1
|
Evans L, Trinder S, Dodgshun A, Eisenstat DD, Whittle JR, Hansford JR, Valvi S. IDH-mutant gliomas in children and adolescents - from biology to clinical trials. Front Oncol 2025; 14:1515538. [PMID: 39876890 PMCID: PMC11773619 DOI: 10.3389/fonc.2024.1515538] [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: 10/23/2024] [Accepted: 12/10/2024] [Indexed: 01/31/2025] Open
Abstract
Gliomas account for nearly 30% of all primary central nervous system (CNS) tumors in children and adolescents and young adults (AYA), contributing to significant morbidity and mortality. The updated molecular classification of gliomas defines molecularly diverse subtypes with a spectrum of tumors associated with age-distinct incidence. In adults, gliomas are characterized by the presence or absence of mutations in isocitrate dehydrogenase (IDH), with mutated IDH (mIDH) gliomas providing favorable outcomes and avenues for targeted therapy with the emergence of mIDH inhibitors. Despite their rarity, IDH mutations have been reported in 5-15% of pediatric glioma cases. Those with primary mismatch-repair deficient mIDH astrocytomas (PMMRDIA) have a particularly poor prognosis. Here, we describe the biology of mIDH gliomas and review the literature regarding the emergence of mIDH inhibitors, including clinical trials in adults. Given the paucity of clinical trial data from pediatric patients with mIDH glioma, we propose guidelines for the inclusion of pediatric and AYA patients with gliomas onto prospective trials and expanded access programs as well as the potential of combined mIDH inhibition and immunotherapy in the treatment of patients with PMMRDIA at high risk of progression.
Collapse
Affiliation(s)
- Louise Evans
- Michael Rice Centre for Hematology and Oncology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
| | - Sarah Trinder
- Kids Cancer Centre, Sydney Children’s Hospital, Sydney, NSW, Australia
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Andrew Dodgshun
- Department of Pediatrics, University of Otago, Christchurch, New Zealand
- Children’s Hematology/Oncology Centre, Christchurch Hospital, Christchurch, New Zealand
| | - David D. Eisenstat
- Children’s Cancer Centre, Royal Children’s Hospital, Melbourne, VIC, Australia
- Department of Stem Cell Medicine, Murdoch Children’s Research Institute, Melbourne, VIC, Australia
- Department of Pediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - James R. Whittle
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Personalized Oncology Division, Walter and Eliza Hall Institute (WEHI), Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Jordan R. Hansford
- Michael Rice Centre for Hematology and Oncology, Women’s and Children’s Hospital, North Adelaide, SA, Australia
- Pediatric Neuro-Oncology, Precision Cancer Medicine, South Australia Health and Medical Reseach Institute, Adelaide, SA, Australia
- South Australia ImmunoGENomics Cancer Institute, University of Adelaide, Adelaide, SA, Australia
| | - Santosh Valvi
- Department of Pediatric and Adolescent Oncology/Hematology, Perth Children’s Hospital, Nedlands, WA, Australia
- Brain Tumor Research Program, Telethon Kids Institute, Nedlands, WA, Australia
- School of Medicine, Division of Pediatrics, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
2
|
Wanis HA, Møller H, Ashkan K, Davies EA. Association of IDH1 Mutation and MGMT Promoter Methylation with Clinicopathological Parameters in an Ethnically Diverse Population of Adults with Gliomas in England. Biomedicines 2024; 12:2732. [PMID: 39767640 PMCID: PMC11726743 DOI: 10.3390/biomedicines12122732] [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: 09/30/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 01/16/2025] Open
Abstract
Background: Molecular profiles can predict which patients will respond to current standard treatment and new targeted therapy regimens. Using data from a highly diverse population of approximately three million in Southeast London and Kent, this study aims to evaluate the prevalence of IDH1 mutation and MGMT promoter methylation in the gliomas diagnosed in adult patients and to explore correlations with patients' demographic and clinicopathological characteristics. Methods: Anonymised data on 749 adult patients diagnosed with a glioma in 2015-2019 at King's College Hospital were extracted. Univariable and multivariable logistic regressions were used to estimate odds ratios (ORs) for expressing IDH1 mutation and MGMT promoter methylation, based on each patient's age, sex, ethnicity, histology, tumour location and extent of resection. The Kaplan-Meier method was used to estimate the overall survival functions. Results: A total of 19.5% of cases were IDH1-mutated. Being 39 years and younger (OR 5.48, 95% CI 3.17-9.47), from Asian/Asian British background (OR 3.68, 95% CI 1.05-12.97), having MGMT methylation (OR 15.92, 95% CI 7.30-34.75), an oligodendroglioma diagnosis (OR 7.45, 95% CI 2.90-19.13) and receiving a gross total/total microscopic resection (OR 1.95, 95% CI 1.24-3.08) were each univariately correlated with IDH1 mutation. MGMT methylation association persisted on adjustment (OR 14.13, 95% CI 3.88-51.43). MGMT promoter methylation was seen in 54.3% of gliomas. In the univariate adjusted ORs, being younger than 39 years (OR 2.56, 95% CI 1.48-4.43), female (OR 1.52, 95% CI 1.11-2.08), having IDH1 mutation (OR 15.92, 95% CI 7.30-34.75) and an oligodendroglioma diagnosis (OR 6.20, 95% CI 1.33-28.88) were associated with MGMT methylation. Being female (OR 1.75, 95% CI 1.22-2.51) and having an IDH1 mutation (OR 15.54, 95% CI 4.73-51.05) persisted after adjustment for age, sex, ethnicity, histology, tumour location and extent of resection. IDH1 mutant and MGMT methylated gliomas were associated with frontal lobe location. Survival analysis showed that patients with both IDH1 mutation and MGMT methylation had significantly better survival than those with either molecular marker alone. Over a 3-year period, women with unmethylated MGMT promoters generally had better survival than men with unmethylated MGMT. Conclusion: This study showed that the molecular markers of IDH1 mutation and MGMT promoter methylation were associated with age, sex, Asian/Asian British ethnic group, tumour histology, anatomical location and extent of resection. This study has demonstrated the importance of assessing glioma molecular markers in the clinical setting and the need to stratify patients according to their clinicopathological characteristics.
Collapse
Affiliation(s)
- Hiba A. Wanis
- Cancer Epidemiology and Cancer Services Research, Centre for Cancer, Society & Public Health, Bermondsey Wing, King’s College London, 3rd Floor, Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Henrik Møller
- Cancer Epidemiology and Cancer Services Research, Centre for Cancer, Society & Public Health, Bermondsey Wing, King’s College London, 3rd Floor, Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK
- Danish Centre for Health Services Research, Aalborg University, 9220 Aalborg, Denmark
| | - Keyoumars Ashkan
- Department of Neurosurgery, King’s College Hospital NHS Trust, Denmark Hill, London SE5 9RS, UK
| | - Elizabeth A. Davies
- Cancer Epidemiology and Cancer Services Research, Centre for Cancer, Society & Public Health, Bermondsey Wing, King’s College London, 3rd Floor, Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
3
|
Trejo-Solís C, Serrano-García N, Castillo-Rodríguez RA, Robledo-Cadena DX, Jimenez-Farfan D, Marín-Hernández Á, Silva-Adaya D, Rodríguez-Pérez CE, Gallardo-Pérez JC. Metabolic dysregulation of tricarboxylic acid cycle and oxidative phosphorylation in glioblastoma. Rev Neurosci 2024; 35:813-838. [PMID: 38841811 DOI: 10.1515/revneuro-2024-0054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
Glioblastoma multiforme (GBM) exhibits genetic alterations that induce the deregulation of oncogenic pathways, thus promoting metabolic adaptation. The modulation of metabolic enzyme activities is necessary to generate nucleotides, amino acids, and fatty acids, which provide energy and metabolic intermediates essential for fulfilling the biosynthetic needs of glioma cells. Moreover, the TCA cycle produces intermediates that play important roles in the metabolism of glucose, fatty acids, or non-essential amino acids, and act as signaling molecules associated with the activation of oncogenic pathways, transcriptional changes, and epigenetic modifications. In this review, we aim to explore how dysregulated metabolic enzymes from the TCA cycle and oxidative phosphorylation, along with their metabolites, modulate both catabolic and anabolic metabolic pathways, as well as pro-oncogenic signaling pathways, transcriptional changes, and epigenetic modifications in GBM cells, contributing to the formation, survival, growth, and invasion of glioma cells. Additionally, we discuss promising therapeutic strategies targeting key players in metabolic regulation. Therefore, understanding metabolic reprogramming is necessary to fully comprehend the biology of malignant gliomas and significantly improve patient survival.
Collapse
Affiliation(s)
- Cristina Trejo-Solís
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Neurobiología Molecular y Celular, Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico
| | - Norma Serrano-García
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Neurobiología Molecular y Celular, Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico
| | - Rosa Angelica Castillo-Rodríguez
- CICATA Unidad Morelos, Instituto Politécnico Nacional, Boulevard de la Tecnología, 1036 Z-1, P 2/2, Atlacholoaya, Xochitepec 62790, Mexico
| | - Diana Xochiquetzal Robledo-Cadena
- Departamento de Fisiopatología Cardio-Renal, Departamento de Bioquímica, Instituto Nacional de Cardiología, Ciudad de México 14080, Mexico
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Coyoacán, 04510, Ciudad de México, Mexico
| | - Dolores Jimenez-Farfan
- Laboratorio de Inmunología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico
| | - Álvaro Marín-Hernández
- Departamento de Fisiopatología Cardio-Renal, Departamento de Bioquímica, Instituto Nacional de Cardiología, Ciudad de México 14080, Mexico
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Coyoacán, 04510, Ciudad de México, Mexico
| | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Neurobiología Molecular y Celular, Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico
| | - Citlali Ekaterina Rodríguez-Pérez
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Neurobiología Molecular y Celular, Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico
| | - Juan Carlos Gallardo-Pérez
- Departamento de Fisiopatología Cardio-Renal, Departamento de Bioquímica, Instituto Nacional de Cardiología, Ciudad de México 14080, Mexico
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Coyoacán, 04510, Ciudad de México, Mexico
| |
Collapse
|
4
|
Kinslow CJ, Roy S, Iwamoto FM, Brown PD, DeStephano DM, Canoll PD, Qureshi SS, Gallito M, Sisti MB, Bruce JN, Horowitz DP, Kachnic LA, Neugut AI, Yu JB, Mehta MP, Cheng SK, Wang TJC. The IDH paradox: Meta-analysis of alkylating chemotherapy in IDH-wild type and -mutant lower grade gliomas. Neuro Oncol 2024; 26:1839-1849. [PMID: 38943513 PMCID: PMC11449043 DOI: 10.1093/neuonc/noae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND IDH-wild type (-wt) status is a prerequisite for the diagnosis of glioblastoma (GBM); however, IDH-wt gliomas with low-grade or anaplastic morphology have historically been excluded from GBM trials and may represent a distinct prognostic entity. While alkylating agent chemotherapy improves overall survival (OS) and progression-free survival (PFS) for IDH-wt GBM and also IDH-mutant gliomas, irrespective of grade, the benefit for IDH-wt diffuse histologic lower-grade gliomas is unclear. METHODS We performed a meta-analysis of randomized clinical trials for World Health Organization (WHO) grades 2-3 gliomas (2009 to present) to determine the effect of alkylating chemotherapy on IDH-wt and -mutant gliomas using a random-effects model with inverse-variance pooling. RESULTS We identified 6 trials with 1204 patients (430 IDH-wt, 774 IDH-mutant) that evaluated alkylating chemoradiotherapy versus radiotherapy alone, allowing us to perform an analysis focused on the value of adding alkylating chemotherapy to radiotherapy. For patients with IDH-wt tumors, alkylating chemotherapy added to radiotherapy was associated with improved PFS (HR:0.77 [95% CI: 0.62-0.97], P = .03) but not OS (HR:0.87 [95% CI: 0.64-1.18], P = .17). For patients with IDH-mutant tumors, alkylating chemotherapy added to radiotherapy improved both OS (HR:0.52 [95% CI: 0.42-0.64], P < .001) and PFS (HR = 0.47 [95% CI: 0.39-0.57], P < .001) compared to radiotherapy alone. The magnitude of benefit was similar for IDH-mutant gliomas with or without 1p19q-codeletion. CONCLUSIONS Alkylating chemotherapy reduces mortality by 48% and progression by 53% for patients with IDH-mutant gliomas. Optimal management of IDH-wt diffuse histologic lower-grade gliomas remains to be determined, as there is little evidence supporting an OS benefit from alkylating chemotherapy.
Collapse
Affiliation(s)
- Connor J Kinslow
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Soumyajit Roy
- Department of Radiation Oncology, Rush University Medical Center, Chicago, Illinois, USA
| | - Fabio M Iwamoto
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - David M DeStephano
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Peter D Canoll
- Departments of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Summer S Qureshi
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Matthew Gallito
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Michael B Sisti
- Department of Neurological Surgery, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - David P Horowitz
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Lisa A Kachnic
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Alfred I Neugut
- Department of Medicine, Vagelos College of Physicians and Surgeons, and Department of Epidemiology, Mailman School of Public Health, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - James B Yu
- Department of Medical Oncology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Radiation Oncology Medical Oncology, Saint Francis Hospital, Hartford, Connecticut, USA
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Simon K Cheng
- Department of Radiation Oncology, James J. Peters Veterans Affairs Medical Center, Bronx, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| | - Tony J C Wang
- Herbert Irving Comprehensive Cancer Center, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
- Department of Radiation Oncology, Columbia University Vagelos College of Physicians and Surgeons and NewYork-Presbyterian, New York, New York, USA
| |
Collapse
|
5
|
Huang YR, Fan HQ, Kuang YY, Wang P, Lu S. The Relationship Between the Molecular Phenotypes of Brain Gliomas and the Imaging Features and Sensitivity of Radiotherapy and Chemotherapy. Clin Oncol (R Coll Radiol) 2024; 36:541-551. [PMID: 38821723 DOI: 10.1016/j.clon.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/28/2024] [Accepted: 05/10/2024] [Indexed: 06/02/2024]
Abstract
Gliomas are the most common primary malignant tumors of the brain, accounting for about 80% of all central nervous system malignancies. With the development of molecular biology, the molecular phenotypes of gliomas have been shown to be closely related to the process of diagnosis and treatment. The molecular phenotype of glioma also plays an important role in guiding treatment plans and evaluating treatment effects and prognosis. However, due to the heterogeneity of the tumors and the trauma associated with the surgical removal of tumor tissue, the application of molecular phenotyping in glioma is limited. With the development of imaging technology, functional magnetic resonance imaging (MRI) can provide structural and function information about tumors in a noninvasive and radiation-free manner. MRI is very important for the diagnosis of intracranial lesions. In recent years, with the development of the technology for tumor molecular diagnosis and imaging, the use of molecular phenotype information and imaging procedures to evaluate the treatment outcome of tumors has become a hot topic. By reviewing the related literature on glioma treatment and molecular typing that has been published in the past 20 years, and referring to the latest 2020 NCCN treatment guidelines, summarizing the imaging characteristic and sensitivity of radiotherapy and chemotherapy of different molecular phenotypes of glioma. In this article, we briefly review the imaging characteristics of different molecular phenotypes in gliomas and their relationship with radiosensitivity and chemosensitivity of gliomas.
Collapse
Affiliation(s)
- Y-R Huang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - H-Q Fan
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Y-Y Kuang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - P Wang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - S Lu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| |
Collapse
|
6
|
Grimi A, Bono BC, Lazzarin SM, Marcheselli S, Pessina F, Riva M. Gliomagenesis, Epileptogenesis, and Remodeling of Neural Circuits: Relevance for Novel Treatment Strategies in Low- and High-Grade Gliomas. Int J Mol Sci 2024; 25:8953. [PMID: 39201639 PMCID: PMC11354416 DOI: 10.3390/ijms25168953] [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: 05/07/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 09/02/2024] Open
Abstract
Gliomas present a complex challenge in neuro-oncology, often accompanied by the debilitating complication of epilepsy. Understanding the biological interaction and common pathways between gliomagenesis and epileptogenesis is crucial for improving the current understanding of tumorigenesis and also for developing effective management strategies. Shared genetic and molecular mechanisms, such as IDH mutations and dysregulated glutamate signaling, contribute to both tumor progression and seizure development. Targeting these pathways, such as through direct inhibition of mutant IDH enzymes or modulation of glutamate receptors, holds promise for improving patient outcomes. Additionally, advancements in surgical techniques, like supratotal resection guided by connectomics, offer opportunities for maximally safe tumor resection and enhanced seizure control. Advanced imaging modalities further aid in identifying epileptogenic foci and tailoring treatment approaches based on the tumor's metabolic characteristics. This review aims to explore the complex interplay between gliomagenesis, epileptogenesis, and neural circuit remodeling, offering insights into shared molecular pathways and innovative treatment strategies to improve outcomes for patients with gliomas and associated epilepsy.
Collapse
Affiliation(s)
- Alessandro Grimi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Beatrice C. Bono
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | | | | | - Federico Pessina
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Marco Riva
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| |
Collapse
|
7
|
Tecik M, Adan A. Emerging DNA Methylome Targets in FLT3-ITD-Positive Acute Myeloid Leukemia: Combination Therapy with Clinically Approved FLT3 Inhibitors. Curr Treat Options Oncol 2024; 25:719-751. [PMID: 38696033 PMCID: PMC11222205 DOI: 10.1007/s11864-024-01202-7] [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] [Accepted: 04/01/2024] [Indexed: 07/04/2024]
Abstract
OPINION STATEMENT The internal tandem duplication (ITD) mutation of the FMS-like receptor tyrosine kinase 3 (FLT3-ITD) is the most common mutation observed in approximately 30% of acute myeloid leukemia (AML) patients. It represents poor prognosis due to continuous activation of downstream growth-promoting signaling pathways such as STAT5 and PI3K/AKT. Hence, FLT3 is considered an attractive druggable target; selective small FLT3 inhibitors (FLT3Is), such as midostaurin and quizartinib, have been clinically approved. However, patients possess generally poor remission rates and acquired resistance when FLT3I used alone. Various factors in patients could cause these adverse effects including altered epigenetic regulation, causing mainly abnormal gene expression patterns. Epigenetic modifications are required for hematopoietic stem cell (HSC) self-renewal and differentiation; however, critical driver mutations have been identified in genes controlling DNA methylation (such as DNMT3A, TET2, IDH1/2). These regulators cause leukemia pathogenesis and affect disease diagnosis and prognosis when they co-occur with FLT3-ITD mutation. Therefore, understanding the role of different epigenetic alterations in FLT3-ITD AML pathogenesis and how they modulate FLT3I's activity is important to rationalize combinational treatment approaches including FLT3Is and modulators of methylation regulators or pathways. Data from ongoing pre-clinical and clinical studies will further precisely define the potential use of epigenetic therapy together with FLT3Is especially after characterized patients' mutational status in terms of FLT3 and DNA methlome regulators.
Collapse
Affiliation(s)
- Melisa Tecik
- Bioengineering Program, Graduate School of Engineering and Science, Abdullah Gul University, Kayseri, Turkey
| | - Aysun Adan
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri, Turkey.
| |
Collapse
|
8
|
Wu Q, Berglund AE, Macaulay RJ, Etame AB. The Role of Mesenchymal Reprogramming in Malignant Clonal Evolution and Intra-Tumoral Heterogeneity in Glioblastoma. Cells 2024; 13:942. [PMID: 38891074 PMCID: PMC11171993 DOI: 10.3390/cells13110942] [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: 04/30/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma (GBM) is the most common yet uniformly fatal adult brain cancer. Intra-tumoral molecular and cellular heterogeneities are major contributory factors to therapeutic refractoriness and futility in GBM. Molecular heterogeneity is represented through molecular subtype clusters whereby the proneural (PN) subtype is associated with significantly increased long-term survival compared to the highly resistant mesenchymal (MES) subtype. Furthermore, it is universally recognized that a small subset of GBM cells known as GBM stem cells (GSCs) serve as reservoirs for tumor recurrence and progression. The clonal evolution of GSC molecular subtypes in response to therapy drives intra-tumoral heterogeneity and remains a critical determinant of GBM outcomes. In particular, the intra-tumoral MES reprogramming of GSCs using current GBM therapies has emerged as a leading hypothesis for therapeutic refractoriness. Preventing the intra-tumoral divergent evolution of GBM toward the MES subtype via new treatments would dramatically improve long-term survival for GBM patients and have a significant impact on GBM outcomes. In this review, we examine the challenges of the role of MES reprogramming in the malignant clonal evolution of glioblastoma and provide future perspectives for addressing the unmet therapeutic need to overcome resistance in GBM.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Anders E. Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Robert J. Macaulay
- Departments of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Arnold B. Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
9
|
Douglas C, Lomeli N, Vu T, Pham J, Bota DA. WITHDRAWN: LonP1 Drives Proneural Mesenchymal Transition in IDH1-R132H Diffuse Glioma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.13.536817. [PMID: 37131765 PMCID: PMC10153221 DOI: 10.1101/2023.04.13.536817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The authors have withdrawn their manuscript owing to massive revision and data validation. Therefore, the authors do not wish this work to be cited as reference for the project. If you have any questions, please contact the corresponding author.
Collapse
|
10
|
Gu Q, An Y, Xu M, Huang X, Chen X, Li X, Shan H, Zhang M. Disulfidptosis, A Novel Cell Death Pathway: Molecular Landscape and Therapeutic Implications. Aging Dis 2024:AD.2024.0083. [PMID: 38739940 DOI: 10.14336/ad.2024.0083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
Programmed cell death is pivotal for several physiological processes, including immune defense. Further, it has been implicated in the pathogenesis of developmental disorders and the onset of numerous diseases. Multiple modes of programmed cell death, including apoptosis, pyroptosis, necroptosis, and ferroptosis, have been identified, each with their own unique characteristics and biological implications. In February 2023, Liu Xiaoguang and his team discovered "disulfidptosis," a novel pathway of programmed cell death. Their findings demonstrated that disulfidptosis is triggered in glucose-starved cells exhibiting high expression of a protein called SLC7A11. Furthermore, disulfidptosis is marked by a drastic imbalance in the NADPH/NADP+ ratio and the abnormal accumulation of disulfides like cystine. These changes ultimately lead to the destabilization of the F-actin network, causing cell death. Given that high SLC7A11 expression is a key feature of certain cancers, these findings indicate that disulfidptosis could serve as the basis of innovative anti-cancer therapies. Hence, this review delves into the discovery of disulfidptosis, its underlying molecular mechanisms and metabolic regulation, and its prospective applications in disease treatment.
Collapse
Affiliation(s)
- Qiuyang Gu
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| | - Yumei An
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| | - Mingyuan Xu
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| | - Xinqi Huang
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| | - Xueshi Chen
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| | - Xianzhe Li
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| | - Haiyan Shan
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Mingyang Zhang
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, China
| |
Collapse
|
11
|
Lin H, Liu C, Hu A, Zhang D, Yang H, Mao Y. Understanding the immunosuppressive microenvironment of glioma: mechanistic insights and clinical perspectives. J Hematol Oncol 2024; 17:31. [PMID: 38720342 PMCID: PMC11077829 DOI: 10.1186/s13045-024-01544-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Glioblastoma (GBM), the predominant and primary malignant intracranial tumor, poses a formidable challenge due to its immunosuppressive microenvironment, thereby confounding conventional therapeutic interventions. Despite the established treatment regimen comprising surgical intervention, radiotherapy, temozolomide administration, and the exploration of emerging modalities such as immunotherapy and integration of medicine and engineering technology therapy, the efficacy of these approaches remains constrained, resulting in suboptimal prognostic outcomes. In recent years, intensive scrutiny of the inhibitory and immunosuppressive milieu within GBM has underscored the significance of cellular constituents of the GBM microenvironment and their interactions with malignant cells and neurons. Novel immune and targeted therapy strategies have emerged, offering promising avenues for advancing GBM treatment. One pivotal mechanism orchestrating immunosuppression in GBM involves the aggregation of myeloid-derived suppressor cells (MDSCs), glioma-associated macrophage/microglia (GAM), and regulatory T cells (Tregs). Among these, MDSCs, though constituting a minority (4-8%) of CD45+ cells in GBM, play a central component in fostering immune evasion and propelling tumor progression, angiogenesis, invasion, and metastasis. MDSCs deploy intricate immunosuppressive mechanisms that adapt to the dynamic tumor microenvironment (TME). Understanding the interplay between GBM and MDSCs provides a compelling basis for therapeutic interventions. This review seeks to elucidate the immune regulatory mechanisms inherent in the GBM microenvironment, explore existing therapeutic targets, and consolidate recent insights into MDSC induction and their contribution to GBM immunosuppression. Additionally, the review comprehensively surveys ongoing clinical trials and potential treatment strategies, envisioning a future where targeting MDSCs could reshape the immune landscape of GBM. Through the synergistic integration of immunotherapy with other therapeutic modalities, this approach can establish a multidisciplinary, multi-target paradigm, ultimately improving the prognosis and quality of life in patients with GBM.
Collapse
Affiliation(s)
- Hao Lin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Chaxian Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Ankang Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Duanwu Zhang
- Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
12
|
Gunn K, Losman JA. Isocitrate Dehydrogenase Mutations in Cancer: Mechanisms of Transformation and Metabolic Liability. Cold Spring Harb Perspect Med 2024; 14:a041537. [PMID: 38191174 PMCID: PMC11065172 DOI: 10.1101/cshperspect.a041537] [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: 01/10/2024]
Abstract
Isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) are metabolic enzymes that interconvert isocitrate and 2-oxoglutarate (2OG). Gain-of-function mutations in IDH1 and IDH2 occur in a number of cancers, including acute myeloid leukemia, glioma, cholangiocarcinoma, and chondrosarcoma. These mutations cripple the wild-type activity of IDH and cause the enzymes to catalyze a partial reverse reaction in which 2OG is reduced but not carboxylated, resulting in production of the (R)-enantiomer of 2-hydroxyglutarate ((R)-2HG). (R)-2HG accumulation in IDH-mutant tumors results in profound dysregulation of cellular metabolism. The most well-characterized oncogenic effects of (R)-2HG involve the dysregulation of 2OG-dependent epigenetic tumor-suppressor enzymes. However, (R)-2HG has many other effects in IDH-mutant cells, some that promote transformation and others that induce metabolic dependencies. Herein, we review how cancer-associated IDH mutations impact epigenetic regulation and cellular metabolism and discuss how these effects can potentially be leveraged to therapeutically target IDH-mutant tumors.
Collapse
Affiliation(s)
- Kathryn Gunn
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Julie-Aurore Losman
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| |
Collapse
|
13
|
Dvorakova K, Skarkova V, Vitovcova B, Soukup J, Vosmikova H, Pleskacova Z, Skarka A, Bartos MC, Krupa P, Kasparova P, Petera J, Rudolf E. Expression of STAT3 and hypoxia markers in long-term surviving malignant glioma patients. BMC Cancer 2024; 24:509. [PMID: 38654280 PMCID: PMC11036726 DOI: 10.1186/s12885-024-12221-w] [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: 11/22/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Glioblastoma is a malignant and aggressive type of central nevous system malignancy characterized by many distinct biological features including extensive hypoxia. Hypoxia in glioblatoma associates with complex signaling patterns including activation of several pathways such as MAPK, PI3K-AKT/mTOR and IL-6/JAK/STAT3 with the master regulator HIF-1, which in turn drive particular tumor behaviors determining, in the end, treatment outcomes and patients fate. Thus, the present study was designed to investigate the expression of selected hypoxia related factors including STAT3 in a small set of long-term surviving glioma patients. METHODS The expression of selected hypoxia related factors including STAT3 was evaluated in a time series of formalin fixed paraffin embedded and cryopreserved glioma samples from repeatedly resected patients. In addition, comparative studies were also conducted on primary glioma cells derived from original patient samples, stabilized glioma cell lines and tumor-xenograft mice model. Obtained data were correlated with clinical findings too. RESULTS Glioblastoma samples of the analyzed patients displayed heterogeneity in the expression of hypoxia- related and EMT markers with most interesting trend being observed in pSTAT3. This heterogeneity was subsequently confirmed in other employed models (primocultures derived from glioblastoma tissue resections, cryopreserved tumor specimens, stabilized glioblastoma cell line in vitro and in vivo) and concerned, in particular, STAT3 expression which remained stable. In addition, subsequent studies on the role of STAT3 in the context of glioblastoma hypoxia demonstrated opposing effects of its deletion on cell viability as well as the expression of hypoxia and EMT markers. CONCLUSIONS Our results suport the importance of STAT3 expression and activity in the context of hypoxia in malignant glioblastoma long-term surviving glioma patients while emphasizing heterogeneity of biological outcomes in varying employed tumor models.
Collapse
Affiliation(s)
- Katerina Dvorakova
- Department of Medical Biology and Genetics, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Veronika Skarkova
- Department of Medical Biology and Genetics, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Barbora Vitovcova
- Department of Medical Biology and Genetics, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Jiri Soukup
- The Fingerland Department of Pathology, Faculty of Medicine n Hradec Kralove, Charles University, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
- Department of Pathology, Military University Hospital Prague, Prague, Czech Republic
- Department of Pathology, First Faculty of Medicine, Charles University, General University Hospital in Prague, Prague, Czech Republic
| | - Hana Vosmikova
- The Fingerland Department of Pathology, Faculty of Medicine n Hradec Kralove, Charles University, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Zuzana Pleskacova
- Department of Oncology and Radiotherapy, Faculty of Medicine in Hradec Kralove, Charles University, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Adam Skarka
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Michael Christian Bartos
- Department of Neurosurgery, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Petr Krupa
- Department of Neurosurgery, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Petra Kasparova
- The Fingerland Department of Pathology, Faculty of Medicine n Hradec Kralove, Charles University, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jiri Petera
- Department of Oncology and Radiotherapy, Faculty of Medicine in Hradec Kralove, Charles University, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Emil Rudolf
- Department of Medical Biology and Genetics, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.
| |
Collapse
|
14
|
El-Kamand S, Quinn JW, Sareen H, Becker T, Wong-Erasmus M, Cowley M. CRUX, a platform for visualising, exploring and analysing cancer genome cohort data. NAR Genom Bioinform 2024; 6:lqae003. [PMID: 38304083 PMCID: PMC10833466 DOI: 10.1093/nargab/lqae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/11/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024] Open
Abstract
To better understand how tumours develop, identify prognostic biomarkers and find new treatments, researchers have generated vast catalogues of cancer genome data. However, these datasets are complex, so interpreting their important features requires specialized computational skills and analytical tools, which presents a significant technical challenge. To address this, we developed CRUX, a platform for exploring genomic data from cancer cohorts. CRUX enables researchers to perform common analyses including cohort comparisons, biomarker discovery, survival analysis, and to create visualisations including oncoplots and lollipop charts. CRUX simplifies cancer genome analysis in several ways: (i) it has an easy-to-use graphical interface; (ii) it enables users to create custom cohorts, as well as analyse precompiled public and private user-created datasets; (iii) it allows analyses to be run locally to address data privacy concerns (though an online version is also available) and (iv) it makes it easy to use additional specialized tools by exporting data in the correct formats. We showcase CRUX's capabilities with case studies employing different types of cancer genome analysis, demonstrating how it can be used flexibly to generate valuable insights into cancer biology. CRUX is freely available at https://github.com/CCICB/CRUX and https://ccicb.shinyapps.io/crux (DOI: 10.5281/zenodo.8015714).
Collapse
Affiliation(s)
- Sam El-Kamand
- Children's Cancer Institute, Randwick, NSW 2031, Australia
| | | | - Heena Sareen
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW 2170, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool, NSW 2170, Australia
| | - Therese M Becker
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, 1 Campbell St, Liverpool, NSW 2170, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool, NSW 2170, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Marie Wong-Erasmus
- Children's Cancer Institute, Randwick, NSW 2031, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| | - Mark J Cowley
- Children's Cancer Institute, Randwick, NSW 2031, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
15
|
Han X, Zhou H, Sun W, Hou L, Wang Y, Wang H, Lv Z, Xue X. IDH1 R132H mutation increases radiotherapy efficacy and a 4-gene radiotherapy-related signature of WHO grade 4 gliomas. Sci Rep 2023; 13:19659. [PMID: 37952042 PMCID: PMC10640646 DOI: 10.1038/s41598-023-46335-1] [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: 10/17/2022] [Accepted: 10/31/2023] [Indexed: 11/14/2023] Open
Abstract
The prognosis for the WHO grade 4 IDH-mutant astrocytoma is better than IDH-wildtype glioblastoma (GBM) patients. The purpose of this study is to explore the potential mechanism of how IDH1 mutation can increase the efficacy of radiotherapy and to establish a risk-score model to predict the efficacy of radiotherapy in WHO grade 4 gliomas. First, we conducted experimental study on the effect of IDH1R132H mutation on glioma cells in vitro. Radiosensitivity of glioma cells was detected by γ-H2AX after 5 Gy radiation. Cell proliferation, migration and invasion were determined respectively by CCK-8, EDU, monolayer cell migration scratch assay and Transwell assay. Then we analyzed IDH1 gene status and the survival of WHO grade 4 glioma patients received radiotherapy in our center and verified our results by analyzing CGGA and TCGA database. For the risk-score model, we use CGGA data to find genetic differences between WHO grade 4 IDH-mutant astrocytoma and IDH-wildtype GBM patients, and determined a 4-gene radiotherapy-related signature through survival analysis by R software. Evaluation and verification through different glioma validation sets and different statistical methods. For in vitro experiments, we established glioma cells stably overexpressing IDH1 wild-type and IDH1-mutant proteins. γ-H2AX assay showed that IDH1-mutant glioma cells had higher radiosensitivity than wild-type. CCK-8 and EDU assay showed that proliferation capacity of IDH1-mutant glioma cells declined. Transwell assay and monolayer cell migration scratch assay also showed that IDH1-mutant glioma cells reduced migration and invasion capabilities. Among the 83 WHO grade 4 glioma patients who received radiotherapy in our center, WHO grade 4 IDH-mutant astrocytoma patients had longer OS and PFS versus IDH-wildtype GBM (P = 0.0336, P = 0.0324, respectively). TCGA and CGGA database analysis had the similar results. Through complex analysis of CGGA and TCGA databases, we established a risk-model that can predict the efficacy of radiotherapy for WHO grade 4 glioma patients. The 4-gene radiotherapy-related signature including ADD3, GRHPR, RHBDL1 and SLC9A9. Patients in the high-risk group had worse OS compared to low-risk group (P = 0.0001). High- and low-risk groups of patients receiving radiotherapy have significant survival differences, while patients who did not receive radiotherapy have no survival difference both in CGGA and TCGA databases. WHO grade 4 IDH-mutant astrocytoma is more radiosensitive than IDH-wildtype GBM patients. Our 4-gene radiotherapy-related signature can predict the radiation efficacy of WHO grade 4 glioma patients, and it may provide some reference for clinical treatment options.
Collapse
Affiliation(s)
- Xuetao Han
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, China
| | - Huandi Zhou
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, China
- Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
- Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Wei Sun
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, China
| | - Liubing Hou
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, China
- Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Yanqiang Wang
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, China
| | - Hong Wang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Zhongqiang Lv
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, China.
| | - Xiaoying Xue
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, China.
- Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
| |
Collapse
|
16
|
Penkova A, Kuziakova O, Gulaia V, Tiasto V, Goncharov NV, Lanskikh D, Zhmenia V, Baklanov I, Farniev V, Kumeiko V. Comprehensive clinical assays for molecular diagnostics of gliomas: the current state and future prospects. Front Mol Biosci 2023; 10:1216102. [PMID: 37908227 PMCID: PMC10613994 DOI: 10.3389/fmolb.2023.1216102] [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: 06/08/2023] [Accepted: 09/04/2023] [Indexed: 11/02/2023] Open
Abstract
Glioma is one of the most intractable types of cancer, due to delayed diagnosis at advanced stages. The clinical symptoms of glioma are unclear and due to a variety of glioma subtypes, available low-invasive testing is not effective enough to be introduced into routine medical laboratory practice. Therefore, recent advances in the clinical diagnosis of glioma have focused on liquid biopsy approaches that utilize a wide range of techniques such as next-generation sequencing (NGS), droplet-digital polymerase chain reaction (ddPCR), and quantitative PCR (qPCR). Among all techniques, NGS is the most advantageous diagnostic method. Despite the rapid cheapening of NGS experiments, the cost of such diagnostics remains high. Moreover, high-throughput diagnostics are not appropriate for molecular profiling of gliomas since patients with gliomas exhibit only a few diagnostic markers. In this review, we highlighted all available assays for glioma diagnosing for main pathogenic glioma DNA sequence alterations. In the present study, we reviewed the possibility of integrating routine molecular methods into the diagnosis of gliomas. We state that the development of an affordable assay covering all glioma genetic aberrations could enable early detection and improve patient outcomes. Moreover, the development of such molecular diagnostic kits could potentially be a good alternative to expensive NGS-based approaches.
Collapse
Affiliation(s)
- Alina Penkova
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Olga Kuziakova
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Valeriia Gulaia
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Vladlena Tiasto
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Nikolay V. Goncharov
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- A. V. Zhirmunsky National Scientific Center of Marine Biology, FEB RAS, Vladivostok, Russia
| | - Daria Lanskikh
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Valeriia Zhmenia
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Ivan Baklanov
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- A. V. Zhirmunsky National Scientific Center of Marine Biology, FEB RAS, Vladivostok, Russia
| | - Vladislav Farniev
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Vadim Kumeiko
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- A. V. Zhirmunsky National Scientific Center of Marine Biology, FEB RAS, Vladivostok, Russia
| |
Collapse
|
17
|
Uson Junior PLS, Borad MJ. Clinical Utility of Ivosidenib in the Treatment of IDH1-Mutant Cholangiocarcinoma: Evidence To Date. Cancer Manag Res 2023; 15:1025-1031. [PMID: 37746315 PMCID: PMC10516216 DOI: 10.2147/cmar.s326060] [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: 04/27/2023] [Accepted: 08/31/2023] [Indexed: 09/26/2023] Open
Abstract
Ivosidenib is an isocitrate dehydrogenase 1 (IDH1) inhibitor that is FDA approved for patients with IDH1 mutation and acute myeloid leukemia and previously treated locally advanced or metastatic cholangiocarcinoma. In the Phase III trial ClarIDHy ivosidenib improved progression-free survival, 2.7 months versus 1.4 months (p < 0.0001) and overall survival (OS), median OS was 10.8 months for ivosidenib and 9.7 months for the placebo arm (p = 0.06) for patients with previously treated and IDH1 mutated cholangiocarcinoma. In this review article, we will address the mechanism of action of ivosidenib and data from early trials and safety from the randomized trial in cholangiocarcinoma. As a conclusion, future perspectives of IDH1 inhibition in IDH1 mutated tumors and possible strategies of sequencing and combinations will be reviewed and discussed.
Collapse
Affiliation(s)
- Pedro Luiz Serrano Uson Junior
- Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ, USA
- HCOR, Hospital do Coração, Sao Paulo, Brazil
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Mitesh J Borad
- Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
18
|
Yin WJ. A bacterial enzyme may correct 2-HG accumulation in human cancers. Front Oncol 2023; 13:1235191. [PMID: 37546420 PMCID: PMC10399246 DOI: 10.3389/fonc.2023.1235191] [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: 06/05/2023] [Accepted: 06/30/2023] [Indexed: 08/08/2023] Open
Abstract
A significant proportion of lower-grade glioma as well as many other types of human cancers are associated with neomorphic mutations in IDH1/2 genes (mIDH1/2). These mutations lead to an aberrant accumulation of 2-hydroxyglutarate (2-HG). Interestingly, even cancers without mIDH1/2 can exhibit increased levels of 2-HG due to factors like hypoxia and extracellular acidity. Mounting evidence demonstrates that 2-HG competitively inhibits α-ketoglutarate dependent enzymes, such as JmjC-domain-containing histone demethylases (JHDMs), ten-eleven translocation enzymes (TETs), and various dioxygenases (e.g., RNA m6A demethylases and prolyl hydroxylases). Consequently, the hypermethylation of DNA, RNA, and histones, and the abnormal activities of hypoxia-inducible factors (HIFs) have profound impacts on the establishment of cancer metabolism and microenvironment, which promote tumor progression. This connection between the oncometabolite 2-HG and glioma holds crucial implications for treatments targeting this disease. Here, I hypothesize that an ectopic introduction of a bacterial 2-hydroxyglutarate synthase (2-HG synthase) enzyme into cancer cells with 2-HG accumulation could serve as a promising enzyme therapy for glioma and other types of cancers. While absent in human metabolism, 2-HG synthase in bacterial species catalyzes the conversion of 2-HG into propionyl-CoA and glyoxylate, two metabolites that potentially possess anti-tumor effects. For a broad spectrum of human cancers with 2-HG accumulation, 2-HG synthase-based enzyme therapy holds the potential to not only correct 2-HG induced cancer metabolism but also transform an oncometabolite into metabolic challenges within cancer cells.
Collapse
Affiliation(s)
- William J. Yin
- Oconee County High School, Watkinsville, GA, United States
- Bio-Imaging Research Center, The University of Georgia, Athens, GA, United States
| |
Collapse
|
19
|
Lathoria K, Gowda P, Umdor SB, Patrick S, Suri V, Sen E. PRMT1 driven PTX3 regulates ferritinophagy in glioma. Autophagy 2023; 19:1997-2014. [PMID: 36647288 PMCID: PMC10283415 DOI: 10.1080/15548627.2023.2165757] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/18/2023] Open
Abstract
Mutations in the Krebs cycle enzyme IDH1 (isocitrate dehydrogenase (NADP(+)) 1) are associated with better prognosis in gliomas. Though IDH1 mutant (IDH1R132H) tumors are characterized by their antiproliferative signatures maintained through hypermethylation of DNA and chromatin, mechanisms affecting cell death pathways in these tumors are not well elucidated. On investigating the crosstalk between the IDH1 mutant epigenome, ferritinophagy and inflammation, diminished expression of PRMT1 (protein arginine methyltransferase 1) and its associated asymmetric dimethyl epigenetic mark H4R3me2a was observed in IDH1R132H gliomas. Reduced expression of PRMT1 was concurrent with diminished levels of PTX3, a key secretory factor involved in cancer-related inflammation. Lack of PRMT1 H4R3me2a in IDH1 mutant glioma failed to epigenetically activate the expression of PTX3 with a reduction in YY1 (YY1 transcription factor) binding on its promoter. Transcriptional activation and subsequent secretion of PTX3 from cells was required for maintaining macroautophagic/autophagic balance as pharmacological or genetic ablation of PTX3 secretion in wild-type IDH1 significantly increased autophagic flux. Additionally, PTX3-deficient IDH1 mutant gliomas exhibited heightened autophagic signatures. Furthermore, we demonstrate that the PRMT1-PTX3 axis is important in regulating the levels of ferritin genes/iron storage and inhibition of this axis triggered ferritinophagic flux. This study highlights the conserved role of IDH1 mutants in augmenting ferritinophagic flux in gliomas irrespective of genetic landscape through inhibition of the PRMT1-PTX3 axis. This is the first study describing ferritinophagy in IDH1 mutant gliomas with mechanistic details. Of clinical importance, our study suggests that the PRMT1-PTX3 ferritinophagy regulatory circuit could be exploited for therapeutic gains.Abbreviations: 2-HG: D-2-hydroxyglutarate; BafA1: bafilomycin A1; ChIP: chromatin immunoprecipitation; FTH1: ferritin heavy chain 1; FTL: ferritin light chain; GBM: glioblastoma; HMOX1/HO-1: heme oxygenase 1; IHC: immunohistochemistry; IDH1: isocitrate dehydrogenase(NADP(+))1; MDC: monodansylcadaverine; NCOA4: nuclear receptor coactivator 4; NFE2L2/Nrf2: NFE2 like bZIP transcription factor 2; PTX3/TSG-14: pentraxin 3; PRMT: protein arginine methyltransferase; SLC40A1: solute carrier family 40 member 1; Tan IIA: tanshinone IIA; TCA: trichloroacetic acid; TEM: transmission electron microscopy; TNF: tumor necrosis factor.
Collapse
Affiliation(s)
- Kirti Lathoria
- Division of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, India
| | - Pruthvi Gowda
- Division of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, India
| | - Sonia B Umdor
- Division of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, India
| | - Shruti Patrick
- Division of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, India
| | - Vaishali Suri
- Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Ellora Sen
- Division of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar, India
| |
Collapse
|
20
|
Bassal MA. The Interplay between Dysregulated Metabolism and Epigenetics in Cancer. Biomolecules 2023; 13:944. [PMID: 37371524 DOI: 10.3390/biom13060944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/21/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular metabolism (or energetics) and epigenetics are tightly coupled cellular processes. It is arguable that of all the described cancer hallmarks, dysregulated cellular energetics and epigenetics are the most tightly coregulated. Cellular metabolic states regulate and drive epigenetic changes while also being capable of influencing, if not driving, epigenetic reprogramming. Conversely, epigenetic changes can drive altered and compensatory metabolic states. Cancer cells meticulously modify and control each of these two linked cellular processes in order to maintain their tumorigenic potential and capacity. This review aims to explore the interplay between these two processes and discuss how each affects the other, driving and enhancing tumorigenic states in certain contexts.
Collapse
Affiliation(s)
- Mahmoud Adel Bassal
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
21
|
Wang QX, Zhang PY, Li QQ, Tong ZJ, Wu JZ, Yu SP, Yu YC, Ding N, Leng XJ, Chang L, Xu JG, Sun SL, Yang Y, Li NG, Shi ZH. Challenges for the development of mutant isocitrate dehydrogenases 1 inhibitors to treat glioma. Eur J Med Chem 2023; 257:115464. [PMID: 37235998 DOI: 10.1016/j.ejmech.2023.115464] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023]
Abstract
Glioma is one of the most common types of brain tumors, and its high recurrence and mortality rates threaten human health. In 2008, the frequent isocitrate dehydrogenase 1 (IDH1) mutations in glioma were reported, which brought a new strategy in the treatment of this challenging disease. In this perspective, we first discuss the possible gliomagenesis after IDH1 mutations (mIDH1). Subsequently, we systematically investigate the reported mIDH1 inhibitors and present a comparative analysis of the ligand-binding pocket in mIDH1. Additionally, we also discuss the binding features and physicochemical properties of different mIDH1 inhibitors to facilitate the future development of mIDH1 inhibitors. Finally, we discuss the possible selectivity features of mIDH1 inhibitors against WT-IDH1 and IDH2 by combining protein-based and ligand-based information. We hope that this perspective can inspire the development of mIDH1 inhibitors and bring potent mIDH1 inhibitors for the treatment of glioma.
Collapse
Affiliation(s)
- Qing-Xin Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Peng-Yu Zhang
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Qing-Qing Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Zhen-Jiang Tong
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Jia-Zhen Wu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Shao-Peng Yu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Yan-Cheng Yu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Ning Ding
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Xue-Jiao Leng
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Liang Chang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Jin-Guo Xu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China
| | - Shan-Liang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China.
| | - Ye Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China.
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu, 210023, China.
| | - Zhi-Hao Shi
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
22
|
Nelson EJ, Gubbiotti MA, Carlin AM, Nasrallah MP, Van Deerlin VM, Herlihy SE. Clinical Evaluation of IDH Mutation Status in Formalin-Fixed Paraffin-Embedded Tissue in Gliomas. Mol Diagn Ther 2023; 27:371-381. [PMID: 36690887 PMCID: PMC9870658 DOI: 10.1007/s40291-022-00638-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND AND OBJECTIVE Determination of isocitrate dehydrogenase (IDH) 1/2 mutational status is crucial for a glioma diagnosis. It is common for IDH mutational status to be determined via a two-step algorithm that utilizes immunohistochemistry studies for IDH1 R132H, the most frequent variant, followed by next-generation sequencing studies for immunohistochemistry-negative or immunohistochemistry-equivocal cases. The objective of this study was to evaluate adding a rapid real-time polymerase chain reaction (RT-PCR) assay to the testing algorithm. METHODS: We validated a modified, commercial, qualitative, RT-PCR assay with the ability to detect 14 variants in IDH1/2 in formalin-fixed paraffin-embedded glioma tumor specimens. The assay was validated using 51 tumor formalin-fixed paraffin-embedded specimens. During clinical implementation of this assay, 48 brain tumor specimens were assessed for IDH result concordance and turnaround time to result. RESULTS Concordance between the RT-PCR and sequencing and IHC studies was 100%. This RT-PCR assay also showed concordant results with IHC for IDH1 R132H for 11 of the 12 (92%) tumor specimens with IDH mutations. The RT-PCR assay yielded faster results (average 2.6 days turnaround time) in comparison to sequencing studies (17.9 days), with complete concordance. CONCLUSIONS In summary, we report that this RT-PCR assay can reliably be performed on formalin-fixed paraffin-embedded specimens and has a faster turnaround time than sequencing assays and can be clinically implemented for determination of IDH mutation status for patients with glioma.
Collapse
Affiliation(s)
- Ernest J Nelson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Maria A Gubbiotti
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Alicia M Carlin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - MacLean P Nasrallah
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Vivianna M Van Deerlin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Sarah E Herlihy
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
23
|
Teraiya M, Perreault H, Chen VC. An overview of glioblastoma multiforme and temozolomide resistance: can LC-MS-based proteomics reveal the fundamental mechanism of temozolomide resistance? Front Oncol 2023; 13:1166207. [PMID: 37182181 PMCID: PMC10169742 DOI: 10.3389/fonc.2023.1166207] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/23/2023] [Indexed: 05/16/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a primary type of lethal brain tumor. Over the last two decades, temozolomide (TMZ) has remained the primary chemotherapy for GBM. However, TMZ resistance in GBM constitutes an underlying factor contributing to high rates of mortality. Despite intense efforts to understand the mechanisms of therapeutic resistance, there is currently a poor understanding of the molecular processes of drug resistance. For TMZ, several mechanisms linked to therapeutic resistance have been proposed. In the past decade, significant progress in the field of mass spectrometry-based proteomics has been made. This review article discusses the molecular drivers of GBM, within the context of TMZ resistance with a particular emphasis on the potential benefits and insights of using global proteomic techniques.
Collapse
Affiliation(s)
- Milan Teraiya
- Chemistry Department, University of Manitoba, Winnipeg, MB, Canada
| | - Helene Perreault
- Chemistry Department, University of Manitoba, Winnipeg, MB, Canada
| | - Vincent C. Chen
- Chemistry Department, Brandon University, Brandon, MB, Canada
| |
Collapse
|
24
|
Burko P, D’Amico G, Miltykh I, Scalia F, Conway de Macario E, Macario AJL, Giglia G, Cappello F, Caruso Bavisotto C. Molecular Pathways Implicated in Radioresistance of Glioblastoma Multiforme: What Is the Role of Extracellular Vesicles? Int J Mol Sci 2023; 24:ijms24054883. [PMID: 36902314 PMCID: PMC10003080 DOI: 10.3390/ijms24054883] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/16/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a primary brain tumor that is very aggressive, resistant to treatment, and characterized by a high degree of anaplasia and proliferation. Routine treatment includes ablative surgery, chemotherapy, and radiotherapy. However, GMB rapidly relapses and develops radioresistance. Here, we briefly review the mechanisms underpinning radioresistance and discuss research to stop it and install anti-tumor defenses. Factors that participate in radioresistance are varied and include stem cells, tumor heterogeneity, tumor microenvironment, hypoxia, metabolic reprogramming, the chaperone system, non-coding RNAs, DNA repair, and extracellular vesicles (EVs). We direct our attention toward EVs because they are emerging as promising candidates as diagnostic and prognostication tools and as the basis for developing nanodevices for delivering anti-cancer agents directly into the tumor mass. EVs are relatively easy to obtain and manipulate to endow them with the desired anti-cancer properties and to administer them using minimally invasive procedures. Thus, isolating EVs from a GBM patient, supplying them with the necessary anti-cancer agent and the capability of recognizing a specified tissue-cell target, and reinjecting them into the original donor appears, at this time, as a reachable objective of personalized medicine.
Collapse
Affiliation(s)
- Pavel Burko
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90133 Palermo, Italy
| | - Giuseppa D’Amico
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90133 Palermo, Italy
| | - Ilia Miltykh
- Department of Human Anatomy, Institute of Medicine, Penza State University, 440026 Penza, Russia
| | - Federica Scalia
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90133 Palermo, Italy
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA
| | - Everly Conway de Macario
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Alberto J. L. Macario
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Giuseppe Giglia
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
- Section of Human Physiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90133 Palermo, Italy
| | - Francesco Cappello
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90133 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Celeste Caruso Bavisotto
- Section of Human Anatomy, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90133 Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
- Correspondence: ; Tel.: +39-0916553501
| |
Collapse
|
25
|
Fortin Ensign SP, Jenkins RB, Giannini C, Sarkaria JN, Galanis E, Kizilbash SH. Translational significance of CDKN2A/B homozygous deletion in isocitrate dehydrogenase-mutant astrocytoma. Neuro Oncol 2023; 25:28-36. [PMID: 35973817 PMCID: PMC9825307 DOI: 10.1093/neuonc/noac205] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Indexed: 01/26/2023] Open
Abstract
Isocitrate dehydrogenase (IDH) 1 or 2 mutations confer a favorable prognosis compared to IDH-wildtype in astrocytoma, frequently denoting a lower grade malignancy. However, recent molecular profiling has identified specific aggressive tumor subgroups with clear clinical prognostic implications that are independent of histologic grading. The homozygous deletion of CDKN2A/B is the strongest implicated independent indicator of the poor prognosis within IDH-mutant astrocytoma, and the identification of this alteration in these lower histologic grade tumors transforms their biology toward an aggressive grade 4 phenotype clinically. CDKN2A/B homozygous deletion is now sufficient to define a grade 4 tumor in IDH-mutant astrocytomas regardless of histologic appearance, yet there are currently no effective molecularly informed targeted therapies for these tumors. The biological impact of CDKN2A/B homozygous deletion in IDH-mutant tumors and the optimal treatment strategy for this molecular subgroup remains insufficiently explored. Here we review the current understanding of the translational significance of homozygous deletion of CDKN2A/B gene expression in IDH-mutant astrocytoma and associated diagnostic and therapeutic implications.
Collapse
Affiliation(s)
| | - Robert B Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Caterina Giannini
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | |
Collapse
|
26
|
Trifănescu OG, Trifănescu RA, Mitrică R, Mitrea D, Ciornei A, Georgescu M, Butnariu I, Galeș LN, Șerbănescu L, Anghel RM, Păun MA. Upstaging and Downstaging in Gliomas-Clinical Implications for the Fifth Edition of the World Health Organization Classification of Tumors of the Central Nervous System. Diagnostics (Basel) 2023; 13:diagnostics13020197. [PMID: 36673007 PMCID: PMC9858599 DOI: 10.3390/diagnostics13020197] [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: 12/03/2022] [Revised: 12/28/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
In 2021, the 5th edition of the WHO Classification of Tumors of the Central Nervous System (WHO-CNS5) was published as the sixth volume of the international standard for brain and spinal cord tumor classification. The most remarkable practical change in the current classification involves grading gliomas according to molecular characterization. IDH mutant (10%) and IDH wild-type tumors (90%) are two different entities that possess unique biological features and various clinical outcomes regarding treatment response and overall survival. This article presents two comparative cases that highlight the clinical importance of these new classification standards. The first clinical case aimed to provide a comprehensive argument for determining the IDH status in tumors initially appearing as low-grade astrocytoma upon histologic examination, thus underlining the importance of the WHO-CNS5. The second case showed the implications of the histologic overdiagnosis of glioblastoma using the previous classification system with a treatment span of 7 years that proceeded through full-dose re-irradiation up to metronomic therapy. The new WHO-CNS5 classification significantly impacted complex neurooncological cases, thus changing the initial approach to a more precise therapeutic management.
Collapse
Affiliation(s)
- Oana Gabriela Trifănescu
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Raluca Alexandra Trifănescu
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- “C. I. Parhon” Bucharest Institute of Endocrinology, 011863 Bucharest, Romania
| | - Radu Mitrică
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
- Correspondence: (R.M.); (D.M.); Tel.: +40-741964311 (R.M.); +40-723226233 (D.M.)
| | - Dan Mitrea
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
- Neuroaxis Neurology Clinic, 011302 Bucharest, Romania
- Correspondence: (R.M.); (D.M.); Tel.: +40-741964311 (R.M.); +40-723226233 (D.M.)
| | - Ana Ciornei
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Mihai Georgescu
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Ioana Butnariu
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, 041914 Bucharest, Romania
| | - Laurenția Nicoleta Galeș
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Medical Oncology II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Luiza Șerbănescu
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Rodica Maricela Anghel
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Mihai-Andrei Păun
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| |
Collapse
|
27
|
Miretti M, Graglia MAG, Suárez AI, Prucca CG. Photodynamic Therapy for glioblastoma: a light at the end of the tunnel. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2023. [DOI: 10.1016/j.jpap.2023.100161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
28
|
Chen J, Wang T, Liu W, Qiu H, Zhang N, Chen X, Ding X, Zhang L. Extended adjuvant temozolomide in newly diagnosed glioblastoma: A single-center retrospective study. Front Oncol 2022; 12:1000501. [PMID: 36483042 PMCID: PMC9723160 DOI: 10.3389/fonc.2022.1000501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/02/2022] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To investigate whether extending adjuvant temozolomide (TMZ) improved the prognosis of newly diagnosed glioblastoma (GBM) patients with different mutation statuses of O6-methylguanine DNA methyltransferase (MGMT), isocitrate dehydrogenase 1 (IDH1), p53 and different expression level of Ki67. METHODS This study was a retrospective cohort study that postoperative patients with newly diagnosed GBM who did not progress after receiving radiotherapy with concomitant and 6 cycles of adjuvant TMZ were enrolled in control group, and those received more than 6 cycles of adjuvant TMZ were incorporated in extended group. Patients were stratified by MGMT expression, IDH1 mutation, p53 mutation and expression level of Ki67. The primary endpoints were overall survival (OS) and progression-free survival (PFS). RESULT A total of 93 postoperative patients with newly diagnosed GBM were included in this study, 40 and 53 cases were included in control group and extended group, respectively. On the whole, extended adjuvant TMZ chemotherapy significantly prolonged OS and PFS of patients with newly diagnosed GBM [median OS (mOS): 29.00 months vs. 16.70 months, P < 0.001; median PFS (mPFS): 13.80 months vs. 9.60 months, P = 0.002]. The results of subgroup analysis showed that patients with methylated MGMT in extended group had significantly longer OS and PFS than those in control group; patients with IDH1 mutation benefited more from extended adjuvant TMZ chemotherapy than those with wild-type IDH1; there was no significant difference in the effect of extended TMZ chemotherapy on OS between GBM patients with wild-type p53 and those with mutant p53; compared with GBM patients with lower expression of Ki67, extended adjuvant TMZ treatment dramatically improved the OS and PFS of those with higher expression of Ki67. CONCLUSION The therapeutic schedule of extended adjuvant TMZ significantly prolonged OS and PFS of patients with newly diagnosed GBM regardless of p53 mutation status, and patients with different MGMT methylation, IDH1 mutation and Ki67 expression level benefited differently from extended adjuvant TMZ chemotherapy.
Collapse
Affiliation(s)
- Jie Chen
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China,Cancer Center, Xuzhou Medical University, Xuzhou, China
| | - Tingting Wang
- First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Wanming Liu
- First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Hui Qiu
- Cancer Center, Xuzhou Medical University, Xuzhou, China
| | - Nie Zhang
- First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Xueting Chen
- First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Xin Ding
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China,*Correspondence: Longzhen Zhang, ; Xin Ding,
| | - Longzhen Zhang
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China,Cancer Center, Xuzhou Medical University, Xuzhou, China,*Correspondence: Longzhen Zhang, ; Xin Ding,
| |
Collapse
|
29
|
Fan X, Nie X, Huang J, Zhang L, Wang X, Lu M. A Composite Bioinformatic Analysis to Explore Endoplasmic Reticulum Stress-Related Prognostic Marker and Potential Pathogenic Mechanisms in Glioma by Integrating Multiomics Data. JOURNAL OF ONCOLOGY 2022; 2022:9886044. [PMID: 36245971 PMCID: PMC9553508 DOI: 10.1155/2022/9886044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/18/2022] [Accepted: 08/14/2022] [Indexed: 11/18/2022]
Abstract
In recent years, abnormal endoplasmic reticulum stress (ERS) response, as an important regulator of immunity, may play a vital role in the occurrence, development, and treatment of glioma. Weighted correlation network analysis (WGCNA) based on six glioma datasets was used to screen eight prognostic-related differentially expressed ERS-related genes (PR-DE-ERSGs) and to construct a prognostic model. BMP2 and HEY2 were identified as protective factors (HR < 1), and NUP107, DRAM1, F2R, PXDN, RNF19A, and SCG5 were identified as risk factors for glioma (HR > 1). QRT-PCR further supported significantly higher DRAM1 and lower SCG5 relative mRNA expression in gliomas. Our model has demonstrated excellent performance in predicting the prognosis of glioma patients from numerous datasets. In addition, the model shows good stability in multiple tests. Our model also shows broad clinical promise in predicting drug treatment effects. More immune cells/processes in the high-risk population with poor prognosis illustrate the importance of the tumor immunosuppressive environment in glioma. The potential role of the HEY2-based competitive endogenous RNA (ceRNA) regulatory network in glioma was validated and revealed the possible important role of glycolysis in glioma ERS. IDH1 and TP53 mutations with better prognosis were strongly associated with the risk score and PR-DE-ERSGs expression in the model. mDNAsi was also closely related to the risk score and clinical characteristics.
Collapse
Affiliation(s)
- Xin Fan
- Department of Emergency, Shangrao Hospital Affiliated to Nanchang University, Shangrao People's Hospital, Shangrao 334000, China
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Xiyi Nie
- Department of Neurosurgery, Yichun Hospital Affiliated to Nanchang University, Yichun People's Hospital, Yichun 334000, China
| | - Junwen Huang
- The First Clinical Medical College of Nanchang University, Nanchang 330000, China
| | - Lingling Zhang
- School of Stomatology, Nanchang University, Nanchang 330000, China
| | - Xifu Wang
- Department of Emergency, Shangrao Hospital Affiliated to Nanchang University, Shangrao People's Hospital, Shangrao 334000, China
| | - Min Lu
- Department of Emergency, Shangrao Hospital Affiliated to Nanchang University, Shangrao People's Hospital, Shangrao 334000, China
| |
Collapse
|
30
|
Chen S, Zhang S, Yuan Y, Wang Z, Li J, Li T, Zuo M, Feng W, Chen M, Liu Y. Prognostic value of cuproptosis-related genes signature and its impact on the reshaped immune microenvironment of glioma. Front Pharmacol 2022; 13:1016520. [PMID: 36267281 PMCID: PMC9576857 DOI: 10.3389/fphar.2022.1016520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/07/2022] [Indexed: 12/04/2022] Open
Abstract
Glioma is the most prevalent malignancy in the central nervous system. The impact of ion-induced cell death on malignant tumors’ development and immune microenvironment has attracted broad attention in recent years. Cuproptosis is a novel copper-dependent mechanism that could potentially regulate tumor cell death by targeting mitochondria respiration. However, the role of cuproptosis in gliomas remains unclear. In the present study, we investigated the relationships between the expression of cuproptosis-related genes (CRGs) and tumor characteristics, including prognosis and microenvironment of glioma, by analyzing multiple public databases and our cohort. Consensus clustering based on the expression of twelve CRGs stratified the glioma patients into three subgroups with significantly different prognosis and immune microenvironment landscapes. Reduced immune infiltration was associated with the less aggressive CRG cluster. A prognostic CRGs risk signature (CRGRS), based on eight critical CRGs, classified the patients into low- and high-risk groups in the training set and was endorsed by validation sets from multiple cohorts. The high-risk group manifested a shorter overall survival, and further survival analysis demonstrated that the CRGRS was an independent prognostic factor. The nomogram combining CRGRS and other clinicopathological factors exhibited good accuracy in predicting the prognosis of glioma patients. Moreover, analyses of tumor immune microenvironment indicated that higher CRGRS was correlated with increased immune cell infiltration but diminished immune function. Gliomas in the high-risk group exhibited higher expression of multiple immune checkpoints, including PD-1 and PD-L1, and a better predicted therapy response to immune checkpoint inhibitors. In conclusion, our study elucidated the connections between CRGs expression and the aggressiveness of gliomas, and the application of CRGRS derived a new robust model for prognosis evaluation of glioma patients. The correlations between the profiles of CRGs expression and immune tumor microenvironment illuminated prospects and potential indications of immunotherapy for glioma.
Collapse
Affiliation(s)
- Siliang Chen
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Shuxin Zhang
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Department of Head and Neck Surgery, Sichuan Cancer Hospital and Institute, Sichuan Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunbo Yuan
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhihao Wang
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junhong Li
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Tengfei Li
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Mingrong Zuo
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Wentao Feng
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Mina Chen
- State Key Laboratory of Biotherapy, Neuroscience & Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Mina Chen, ; Yanhui Liu,
| | - Yanhui Liu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Mina Chen, ; Yanhui Liu,
| |
Collapse
|
31
|
Fan X, Nie X, Huang J, Zhang L, Wang X, Lu M. A Composite Bioinformatic Analysis to Explore Endoplasmic Reticulum Stress-Related Prognostic Marker and Potential Pathogenic Mechanisms in Glioma by Integrating Multiomics Data. JOURNAL OF ONCOLOGY 2022. [DOI: https:/doi.org/10.1155/2022/9886044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
In recent years, abnormal endoplasmic reticulum stress (ERS) response, as an important regulator of immunity, may play a vital role in the occurrence, development, and treatment of glioma. Weighted correlation network analysis (WGCNA) based on six glioma datasets was used to screen eight prognostic-related differentially expressed ERS-related genes (PR-DE-ERSGs) and to construct a prognostic model. BMP2 and HEY2 were identified as protective factors (HR < 1), and NUP107, DRAM1, F2R, PXDN, RNF19A, and SCG5 were identified as risk factors for glioma (HR > 1). QRT-PCR further supported significantly higher DRAM1 and lower SCG5 relative mRNA expression in gliomas. Our model has demonstrated excellent performance in predicting the prognosis of glioma patients from numerous datasets. In addition, the model shows good stability in multiple tests. Our model also shows broad clinical promise in predicting drug treatment effects. More immune cells/processes in the high-risk population with poor prognosis illustrate the importance of the tumor immunosuppressive environment in glioma. The potential role of the HEY2-based competitive endogenous RNA (ceRNA) regulatory network in glioma was validated and revealed the possible important role of glycolysis in glioma ERS. IDH1 and TP53 mutations with better prognosis were strongly associated with the risk score and PR-DE-ERSGs expression in the model. mDNAsi was also closely related to the risk score and clinical characteristics.
Collapse
Affiliation(s)
- Xin Fan
- Department of Emergency, Shangrao Hospital Affiliated to Nanchang University, Shangrao People’s Hospital, Shangrao 334000, China
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Xiyi Nie
- Department of Neurosurgery, Yichun Hospital Affiliated to Nanchang University, Yichun People’s Hospital, Yichun 334000, China
| | - Junwen Huang
- The First Clinical Medical College of Nanchang University, Nanchang 330000, China
| | - Lingling Zhang
- School of Stomatology, Nanchang University, Nanchang 330000, China
| | - Xifu Wang
- Department of Emergency, Shangrao Hospital Affiliated to Nanchang University, Shangrao People’s Hospital, Shangrao 334000, China
| | - Min Lu
- Department of Emergency, Shangrao Hospital Affiliated to Nanchang University, Shangrao People’s Hospital, Shangrao 334000, China
| |
Collapse
|
32
|
Fan X, Nie X, Huang J, Zhang L, Wang X, Lu M. A Composite Bioinformatic Analysis to Explore Endoplasmic Reticulum Stress-Related Prognostic Marker and Potential Pathogenic Mechanisms in Glioma by Integrating Multiomics Data. JOURNAL OF ONCOLOGY 2022. [DOI: doi.org/10.1155/2022/9886044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
In recent years, abnormal endoplasmic reticulum stress (ERS) response, as an important regulator of immunity, may play a vital role in the occurrence, development, and treatment of glioma. Weighted correlation network analysis (WGCNA) based on six glioma datasets was used to screen eight prognostic-related differentially expressed ERS-related genes (PR-DE-ERSGs) and to construct a prognostic model. BMP2 and HEY2 were identified as protective factors (HR < 1), and NUP107, DRAM1, F2R, PXDN, RNF19A, and SCG5 were identified as risk factors for glioma (HR > 1). QRT-PCR further supported significantly higher DRAM1 and lower SCG5 relative mRNA expression in gliomas. Our model has demonstrated excellent performance in predicting the prognosis of glioma patients from numerous datasets. In addition, the model shows good stability in multiple tests. Our model also shows broad clinical promise in predicting drug treatment effects. More immune cells/processes in the high-risk population with poor prognosis illustrate the importance of the tumor immunosuppressive environment in glioma. The potential role of the HEY2-based competitive endogenous RNA (ceRNA) regulatory network in glioma was validated and revealed the possible important role of glycolysis in glioma ERS. IDH1 and TP53 mutations with better prognosis were strongly associated with the risk score and PR-DE-ERSGs expression in the model. mDNAsi was also closely related to the risk score and clinical characteristics.
Collapse
Affiliation(s)
- Xin Fan
- Department of Emergency, Shangrao Hospital Affiliated to Nanchang University, Shangrao People’s Hospital, Shangrao 334000, China
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Xiyi Nie
- Department of Neurosurgery, Yichun Hospital Affiliated to Nanchang University, Yichun People’s Hospital, Yichun 334000, China
| | - Junwen Huang
- The First Clinical Medical College of Nanchang University, Nanchang 330000, China
| | - Lingling Zhang
- School of Stomatology, Nanchang University, Nanchang 330000, China
| | - Xifu Wang
- Department of Emergency, Shangrao Hospital Affiliated to Nanchang University, Shangrao People’s Hospital, Shangrao 334000, China
| | - Min Lu
- Department of Emergency, Shangrao Hospital Affiliated to Nanchang University, Shangrao People’s Hospital, Shangrao 334000, China
| |
Collapse
|
33
|
Karami Fath M, Babakhaniyan K, Anjomrooz M, Jalalifar M, Alizadeh SD, Pourghasem Z, Abbasi Oshagh P, Azargoonjahromi A, Almasi F, Manzoor HZ, Khalesi B, Pourzardosht N, Khalili S, Payandeh Z. Recent Advances in Glioma Cancer Treatment: Conventional and Epigenetic Realms. Vaccines (Basel) 2022; 10:1448. [PMID: 36146527 PMCID: PMC9501259 DOI: 10.3390/vaccines10091448] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/14/2022] [Accepted: 08/27/2022] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma (GBM) is the most typical and aggressive form of primary brain tumor in adults, with a poor prognosis. Successful glioma treatment is hampered by ineffective medication distribution across the blood-brain barrier (BBB) and the emergence of drug resistance. Although a few FDA-approved multimodal treatments are available for glioblastoma, most patients still have poor prognoses. Targeting epigenetic variables, immunotherapy, gene therapy, and different vaccine- and peptide-based treatments are some innovative approaches to improve anti-glioma treatment efficacy. Following the identification of lymphatics in the central nervous system, immunotherapy offers a potential method with the potency to permeate the blood-brain barrier. This review will discuss the rationale, tactics, benefits, and drawbacks of current glioma therapy options in clinical and preclinical investigations.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran 1571914911, Iran
| | - Kimiya Babakhaniyan
- Department of Medical Surgical Nursing, School of Nursing and Midwifery, Iran University of Medical Sciences, Tehran 1996713883, Iran
| | - Mehran Anjomrooz
- Department of Radiology, Shariati Hospital, Tehran University of Medical Sciences, Tehran 1411713135, Iran
| | | | | | - Zeinab Pourghasem
- Department of Microbiology, Islamic Azad University of Lahijan, Gilan 4416939515, Iran
| | - Parisa Abbasi Oshagh
- Department of Biology, Faculty of Basic Sciences, Malayer University, Malayer 6571995863, Iran
| | - Ali Azargoonjahromi
- Department of Nursing, School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz 7417773539, Iran
| | - Faezeh Almasi
- Pharmaceutical Biotechnology Lab, Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran 1411734115, Iran
| | - Hafza Zahira Manzoor
- Experimental and Translational Medicine, University of Insubria, Via jean Henry Dunant 3, 21100 Varese, Italy
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj 3197619751, Iran
| | - Navid Pourzardosht
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht 4193713111, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran 1678815811, Iran
| | - Zahra Payandeh
- Department of Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, SE-17177 Stockholm, Sweden
| |
Collapse
|
34
|
Wang J, Sun X, Wang J, Zhang K, Yuan Y, Guo Y, Yao L, Li X, Shen L. NDRG2 inhibits pyruvate carboxylase-mediated anaplerosis and combines with glutamine blockade to inhibit the proliferation of glioma cells. Am J Cancer Res 2022; 12:3729-3744. [PMID: 36119843 PMCID: PMC9442009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023] Open
Abstract
Due to the rapid proliferation, cancer cells have increased anabolic biosynthesis, which requires anaplerosis to replenish precursor intermediates. The major anaplerotic sources are pyruvate and glutamine, which require the catalysis of pyruvate carboxylase (PC) and glutaminase (GLS) respectively. In GLS-suppressed cancer cells, the PC-mediated pathway for anaplerosis is crucial to maintain cell growth and proliferation. Here, we investigated the regulatory role and molecular mechanism of N-myc downstream-regulated gene 2 (NDRG2) in PC and PC-mediated anaplerosis. NDRG2 interacted with PC and induced the degradation of PC in glutamine-deprived cells. NDRG2 also inhibited the activity of PC and PC-mediated anaplerosis. As a result, NDRG2 significantly inhibited the malignant growth and proliferation of glioma cells in combination with a glutamine antagonist. In addition, NDRG2 more significantly inhibited the protein level of PC in isocitrate dehydrogenase 1 (R132H)-mutant glioma cells than in wild-type glioma cells. These findings indicate that the molecular mechanism of NDRG2 inhibits PC-mediated anaplerosis and collaborates with glutamine antagonist to inhibit the malignant proliferation of glioma cells, thus providing a theoretical and experimental basis for targeting anaplerosis in glioma therapy.
Collapse
Affiliation(s)
- Jiancai Wang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
- Department of Neurosurgery, PLA 982 HospitalTangshan 063099, Hebei, China
| | - Xiang Sun
- Department of Special Diagnosis, School of Stomatology, The Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
| | - Jiayuan Wang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
- Department of Pathogenic Biology, Medical College, Yan’an UniversityYan’an 716000, Shaanxi, China
| | - Kun Zhang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
- Department of Medical Genetics, Medical College, Yan’an UniversityYan’an 716000, Shaanxi, China
| | - Yiyi Yuan
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
| | - Yan Guo
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
| | - Libo Yao
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
| | - Xia Li
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
| | - Lan Shen
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical UniversityXi’an 710032, Shaanxi, China
| |
Collapse
|
35
|
Gong Y, Wei S, Wei Y, Chen Y, Cui J, Yu Y, Lin X, Yan H, Qin H, Yi L. IDH2: A novel biomarker for environmental exposure in blood circulatory system disorders (Review). Oncol Lett 2022; 24:278. [PMID: 35814829 PMCID: PMC9260733 DOI: 10.3892/ol.2022.13398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/24/2022] [Indexed: 11/11/2022] Open
Abstract
As the risk of harmful environmental exposure is increasing, it is important to find suitable targets for the diagnosis and treatment of the diseases caused. Isocitrate dehydrogenase 2 (IDH2) is an enzyme located in the mitochondria; it plays an important role in numerous cell processes, including maintaining redox homeostasis, participating in the tricarboxylic acid cycle and indirectly taking part in the transmission of the oxidative respiratory chain. IDH2 mutations promote progression in acute myeloid leukemia, glioma and other diseases. The present review mainly summarizes the role and mechanism of IDH2 with regard to the biological effects, such as the mitophagy and apoptosis of animal or human cells, caused by environmental pollution such as radiation, heavy metals and other environmental exposure factors. The possible mechanisms of these biological effects are described in terms of IDH2 expression, reduced nicotine adenine dinucleotide phosphate content and reactive oxygen species level, among other variables. The impact of environmental pollution on human health is increasingly attracting attention. IDH2 may therefore become useful as a potential diagnostic and therapeutic target for environmental exposure-induced diseases.
Collapse
Affiliation(s)
- Ya Gong
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shuang Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yuan Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yong Chen
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jian Cui
- Institute of Cardiovascular Disease, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yue Yu
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiang Lin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hong Yan
- Pediatric Intensive Care Unit, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hui Qin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Lan Yi
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
36
|
Wei Z, Ye S, Feng H, Zeng C, Dong X, Zeng X, Zeng L, Lin X, Liu Q, Yao J. Silybin suppresses ovarian cancer cell proliferation by inhibiting isocitrate dehydrogenase 1 activity. Cancer Sci 2022; 113:3032-3043. [PMID: 35730256 PMCID: PMC9459272 DOI: 10.1111/cas.15470] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/01/2022] [Accepted: 06/14/2022] [Indexed: 11/27/2022] Open
Abstract
Metabolic reprogramming is a sign of malignant tumors, and targeting the metabolism of tumor cells has become a promising therapeutic approach. Here, we report that Silybin (a nontoxic flavonoid commonly used for liver protection) exhibits prominent anti‐tumor effects on human ovarian cancer cells. Treatment of an ovarian cancer cell line with Silybin interfered with glutamine metabolism and the tricarboxylic acid cycle. We applied the drug affinity responsive target stability approach to show that Silybin binds to isocitrate dehydrogenase 1 (IDH1). This combination leads to reduced phosphorylation of IDH1 and inhibits enzyme activity. IDH1 dysfunction significantly increases the ratio of NADP/NADPH in the cell, causing an increase in reactive oxygen species generation. Immunohistochemistry demonstrated that IDH1 was increased in ovarian cancer samples compared with normal para‐tumoral tissues. Xenograft murine experiments indicated that Silybin administered orally suppressed the growth of the tumor formed by ovarian cancer cells. In combination, our data strongly suggest that Silybin targets IDH1 in ovarian cancer cells and may be a novel treatment candidate.
Collapse
Affiliation(s)
- Zibo Wei
- Department of Laboratory Medicine and Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Shuangyan Ye
- Medical research center, The eighth affiliated hospital, Sun Yat-Sen University, Shenzhen, China
| | - Haipeng Feng
- Department of Pathology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Chong Zeng
- Department of Laboratory Medicine and Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Xinhuai Dong
- Department of Laboratory Medicine and Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Xiaokang Zeng
- Department of Laboratory Medicine and Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Liming Zeng
- Department of Laboratory Medicine and Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Xu Lin
- Department of Laboratory Medicine and Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Qiuzhen Liu
- Department of Laboratory Medicine and Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Jie Yao
- Department of Laboratory Medicine and Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, Guangdong, China
| |
Collapse
|
37
|
Raimondi V, Ciotti G, Gottardi M, Ciccarese F. 2-Hydroxyglutarate in Acute Myeloid Leukemia: A Journey from Pathogenesis to Therapies. Biomedicines 2022; 10:biomedicines10061359. [PMID: 35740380 PMCID: PMC9220225 DOI: 10.3390/biomedicines10061359] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 12/19/2022] Open
Abstract
The oncometabolite 2-hydroxyglutarate (2-HG) plays a key role in differentiation blockade and metabolic reprogramming of cancer cells. Approximatively 20–30% of acute myeloid leukemia (AML) cases carry mutations in the isocitrate dehydrogenase (IDH) enzymes, leading to a reduction in the Krebs cycle intermediate α-ketoglutarate (α-KG) to 2-HG. Relapse and chemoresistance of AML blasts following initial good response to standard therapy account for the very poor outcome of this pathology, which represents a great challenge for hematologists. The decrease of 2-HG levels through pharmacological inhibition of mutated IDH enzymes induces the differentiation of AML blasts and sensitizes leukemic cells to several anticancer drugs. In this review, we provide an overview of the main genetic mutations in AML, with a focus on IDH mutants and the role of 2-HG in AML pathogenesis. Moreover, we discuss the impact of high levels of 2-HG on the response of AML cells to antileukemic therapies and recent evidence for highly efficient combinations of mutant IDH inhibitors with other drugs for the management of relapsed/refractory (R/R) AML.
Collapse
Affiliation(s)
- Vittoria Raimondi
- Department of Surgery, Oncology and Gastroenterology, University of Padua, 35128 Padova, Italy
- Correspondence:
| | - Giulia Ciotti
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV–IRCCS, 31033 Castelfranco Veneto, Italy; (G.C.); (M.G.)
| | - Michele Gottardi
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV–IRCCS, 31033 Castelfranco Veneto, Italy; (G.C.); (M.G.)
| | - Francesco Ciccarese
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV–IRCCS, 35128 Padova, Italy;
| |
Collapse
|
38
|
Yan Y, Dai W, Mei Q. Multicentric Glioma: An Ideal Model to Reveal the Mechanism of Glioma. Front Oncol 2022; 12:798018. [PMID: 35747806 PMCID: PMC9209746 DOI: 10.3389/fonc.2022.798018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
As a special type of glioma, multicentric glioma provides an ideal pathological model for glioma research. According to the stem-cell-origin theory, multiple lesions of multicentric glioma share the same neuro-oncological origin, both in gene level and in cell level. Although the number of studies focusing on genetic evolution in gliomas with the model of multicentric gliomas were limited, some mutations, including IDH1 mutations, TERTp mutations and PTEN deletions, are found to be at an early stage in the process of genetic aberrance during glioma evolution based on the results of these studies. This article reviews the clinical reports and genetic studies of multicentric glioma, and intends to explain the various clinical phenomena of multicentric glioma from the perspective of genetic aberrance accumulation and tumor cell evolution. The malignant degree of a glioma is determined by both the tumorigenicity of early mutant genes, and the stemness of early suffered cells.
Collapse
Affiliation(s)
- Yong Yan
- Departmentof Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Wei Dai
- Departmentof Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Qiyong Mei
- Departmentof Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
39
|
Different Effects of RNAi-Mediated Downregulation or Chemical Inhibition of NAMPT in an Isogenic IDH Mutant and Wild-Type Glioma Cell Model. Int J Mol Sci 2022; 23:ijms23105787. [PMID: 35628596 PMCID: PMC9143996 DOI: 10.3390/ijms23105787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 12/13/2022] Open
Abstract
The IDH1R132H mutation in glioma results in the neoenzymatic function of IDH1, leading to the production of the oncometabolite 2-hydroxyglutarate (2-HG), alterations in energy metabolism and changes in the cellular redox household. Although shifts in the redox ratio NADPH/NADP+ were described, the consequences for the NAD+ synthesis pathways and potential therapeutic interventions were largely unexplored. Here, we describe the effects of heterozygous IDH1R132H on the redox system in a CRISPR/Cas edited glioblastoma model and compare them with IDH1 wild-type (IDH1wt) cells. Besides an increase in 2-HG and decrease in NADPH, we observed an increase in NAD+ in IDH1R132H glioblastoma cells. RT-qPCR analysis revealed the upregulation of the expression of the NAD+ synthesis enzyme nicotinamide phosphoribosyltransferase (NAMPT). Knockdown of NAMPT resulted in significantly reduced viability in IDH1R132H glioblastoma cells. Given this dependence of IDH1R132H cells on NAMPT expression, we explored the effects of the NAMPT inhibitors FK866, GMX1778 and GNE-617. Surprisingly, these agents were equally cytotoxic to IDH1R132H and IDH1wt cells. Altogether, our results indicate that targeting the NAD+ synthesis pathway is a promising therapeutic strategy in IDH mutant gliomas; however, the agent should be carefully considered since three small-molecule inhibitors of NAMPT tested in this study were not suitable for this purpose.
Collapse
|
40
|
Vilar JB, Christmann M, Tomicic MT. Alterations in Molecular Profiles Affecting Glioblastoma Resistance to Radiochemotherapy: Where Does the Good Go? Cancers (Basel) 2022; 14:cancers14102416. [PMID: 35626024 PMCID: PMC9139489 DOI: 10.3390/cancers14102416] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Glioblastoma is a type of brain cancer that remains incurable. Despite multiple past and ongoing preclinical studies and clinical trials, involving adjuvants to the conventional therapy and based on molecular targeting, no relevant benefit for patients’ survival has been achieved so far. The current first-line treatment regimen is based on ionizing radiation and the monoalkylating compound, temozolomide, and has been administered for more than 15 years. Glioblastoma is extremely resistant to most agents due to a mutational background that elicits quick response to insults and adapts to microenvironmental and metabolic changes. Here, we present the most recent evidence concerning the molecular features and their alterations governing pathways involved in GBM response to the standard radio-chemotherapy and discuss how they collaborate with acquired GBM’s resistance. Abstract Glioblastoma multiforme (GBM) is a brain tumor characterized by high heterogeneity, diffuse infiltration, aggressiveness, and formation of recurrences. Patients with this kind of tumor suffer from cognitive, emotional, and behavioral problems, beyond exhibiting dismal survival rates. Current treatment comprises surgery, radiotherapy, and chemotherapy with the methylating agent, temozolomide (TMZ). GBMs harbor intrinsic mutations involving major pathways that elicit the cells to evade cell death, adapt to the genotoxic stress, and regrow. Ionizing radiation and TMZ induce, for the most part, DNA damage repair, autophagy, stemness, and senescence, whereas only a small fraction of GBM cells undergoes treatment-induced apoptosis. Particularly upon TMZ exposure, most of the GBM cells undergo cellular senescence. Increased DNA repair attenuates the agent-induced cytotoxicity; autophagy functions as a pro-survival mechanism, protecting the cells from damage and facilitating the cells to have energy to grow. Stemness grants the cells capacity to repopulate the tumor, and senescence triggers an inflammatory microenvironment favorable to transformation. Here, we highlight this mutational background and its interference with the response to the standard radiochemotherapy. We discuss the most relevant and recent evidence obtained from the studies revealing the molecular mechanisms that lead these cells to be resistant and indicate some future perspectives on combating this incurable tumor.
Collapse
|
41
|
Fan S, Wu N, Chang S, Chen L, Sun X. The immune regulation of BCL3 in glioblastoma with mutated IDH1. Aging (Albany NY) 2022; 14:3856-3873. [PMID: 35488886 PMCID: PMC9134951 DOI: 10.18632/aging.204048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/25/2022] [Indexed: 11/25/2022]
Abstract
Background: Glioblastoma in the brain is the most malignant solid tumor with a poor prognosis. Screening critical targets and exploring underlying mechanisms will be a benefit for diagnoses and treatment. IDH1 mutation (R132) was used to distinguish glioblastoma grade and predict prognosis as a significant marker. However, the manner of IDH1 mutation regulating glioblastoma development was still unclear. Methods: To study the function of IDH1 mutation, multi-type sequencing data (transcriptome, methylation and copy number variation) from the GEO and TCGA database were analyzed using bioinformatics techniques. The biological functions of IDH1 mutation (R132) would be comprehensively evaluated from the regulatory networks, tumor immune microenvironment and clinical relevance. Then the analysis result would be validated by experimental techniques. Results: Compared with adjacent tissues, IDH1 was up-regulated in glioblastoma, which also positively correlated with the malignant degree and a poor prognosis. To further study the mechanism of mutated IDH1 (R132) function, 5 correlated genes (FABP5, C1RL, MIR155HG, CSTA and BCL3) were identified by different expression gene screening, enrichment analysis and network construction successively. Among them, the BCL3 was a transcription factor that may induce IDH1expression. Through calculating the correlation coefficient, it was found that in IDH1mut glioblastoma, the dendritic cell infiltration was reduced which may result in a better prognosis. In addition, the level of IDH1, FABP5, C1RL, MIR155HG, CSTA and BCL3 might also influence lymphocytes infiltration (eg. CD4+ T cell) and chemokine expression (CXCL family). Conclusions: IDH1 may participate in pathological mechanisms of glioblastoma via expression alteration or gene mutation. Furthermore, IDH1 mutation might improve prognosis via suppressing the expression of FABP5, C1RL, MIR155HG, CSTA and BCL3. Meanwhile, it was identified that BCL3 might perform similar immunomodulatory functions with IDH1 as an upstream transcript factor.
Collapse
Affiliation(s)
- Shibing Fan
- Department of Neurosurgery, Chongqing Medical University, Chongqing, China.,Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| | - Na Wu
- Department of Neurosurgery, Chongqing Medical University, Chongqing, China.,Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| | - Shichuan Chang
- Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, China
| | - Long Chen
- Chongqing University, Shapingba, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, Chongqing Medical University, Chongqing, China
| |
Collapse
|
42
|
Wu S, Zhang X, Rui W, Sheng Y, Yu Y, Zhang Y, Yao Z, Qiu T, Ren Y. A nomogram strategy for identifying the subclassification of IDH mutation and ATRX expression loss in lower-grade gliomas. Eur Radiol 2022; 32:3187-3198. [PMID: 35133485 DOI: 10.1007/s00330-021-08444-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 09/22/2021] [Accepted: 10/25/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVES To construct a radiomics nomogram based on multiparametric MRI data for predicting isocitrate dehydrogenase 1 mutation (IDH +) and loss of nuclear alpha thalassemia/mental retardation syndrome X-linked expression (ATRX -) in patients with lower-grade gliomas (LrGG; World Health Organization [WHO] 2016 grades II and III). METHODS A total of 111 LrGG patients (76 mutated IDH and 35 wild-type IDH) were enrolled, divided into a training set (n = 78) and a validation set (n = 33) for predicting IDH mutation. IDH + LrGG patients were further stratified into the ATRX - (n = 38) and ATRX + (n = 38) subtypes. A total of 250 radiomics features were extracted from the region of interest of each tumor, including that from T2 fluid-attenuated inversion recovery (T2 FLAIR), contrast-enhanced T1 WI, ASL-derived cerebral blood flow (CBF), DWI-derived ADC, and exponential ADC (eADC). A radiomics signature was selected using the Elastic Net regression model, and a radiomics nomogram was finally constructed using the age, gender information, and above features. RESULTS The radiomics nomogram identified LrGG patients for IDH mutation (C-index: training sets = 0.881, validation sets = 0.900) and ATRX loss (C-index: training sets = 0.863, validation sets = 0.840) with good calibration. Decision curve analysis further confirmed the clinical usefulness of the two nomograms for predicting IDH and ATRX status. CONCLUSIONS The nomogram incorporating age, gender, and the radiomics signature provided a clinically useful approach in noninvasively predicting IDH and ATRX mutation status for LrGG patients. The proposed method could facilitate MRI-based clinical decision-making for the LrGG patients. KEY POINTS • Non-invasive determination of IDH and ATRX gene status of LrGG patients can be obtained with a radiomics nomogram. • The proposed nomogram is constructed by radiomics signature selected from 250 radiomics features, combined with age and gender. • The proposed radiomics nomogram exhibited good calibration and discrimination for IDH and ATRX gene mutation stratification of LrGG patients in both training and validation sets.
Collapse
Affiliation(s)
- Shiman Wu
- Department of Radiology, Huashan Hospital, Fudan University, Jing'an District, 12 Middle Urumqi Road, Shanghai, 200040, People's Republic of China
| | - Xi Zhang
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Wenting Rui
- Department of Radiology, Huashan Hospital, Fudan University, Jing'an District, 12 Middle Urumqi Road, Shanghai, 200040, People's Republic of China
| | - Yaru Sheng
- Department of Radiology, Huashan Hospital, Fudan University, Jing'an District, 12 Middle Urumqi Road, Shanghai, 200040, People's Republic of China
| | - Yang Yu
- Department of Radiology, Huashan Hospital, Fudan University, Jing'an District, 12 Middle Urumqi Road, Shanghai, 200040, People's Republic of China
| | - Yong Zhang
- GE Healthcare, Shanghai, People's Republic of China
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Jing'an District, 12 Middle Urumqi Road, Shanghai, 200040, People's Republic of China
| | - Tianming Qiu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Jing'an District, 12 Middle Urumqi Road, Shanghai, 200040, People's Republic of China.
| | - Yan Ren
- Department of Radiology, Huashan Hospital, Fudan University, Jing'an District, 12 Middle Urumqi Road, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
43
|
Yang Z, Hu N, Wang W, Hu W, Zhou S, Shi J, Li M, Jing Z, Chen C, Zhang X, Yang R, Fu X, Wang X. Loss of FBXW7 Correlates with Increased IDH1 Expression in Glioma and Enhances IDH1-Mutant Cancer Cell Sensitivity to Radiation. Cancer Res 2022; 82:497-509. [PMID: 34737211 DOI: 10.1158/0008-5472.can-21-0384] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 09/20/2021] [Accepted: 10/29/2021] [Indexed: 11/16/2022]
Abstract
F-box and WD repeat domain containing 7 (FBXW7) is a substrate receptor of the ubiquitin ligase SKP1-Cullin1-F-box complex and a potent tumor suppressor that prevents unregulated cell growth and tumorigenesis. However, little is known about FBXW7-mediated control of cell metabolism and related functions in cancer therapy. Here, we report that FBXW7 expression inversely correlates with the expression levels of the key metabolic enzyme isocitrate dehydrogenase 1 (IDH1) in patients with glioma and public glioma datasets. Deletion of FBXW7 significantly increased both wild-type (WT) and mutant IDH1 expression, which was mediated by blocking degradation of sterol regulatory element binding protein 1 (SREBP1). The upregulation of neomorphic mutant IDH1 by FBXW7 deletion stimulated production of the oncometabolite 2-hydroxyglutarate at the expense of increasing pentose phosphate pathway activity and NADPH consumption, limiting the buffering ability against radiation-induced oxidative stress. In addition, FBXW7 knockout and IDH1 mutations induced nonhomologous end joining and homologous recombination defects, respectively. In vitro and in vivo, loss of FBXW7 dramatically enhanced the efficacy of radiation treatment in IDH1-mutant cancer cells. Taken together, this work identifies FBXW7 deficiency as a potential biomarker representing both DNA repair and metabolic vulnerabilities that sensitizes IDH1-mutant cancers to radiotherapy. SIGNIFICANCE: Deficiency of FBXW7 causes defects in DNA repair and disrupts NADPH homeostasis in IDH1-mutant glioma cells, conferring high sensitivity to radiotherapy.
Collapse
Affiliation(s)
- Zhuo Yang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, P.R. China
| | - Nan Hu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, P.R. China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Weihua Hu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, P.R. China
| | - Shaolong Zhou
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, P.R. China
| | - Jianxiang Shi
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, P.R. China
| | - Minghe Li
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, P.R. China
| | - Zhou Jing
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, P.R. China
| | - Chao Chen
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, P.R. China
| | - Xuyang Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Ruyi Yang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, P.R. China
| | - Xudong Fu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China.
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, P.R. China
| | - Xinjun Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China.
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, Henan, P.R. China
| |
Collapse
|
44
|
Mazurek M, Szczepanek D, Orzyłowska A, Rola R. Analysis of Factors Affecting 5-ALA Fluorescence Intensity in Visualizing Glial Tumor Cells-Literature Review. Int J Mol Sci 2022; 23:ijms23020926. [PMID: 35055109 PMCID: PMC8779265 DOI: 10.3390/ijms23020926] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 01/27/2023] Open
Abstract
Glial tumors are one of the most common lesions of the central nervous system. Despite the implementation of appropriate treatment, the prognosis is not successful. As shown in the literature, maximal tumor resection is a key element in improving therapeutic outcome. One of the methods to achieve it is the use of fluorescent intraoperative navigation with 5-aminolevulinic acid. Unfortunately, often the level of fluorescence emitted is not satisfactory, resulting in difficulties in the course of surgery. This article summarizes currently available knowledge regarding differences in the level of emitted fluorescence. It may depend on both the histological type and the genetic profile of the tumor, which is reflected in the activity and expression of enzymes involved in the intracellular metabolism of fluorescent dyes, such as PBGD, FECH, UROS, and ALAS. The transport of 5-aminolevulinic acid and its metabolites across the blood–brain barrier and cell membranes mediated by transporters, such as ABCB6 and ABCG2, is also important. Accompanying therapies, such as antiepileptic drugs or steroids, also have an impact on light emission by tumor cells. Accurate determination of the factors influencing the fluorescence of 5-aminolevulinic acid-treated cells may contribute to the improvement of fluorescence navigation in patients with highly malignant gliomas.
Collapse
|
45
|
Sumbly V, Landry I, Rizzo V. Ivosidenib for IDH1 Mutant Cholangiocarcinoma: A Narrative Review. Cureus 2022; 14:e21018. [PMID: 35154988 PMCID: PMC8818329 DOI: 10.7759/cureus.21018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2022] [Indexed: 11/16/2022] Open
Abstract
Cholangiocarcinoma is an uncommon gastrointestinal neoplasm characterized by the abnormal proliferation of cholangiocytes within the biliary duct. This type of malignancy can be subdivided into three major classes: intrahepatic cholangiocarcinoma (iCCA), perihilar cholangiocarcinoma (pCCA), and distal cholangiocarcinoma (dCCA). Based on the results of various clinical trials, ivosidenib was approved for acute myeloid leukemia harboring the IDH1 mutation. It has also been shown that ivosidenib was effective in patients with IDH1 mutated cholangiocarcinoma. In this article, we briefly review the genomics and prognosis of cholangiocarcinoma with a special focus on ivosidenib and the mechanisms by which its approval was met.
Collapse
|
46
|
Alzial G, Renoult O, Paris F, Gratas C, Clavreul A, Pecqueur C. Wild-type isocitrate dehydrogenase under the spotlight in glioblastoma. Oncogene 2022; 41:613-621. [PMID: 34764443 PMCID: PMC8799461 DOI: 10.1038/s41388-021-02056-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/21/2021] [Accepted: 09/30/2021] [Indexed: 01/03/2023]
Abstract
Brain tumors actively reprogram their cellular metabolism to survive and proliferate, thus offering potential therapeutic opportunities. Over the past decade, extensive research has been done on mutant IDH enzymes as markers of good prognosis in glioblastoma, a highly aggressive brain tumor in adults with dismal prognosis. Yet, 95% of glioblastoma are IDH wild-type. Here, we review current knowledge about IDH wild-type enzymes and their putative role in mechanisms driving tumor progression. After a brief overview on tumor metabolic adaptation, we present the diverse metabolic function of IDH enzymes and their roles in glioblastoma initiation, progression and response to treatments. Finally, we will discuss wild-type IDH targeting in primary glioblastoma.
Collapse
Affiliation(s)
- Gabriel Alzial
- Université de Nantes, CRCINA, INSERM, CNRS, F-44000, Nantes, France
| | - Ophelie Renoult
- Université de Nantes, CRCINA, INSERM, CNRS, F-44000, Nantes, France
| | - François Paris
- Université de Nantes, CRCINA, INSERM, CNRS, F-44000, Nantes, France
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Catherine Gratas
- Université de Nantes, CHU Nantes, Inserm, CRCINA, F-44000, Nantes, France
| | - Anne Clavreul
- Université d'Angers, CHU d'Angers, CRCINA, F-49000, Angers, France
- Département de Neurochirurgie, CHU Angers, Angers, France
| | - Claire Pecqueur
- Université de Nantes, CRCINA, INSERM, CNRS, F-44000, Nantes, France.
| |
Collapse
|
47
|
Molenaar RJ, Wilmink JW. IDH1/2 Mutations in Cancer Stem Cells and Their Implications for Differentiation Therapy. J Histochem Cytochem 2021; 70:83-97. [PMID: 34967233 PMCID: PMC8721574 DOI: 10.1369/00221554211062499] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Isocitrate dehydrogenase 1 and 2 (IDH1/2) are enzymes recurrently mutated in various types of cancer, including glioma, cholangiocarcinoma, chondrosarcoma, and acute myeloid leukemia. Mutant IDH1/2 induce a block in differentiation and thereby contribute to the stemness and oncogenesis of their cells of origin. Recently, small-molecule inhibitors of mutant IDH1/2 have been Food and Drug Administration-approved for the treatment of IDH1/2-mutated acute myeloid leukemia. These inhibitors decrease the stemness of the targeted IDH1/2-mutated cancer cells and induce their differentiation to more mature cells. In this review, we elucidate the mechanisms by which mutant IDH1/2 induce a block in differentiation and the biological and clinical effects of the release into differentiation by mutant-IDH1/2 inhibitors. (J Histochem Cytochem 70:83-97, 2022).
Collapse
Affiliation(s)
- Remco J Molenaar
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands.,Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Johanna W Wilmink
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
48
|
Hvinden IC, Cadoux-Hudson T, Schofield CJ, McCullagh JS. Metabolic adaptations in cancers expressing isocitrate dehydrogenase mutations. Cell Rep Med 2021; 2:100469. [PMID: 35028610 PMCID: PMC8714851 DOI: 10.1016/j.xcrm.2021.100469] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The most frequently mutated metabolic genes in human cancer are those encoding the enzymes isocitrate dehydrogenase 1 (IDH1) and IDH2; these mutations have so far been identified in more than 20 tumor types. Since IDH mutations were first reported in glioma over a decade ago, extensive research has revealed their association with altered cellular processes. Mutations in IDH lead to a change in enzyme function, enabling efficient conversion of 2-oxoglutarate to R-2-hydroxyglutarate (R-2-HG). It is proposed that elevated cellular R-2-HG inhibits enzymes that regulate transcription and metabolism, subsequently affecting nuclear, cytoplasmic, and mitochondrial biochemistry. The significance of these biochemical changes for tumorigenesis and potential for therapeutic exploitation remains unclear. Here we comprehensively review reported direct and indirect metabolic changes linked to IDH mutations and discuss their clinical significance. We also review the metabolic effects of first-generation mutant IDH inhibitors and highlight the potential for combination treatment strategies and new metabolic targets.
Collapse
Affiliation(s)
- Ingvild Comfort Hvinden
- Chemistry Research Laboratory, 12 Mansfield Road, Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| | - Tom Cadoux-Hudson
- Chemistry Research Laboratory, 12 Mansfield Road, Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| | - Christopher J. Schofield
- Chemistry Research Laboratory, 12 Mansfield Road, Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
- Ineos Oxford Institute for Antimicrobial Research, 12 Mansfield Road, Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| | - James S.O. McCullagh
- Chemistry Research Laboratory, 12 Mansfield Road, Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| |
Collapse
|
49
|
Nelakurti DD, Rossetti T, Husbands AY, Petreaca RC. Arginine Depletion in Human Cancers. Cancers (Basel) 2021; 13:6274. [PMID: 34944895 PMCID: PMC8699593 DOI: 10.3390/cancers13246274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/04/2021] [Accepted: 12/09/2021] [Indexed: 11/25/2022] Open
Abstract
Arginine is encoded by six different codons. Base pair changes in any of these codons can have a broad spectrum of effects including substitutions to twelve different amino acids, eighteen synonymous changes, and two stop codons. Four amino acids (histidine, cysteine, glutamine, and tryptophan) account for over 75% of amino acid substitutions of arginine. This suggests that a mutational bias, or "purifying selection", mechanism is at work. This bias appears to be driven by C > T and G > A transitions in four of the six arginine codons, a signature that is universal and independent of cancer tissue of origin or histology. Here, we provide a review of the available literature and reanalyze publicly available data from the Catalogue of Somatic Mutations in Cancer (COSMIC). Our analysis identifies several genes with an arginine substitution bias. These include known factors such as IDH1, as well as previously unreported genes, including four cancer driver genes (FGFR3, PPP6C, MAX, GNAQ). We propose that base pair substitution bias and amino acid physiology both play a role in purifying selection. This model may explain the documented arginine substitution bias in cancers.
Collapse
Affiliation(s)
- Devi D. Nelakurti
- Biomedical Science Undergraduate Program, The Ohio State University Medical School, Columbus, OH 43210, USA;
| | - Tiffany Rossetti
- Biology Undergraduate Program, The Ohio State University, Marion, OH 43302, USA;
| | - Aman Y. Husbands
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43215, USA
| | - Ruben C. Petreaca
- Department of Molecular Genetics, The Ohio State University, Marion, OH 43302, USA
- Cancer Biology Program, The Ohio State University James Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
50
|
Uson Junior PLS, Borad MJ. Precision approaches for cholangiocarcinoma: Progress in clinical trials and beyond. Expert Opin Investig Drugs 2021; 31:125-131. [PMID: 34904492 DOI: 10.1080/13543784.2022.2017882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Although a relatively uncommon tumor, the incidence of intrahepatic cholangiocarcinoma is rising globally. Unfortunately, most patients are diagnosed with locally advanced or metastatic disease with poor prognosis. Strategies targeting genomic alterations and incorporation of precision medicine will represent a new therapeutic avenue for these patients. AREAS COVERED In this review article we addressed clinical trials in cholangiocarcinoma with FGFR, IDH, BRAF and ErbB2 targeted therapies. We also reviewed mechanisms of resistance to precision medicine and possible future strategies to overcome clonal evolution. Articles selected for this review were based on reported studies indexed in PubMed (2010-2021). EXPERT OPINION Pemigatinib, infigratinib and futibatinib could eventually be incorporated in the landscape of first-line systemic treatment for advanced cholangiocarcinoma with FGFR2 fusions or rearrangements after the ongoing phase III trials. Circulating tumor DNA could be used as a dynamic tool for evaluating mechanisms of resistance and prediction of response in patients treated with directed therapy.
Collapse
Affiliation(s)
- Pedro Luiz Serrano Uson Junior
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA.,Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Mitesh J Borad
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Medicine, Rochester, MN, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|