKB1 regulates telomerase activity through lactylation-dependent transcriptional inhibition, and therefore, provide new insights into the effects of LKB1-mediated senescence in lung adenocarcinoma. Our research has opened up new possibilities for the creation of new cancer treatments.
Collapse
Affiliation(s)
- Mingdi Liu
- 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
| | - Yuning Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China
| | - Yunkuo Li
- Department of Urology, the First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Lihong Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, Jilin, China
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, 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.
| |
Collapse
|
12
|
Gu L, Liu M, Zhang Y, Zhou H, Wang Y, Xu ZX. Telomere-related DNA damage response pathways in cancer therapy: prospective targets. Front Pharmacol 2024; 15:1379166. [PMID: 38910895 PMCID: PMC11190371 DOI: 10.3389/fphar.2024.1379166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024] Open
Abstract
Maintaining the structural integrity of genomic chromosomal DNA is an essential role of cellular life and requires two important biological mechanisms: the DNA damage response (DDR) mechanism and telomere protection mechanism at chromosome ends. Because abnormalities in telomeres and cellular DDR regulation are strongly associated with human aging and cancer, there is a reciprocal regulation of telomeres and cellular DDR. Moreover, several drug treatments for DDR are currently available. This paper reviews the progress in research on the interaction between telomeres and cellular DNA damage repair pathways. The research on the crosstalk between telomere damage and DDR is important for improving the efficacy of tumor treatment. However, further studies are required to confirm this hypothesis.
Collapse
Affiliation(s)
- Liting Gu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Mingdi Liu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Yuning Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Zhi-Xiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| |
Collapse
|
13
|
Waitkus MS, Erman EN, Reitman ZJ, Ashley DM. Mechanisms of telomere maintenance and associated therapeutic vulnerabilities in malignant gliomas. Neuro Oncol 2024; 26:1012-1024. [PMID: 38285162 PMCID: PMC11145458 DOI: 10.1093/neuonc/noae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Indexed: 01/30/2024] Open
Abstract
A majority of cancers (~85%) activate the enzyme telomerase to maintain telomere length over multiple rounds of cellular division. Telomerase-negative cancers activate a distinct, telomerase-independent mechanism of telomere maintenance termed alternative lengthening of telomeres (ALT). ALT uses homologous recombination to maintain telomere length and exhibits features of break-induced DNA replication. In malignant gliomas, the activation of either telomerase or ALT is nearly ubiquitous in pediatric and adult tumors, and the frequency with which these distinct telomere maintenance mechanisms (TMMs) is activated varies according to genetically defined glioma subtypes. In this review, we summarize the current state of the field of TMMs and their relevance to glioma biology and therapy. We review the genetic alterations and molecular mechanisms leading to telomerase activation or ALT induction in pediatric and adult gliomas. With this background, we review emerging evidence on strategies for targeting TMMs for glioma therapy. Finally, we comment on critical gaps and issues for moving the field forward to translate our improved understanding of glioma telomere maintenance into better therapeutic strategies for patients.
Collapse
Affiliation(s)
- Matthew S Waitkus
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Elise N Erman
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Zachary J Reitman
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - David M Ashley
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
14
|
Lee Y, Park CK, Park SH. Prognostic Impact of TERT Promoter Mutations in Adult-Type Diffuse Gliomas Based on WHO2021 Criteria. Cancers (Basel) 2024; 16:2032. [PMID: 38893152 PMCID: PMC11171308 DOI: 10.3390/cancers16112032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/07/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Mutation in the telomerase reverse transcriptase promoter (TERTp )is commonly observed in various malignancies, such as central nervous system (CNS) tumors, malignant melanoma, bladder cancer, and thyroid carcinoma. These mutations are recognized as significant poor prognostic factors for these tumors. In this investigation, a total of 528 cases of adult-type diffuse gliomas diagnosed at a single institution were reclassified according to the 2021 WHO classifications of CNS tumors, 5th edition (WHO2021). The study analyzed clinicopathological and genetic features, including TERTp mutations in each tumor. The impact of known prognostic factors on patient outcomes was analyzed through Kaplan-Meier survival and Cox regression analysis. TERTp mutations were predominantly identified in 94.1% of oligodendrogliomas (ODG), followed by 66.3% in glioblastoma, IDH-wildtype (GBM-IDHwt), and 9.2% of astrocytomas, IDH-mutant (A-IDHm). When considering A-IDHm and GBM as astrocytic tumors (Group 1) and ODGs (Group 2), TERTp mutations emerged as a significant adverse prognostic factor (p = 0.013) in Group 1. However, within each GBM-IDHwt and A-IDHm, the presence of TERTp mutations did not significantly impact patient prognosis (p = 0.215 and 0.268, respectively). Due to the high frequency of TERTp mutations in Group 2 (ODG) and their consistent prolonged survival, a statistical analysis to evaluate their impact on overall survival was deemed impractical. When considering MGMTp status, the combined TERTp-mutated and MGMTp-unmethylated group exhibited the worst prognosis in OS (p = 0.018) and PFS (p = 0.034) of GBM. This study confirmed that the classification of tumors according to the WHO2021 criteria effectively reflected prognosis. Both uni- and multivariate analyses in GBM, age, MGMTp methylation, and CDKN2A/B homozygous deletion were statistically significant prognostic factors while in univariate analysis in A-IDHm, grade 4, the Ki-67 index and MYCN amplifications were statistically significant prognostic factors. This study suggests that it is important to classify and manage tumors based on their genetic characteristics in adult-type diffuse gliomas.
Collapse
Affiliation(s)
- Yujin Lee
- Department of Hospital Pathology, St. Vincent’s Hospital, The Catholic University of Korea College of Medicine, 93, Jungbu-daero, Paldal-gu, Suwon 16247, Gyeonggi-do, Republic of Korea;
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University College of Medicine, 103 Deahak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, 103 Deahak-ro, Jongno-gu, Seoul 03080, Republic of Korea
- Neuroscience Institute, Seoul National University College of Medicine, 103 Deahak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| |
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
|
Jayaprasad AG, Chandrasekharan A, Arun Jyothi SP, John Sam SM, Santhoshkumar TR, Pillai MR. Telomerase inhibitors induce mitochondrial oxidation and DNA damage-dependent cell death rescued by Bcl-2/Bcl-xL. Int J Biol Macromol 2024; 264:130151. [PMID: 38403227 DOI: 10.1016/j.ijbiomac.2024.130151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/05/2024] [Accepted: 02/11/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND Reactivation of telomerase is a hallmark of cancer and the majority of cancers over-express telomerase. Telomerase-dependent telomere length maintenance confers immortality to cancer cells. However, telomere length-independent cell survival functions of telomerase also play a critical role in tumorigenesis. Multiple telomerase inhibitors have been developed as therapeutics and include anti-sense oligonucleotides, telomerase RNA component targeting agents, chemical inhibitors of telomerase, small molecule inhibitors of hTERT, and telomerase vaccine. In general, telomerase inhibitors affect cell proliferation and survival of cells depending on the telomere length reduction, culminating in replicative senescence or cell death by crisis. However, most telomerase inhibitors kill cancer cells prior to significant reduction in telomere length, suggesting telomere length independent role of telomerase in early telomere dysfunction-dependent cell death. METHODS In this study, we explored the mechanism of cell death induced by three prominent telomerase inhibitors utilizing a series of genetically encoded sensor cells including redox and DNA damage sensor cells. RESULTS We report that telomerase inhibitors induce early cell cycle inhibition, followed by redox alterations at cytosol and mitochondria. Massive mitochondrial oxidation and DNA damage induce classical cell death involving mitochondrial transmembrane potential loss and mitochondrial permeabilization. Real-time imaging of the progression of mitochondrial oxidation revealed that treated cells undergo a biphasic mitochondrial redox alteration during telomerase inhibition, emphasizing the potential role of telomerase in the redox regulation at mitochondria. Additionally, silencing of hTERT confirmed its predominant role in maintaining mitochondrial redox homeostasis. Interestingly, the study also demonstrated that anti-apoptotic Bcl-2 family proteins still confer protection against cell death induced by telomerase inhibitors. CONCLUSION The study demonstrates that redox alterations and DNA damage contribute to early cell death by telomerase inhibitors and anti-apoptotic Bcl-2 family proteins confer protection from cell death by their ability to safeguard mitochondria from oxidation damage.
Collapse
Affiliation(s)
- Aparna Geetha Jayaprasad
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud, Thiruvananthapuram 695014, Kerala, India; PhD Program, Manipal Academy of Higher Education (MAHE), Madhav Nagar, Manipal, Karnataka 576104, India
| | - Aneesh Chandrasekharan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud, Thiruvananthapuram 695014, Kerala, India
| | - S P Arun Jyothi
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud, Thiruvananthapuram 695014, Kerala, India
| | - S M John Sam
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud, Thiruvananthapuram 695014, Kerala, India
| | - T R Santhoshkumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud, Thiruvananthapuram 695014, Kerala, India.
| | - M Radhakrishna Pillai
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud, Thiruvananthapuram 695014, Kerala, India.
| |
Collapse
|
17
|
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
|
18
|
Hicks WH, Gattie LC, Traylor JI, Davar D, Najjar YG, Richardson DO TE, McBrayer SK, Abdullah KG. Matched three-dimensional organoids and two-dimensional cell lines of melanoma brain metastases mirror response to targeted molecular therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576318. [PMID: 38328251 PMCID: PMC10849477 DOI: 10.1101/2024.01.18.576318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Purpose Despite significant advances in the treatment paradigm for patients with metastatic melanoma, melanoma brain metastasis (MBM) continues to represent a significant treatment challenge. The study of MBM is limited, in part, by shortcomings in existing preclinical models. Surgically eXplanted Organoids (SXOs) are ex vivo, three-dimensional cultures prepared from primary tissue samples with minimal processing that recapitulate genotypic and phenotypic features of parent tumors and are grown without artificial extracellular scaffolding. We aimed to develop the first matched patient-derived SXO and cell line models of MBM to investigate responses to targeted therapy. Methods MBM SXOs were created by a novel protocol incorporating techniques for establishing glioma and cutaneous melanoma organoids. A BRAFV600K-mutant and BRAF-wildtype MBM sample were collected directly from the operating room for downstream experiments. Organoids were cultured in an optimized culture medium without an artificial extracellular scaffold. Concurrently, matched patient-derived cell lines were created. Drug screens were conducted to assess treatment response in SXOs and cell lines. Results Organoid growth was observed within 3-4 weeks, and MBM SXOs retained histological features of the parent tissue, including pleomorphic epithelioid cells with abundant cytoplasm, large nuclei, focal melanin accumulation, and strong SOX10 positivity. After sufficient growth, organoids could be manually parcellated to increase the number of replicates. Matched SXOs and cell lines demonstrated sensitivity to BRAF and MEK inhibitors. Conclusion Here, we describe the creation of a scaffold-free organoid model of MBM. Further study using SXOs may improve the translational relevance of preclinical studies and enable the study of the metastatic melanoma tumor microenvironment.
Collapse
Affiliation(s)
- William H. Hicks
- Department of Neurosurgery, University of Pittsburgh School of Medicine, 200 Lothrop St, Pittsburgh, PA, 15213, USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Lauren C. Gattie
- Department of Neurosurgery, University of Pittsburgh School of Medicine, 200 Lothrop St, Pittsburgh, PA, 15213, USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Jeffrey I Traylor
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, 75235, USA
- Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Diwakar Davar
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Yana G. Najjar
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Timothy E. Richardson DO
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, 11029, USA
| | - Samuel K. McBrayer
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, 75235, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kalil G. Abdullah
- Department of Neurosurgery, University of Pittsburgh School of Medicine, 200 Lothrop St, Pittsburgh, PA, 15213, USA
- Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| |
Collapse
|
19
|
Wang ZH, Li J, Liu Q, Qian JC, Li QQ, Wang QY, Zeng LT, Li SJ, Gao X, Pan JX, Gao XF, Wu K, Hu GX, Iwakuma T, Cai JP. A modified nucleoside O6-methyl-2'-deoxyguanosine-5'-triphosphate exhibits anti-glioblastoma activity in a caspase-independent manner. Pharmacol Res 2024; 199:106990. [PMID: 37984506 DOI: 10.1016/j.phrs.2023.106990] [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: 10/09/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
Resistance to temozolomide (TMZ), the frontline chemotherapeutic agent for glioblastoma (GBM), has emerged as a formidable obstacle, underscoring the imperative to identify alternative therapeutic strategies to improve patient outcomes. In this study, we comprehensively evaluated a novel agent, O6-methyl-2'-deoxyguanosine-5'-triphosphate (O6-methyl-dGTP) for its anti-GBM activity both in vitro and in vivo. Notably, O6-methyl-dGTP exhibited pronounced cytotoxicity against GBM cells, including those resistant to TMZ and overexpressing O6-methylguanine-DNA methyltransferase (MGMT). Mechanistic investigations revealed that O6-methyl-dGTP could be incorporated into genomic DNA, disrupting nucleotide pools balance, and inducing replication stress, resulting in S-phase arrest and DNA damage. The compound exerted its anti-tumor properties through the activation of AIF-mediated apoptosis and the parthanatos pathway. In vivo studies using U251 and Ln229 cell xenografts supported the robust tumor-inhibitory capacity of O6-methyl-dGTP. In an orthotopic transplantation model with U87MG cells, O6-methyl-dGTP showcased marginally superior tumor-suppressive activity compared to TMZ. In summary, our research, for the first time, underscores the potential of O6-methyl-dGTP as an effective candidate against GBM, laying a robust scientific groundwork for its potential clinical adoption in GBM treatment regimens.
Collapse
Affiliation(s)
- Zi-Hui Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jin Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Qian Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jian-Chang Qian
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qing-Qing Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qing-Yu Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Lv-Tao Zeng
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Si-Jia Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Xin Gao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Jia-Xin Pan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Xu-Fan Gao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Geriatrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Kun Wu
- Wu Xi AppTec (Tianjin) Co., Ltd, China
| | - Guo-Xin Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tomoo Iwakuma
- Children's Mercy Research Institute, Kansas City, MO 64108, USA
| | - Jian-Ping Cai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
20
|
Tu KJ, Stewart CE, Hendrickson PG, Regal JA, Kim SY, Ashley DM, Waitkus MS, Reitman ZJ. Pooled genetic screens to identify vulnerabilities in TERT-promoter-mutant glioblastoma. Oncogene 2023; 42:3274-3286. [PMID: 37741952 PMCID: PMC10615780 DOI: 10.1038/s41388-023-02845-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023]
Abstract
Pooled genetic screens represent a powerful approach to identify vulnerabilities in cancer. Here we used pooled CRISPR/Cas9-based approaches to identify vulnerabilities associated with telomerase reverse transcriptase (TERT) promoter mutations (TPMs) found in >80% of glioblastomas. We first developed a platform to detect perturbations that cause long-term growth defects in a TPM-mutated glioblastoma cell line. However, we could not detect dependencies on either TERT itself or on an E-twenty six transcription (ETS) factor known to activate TPMs. To explore this finding, we cataloged TPM status for 441 cell lines and correlated this with genome-wide screening data. We found that TPM status was not associated with differential dependency on TERT, but that E-twenty six (ETS) transcription factors represent key dependencies in both TPM+ and TPM- lines. Further, we found that TPMs are associated with expression of gene programs regulated by a wide array of ETS-factors in both cell lines and primary glioblastoma tissues. This work contributes a unique TPM cell line reagent, establishes TPM status for many deeply-profiled cell lines, and catalogs TPM-associated vulnerabilities. The results highlight challenges in executing genetic screens to detect TPM-specific vulnerabilities, and suggest redundancy in the genetic network that regulates TPM function with therapeutic implications.
Collapse
Affiliation(s)
- Kevin J Tu
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, 21044, USA
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Connor E Stewart
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Peter G Hendrickson
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Joshua A Regal
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - So Young Kim
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - David M Ashley
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, 27710, USA
- The Preston Robert Tisch Brain Tumor Center at Duke, Durham, NC, 27710, USA
| | - Matthew S Waitkus
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, 27710, USA
- The Preston Robert Tisch Brain Tumor Center at Duke, Durham, NC, 27710, USA
| | - Zachary J Reitman
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, 27710, USA.
- The Preston Robert Tisch Brain Tumor Center at Duke, Durham, NC, 27710, USA.
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
21
|
Chojak R, Fares J, Petrosyan E, Lesniak MS. Cellular senescence in glioma. J Neurooncol 2023; 164:11-29. [PMID: 37458855 DOI: 10.1007/s11060-023-04387-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/01/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION Glioma is the most common primary brain tumor and is often associated with treatment resistance and poor prognosis. Standard treatment typically involves radiotherapy and temozolomide-based chemotherapy, both of which induce cellular senescence-a tumor suppression mechanism. DISCUSSION Gliomas employ various mechanisms to bypass or escape senescence and remain in a proliferative state. Importantly, senescent cells remain viable and secrete a large number of factors collectively known as the senescence-associated secretory phenotype (SASP) that, paradoxically, also have pro-tumorigenic effects. Furthermore, senescent cells may represent one form of tumor dormancy and play a role in glioma recurrence and progression. CONCLUSION In this article, we delineate an overview of senescence in the context of gliomas, including the mechanisms that lead to senescence induction, bypass, and escape. Furthermore, we examine the role of senescent cells in the tumor microenvironment and their role in tumor progression and recurrence. Additionally, we highlight potential therapeutic opportunities for targeting senescence in glioma.
Collapse
Affiliation(s)
- Rafał Chojak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jawad Fares
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Edgar Petrosyan
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA.
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
22
|
Sim HW, Lorrey S, Khasraw M. Advances in Treatment of Isocitrate Dehydrogenase (IDH)-Wildtype Glioblastomas. Curr Neurol Neurosci Rep 2023; 23:263-276. [PMID: 37154886 DOI: 10.1007/s11910-023-01268-0] [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: 03/22/2023] [Indexed: 05/10/2023]
Abstract
PURPOSE OF REVIEW The management of isocitrate dehydrogenase (IDH)-wildtype glioblastomas is an area of unmet need. Despite multimodal therapy incorporating maximal safe resection, radiotherapy, and temozolomide, clinical outcomes remain poor. At disease progression or relapse, available systemic agents such as temozolomide, lomustine, and bevacizumab have limited efficacy. We review the recent advances in the treatment of IDH-wildtype glioblastomas. RECENT FINDINGS A broad repertoire of systemic agents is in the early stages of development, encompassing the areas of precision medicine, immunotherapy, and repurposed medications. The use of medical devices may present opportunities to bypass the blood-brain barrier. Novel clinical trial designs aim to efficiently test treatment options to advance the field. There are a number of emerging treatment options for IDH-wildtype glioblastomas which are undergoing evaluation in clinical trials. Advances in our scientific understanding of IDH-wildtype glioblastomas offer hope and the prospect of incremental improvements in clinical outcomes.
Collapse
Affiliation(s)
- Hao-Wen Sim
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, 2050, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2010, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, NSW, 2050, Australia
| | - Selena Lorrey
- Department of Immunology, Duke University Medical Center, Durham, NC, 27710, USA
- Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Mustafa Khasraw
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, 2050, Australia.
- Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, 27710, USA.
- Duke University School of Medicine, Duke University Medical Center, Box 3624, Durham, NC, 27710, USA.
| |
Collapse
|
23
|
Montella L, Del Gaudio N, Bove G, Cuomo M, Buonaiuto M, Costabile D, Visconti R, Facchini G, Altucci L, Chiariotti L, Della Monica R. Looking Beyond the Glioblastoma Mask: Is Genomics the Right Path? Front Oncol 2022; 12:926967. [PMID: 35875139 PMCID: PMC9306486 DOI: 10.3389/fonc.2022.926967] [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: 04/23/2022] [Accepted: 06/09/2022] [Indexed: 11/15/2022] Open
Abstract
Glioblastomas are the most frequent and malignant brain tumor hallmarked by an invariably poor prognosis. They have been classically differentiated into primary isocitrate dehydrogenase 1 or 2 (IDH1 -2) wild-type (wt) glioblastoma (GBM) and secondary IDH mutant GBM, with IDH wt GBMs being commonly associated with older age and poor prognosis. Recently, genetic analyses have been integrated with epigenetic investigations, strongly implementing typing and subtyping of brain tumors, including GBMs, and leading to the new WHO 2021 classification. GBM genomic and epigenomic profile influences evolution, resistance, and therapeutic responses. However, differently from other tumors, there is a wide gap between the refined GBM profiling and the limited therapeutic opportunities. In addition, the different oncogenes and tumor suppressor genes involved in glial cell transformation, the heterogeneous nature of cancer, and the restricted access of drugs due to the blood–brain barrier have limited clinical advancements. This review will summarize the more relevant genetic alterations found in GBMs and highlight their potential role as potential therapeutic targets.
Collapse
Affiliation(s)
- Liliana Montella
- Oncology Operative Unit, "Santa Maria delle Grazie" Hospital, ASL Napoli 2 NORD-, Pozzuoli, Italy
| | - Nunzio Del Gaudio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Guglielmo Bove
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Mariella Cuomo
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| | - Michela Buonaiuto
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| | - Davide Costabile
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,SEMM-European School of Molecular Medicine, Milano, Italy
| | - Roberta Visconti
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Institute of Experimental Endocrinology and Oncology, Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Gaetano Facchini
- Oncology Operative Unit, "Santa Maria delle Grazie" Hospital, ASL Napoli 2 NORD-, Pozzuoli, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy.,BIOGEM, Ariano Irpino, Italy
| | - Lorenzo Chiariotti
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| | - Rosa Della Monica
- CEINGE Biotecnologie Avanzate scarl, Napoli, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Napoli, Italy
| |
Collapse
|