1
|
Li Y, Yu J, Zhang Y, Peng C, Song Y, Liu S. Advances in targeted therapy of cholangiocarcinoma. Ann Med 2024; 56:2310196. [PMID: 38359439 PMCID: PMC10877652 DOI: 10.1080/07853890.2024.2310196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 01/20/2024] [Indexed: 02/17/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a malignant tumor originating in the bile duct and its branching epithelium. Due to its high heterogeneity, there are no specific clinical indications at the early stage, the diagnosis is often in advanced CCA. With surgical resection, the 5-year postoperative survival rate (long-term survival rate) is very poor. The regimen of gemcitabine combined with platinum has been used as the first-line chemotherapy for advanced patients. In recent years, targeted therapy for a variety of malignant tumors has made great progress, showing good efficacy and safety in advanced CCA. However, the current targeted therapy of CCA still has many challenges, such as adverse reactions, drug resistance, and individual differences. Therefore, the researches need to further explore the targeted therapy mechanism of CCA malignancies in depth, develop more effective and safe drugs, and accurately formulate plans based on patient characteristics to further improve patient prognosis in the future. This article reviews the recent progress of targeted therapy for CCA, aiming to provide a strategy for the research and clinical work of targeted therapy for CCA.
Collapse
Affiliation(s)
- Yuhang Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Jianfeng Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
- Central Laboratory, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, China
| | - Yujing Zhang
- Central Laboratory, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, China
| | - Chuang Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
- Hunan Provincial Key Laboratory of Biliary Disease Prevention and Treatment, Changsha, Hunan Province, China
- Clinical Medical Technology Research Center of Hunan Provincial for Biliary Disease Prevention and Treatment, Changsha, Hunan Province, China
| | - Yinghui Song
- Central Laboratory, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, China
| | - Sulai Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
- Central Laboratory, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, China
- Hunan Provincial Key Laboratory of Biliary Disease Prevention and Treatment, Changsha, Hunan Province, China
- Clinical Medical Technology Research Center of Hunan Provincial for Biliary Disease Prevention and Treatment, Changsha, Hunan Province, China
| |
Collapse
|
2
|
Garcia I, Cornely K, Peterson CN, Berkmen MB. Roles of the oncometabolite enantiomers of 2-hydroxyglutarate and their metabolism by diverse dehydrogenases. Essays Biochem 2024; 68:161-171. [PMID: 38919140 DOI: 10.1042/ebc20230077] [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: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024]
Abstract
2-Hydroxyglutarate (2HG) is an oncometabolite that can contribute to tumor progression. Two enantiomer forms, L-2HG and D-2HG, arise from independent pathways starting from the precursor α-ketoglutarate (αKG). L-2HG production occurs through the promiscuous activities of malate dehydrogenase (MDH) and lactate dehydrogenase (LDH) under acidic and/or hypoxic conditions. D-2HG frequently accumulates by gain-of-function mutations in the genes encoding two isoforms of isocitrate dehydrogenase (IDH1 and IDH2). Cognate metabolite repair enzymes, L- and D-2-hydroxyglutarate dehydrogenases, oxidize the enantiomers and cause abnormally high 2HG accumulation and disease when mutated. Elevated levels of either oncometabolite affect redox homeostasis, metabolism, and immune system functioning. Moreover, the oncometabolites inhibit several α-ketoglutarate-dependent dioxygenases resulting in epigenetic changes such as DNA and histone hypermethylation as well as deficiencies in DNA repair. L-2HG, and D-2HG in some cases, inhibit degradation of hypoxia-inducible factor (HIF1α), a transcription factor that alters gene expression to adapt to hypoxic conditions, favoring tumorigenesis. Patients with the rare disease 2-hydroxyglutaric aciduria (2HGA) have exceedingly high levels of 2HG, which is neurotoxic, causing developmental delays and brain abnormalities. D-2HG also has specific effects on collagen production and NADPH pools. Recently, D-2HG has been targeted in new chemotherapies aimed at disrupting the gain-of-function IDH1 and IDH2 mutants, resulting in successful clinical trials for several cancers.
Collapse
Affiliation(s)
- Ivelitza Garcia
- Department of Chemistry, Allegheny College, Meadville, PA, U.S.A
| | - Kathleen Cornely
- Department of Chemistry and Biochemistry, Providence College, Providence, RI, U.S.A
| | | | - Melanie B Berkmen
- Department of Biochemistry, Chemistry, Environment, and Physics, Suffolk University, Boston, MA, U.S.A
| |
Collapse
|
3
|
Noronha KJ, Lucas KN, Paradkar S, Edmonds J, Friedman S, Murray MA, Liu S, Sajed DP, Sachs C, Spurrier J, Raponi M, Liang J, Zeng H, Sundaram RK, Shuch B, Vasquez JC, Bindra RS. NAPRT Silencing in FH-Deficient Renal Cell Carcinoma Confers Therapeutic Vulnerabilities via NAD+ Depletion. Mol Cancer Res 2024; 22:973-988. [PMID: 38949523 PMCID: PMC11445649 DOI: 10.1158/1541-7786.mcr-23-1003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/10/2024] [Accepted: 06/21/2024] [Indexed: 07/02/2024]
Abstract
Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is caused by loss of function mutations in fumarate hydratase (FH) and results in an aggressive subtype of renal cell carcinoma with limited treatment options. Loss of FH leads to accumulation of fumarate, an oncometabolite that disrupts multiple cellular processes and drives tumor progression. High levels of fumarate inhibit alpha ketoglutarate-dependent dioxygenases, including the ten-eleven translocation (TET) enzymes, and can lead to global DNA hypermethylation. Here, we report patterns of hypermethylation in FH-mutant cell lines and tumor samples are associated with the silencing of nicotinate phosphoribosyl transferase (NAPRT), a rate-limiting enzyme in the Preiss-Handler pathway of NAD+ biosynthesis, in a subset of HLRCC cases. NAPRT is hypermethylated at a CpG island in the promoter in cell line models and patient samples, resulting in loss of NAPRT expression. We find that FH-deficient RCC models with loss of NAPRT expression, as well as other oncometabolite-producing cancer models that silence NAPRT, are extremely sensitive to nicotinamide phosphoribosyl transferase inhibitors (NAMPTi). NAPRT silencing was also associated with synergistic tumor cell killing with PARP inhibitors and NAMPTis, which was associated with effects on PAR-mediated DNA repair. Overall, our findings indicate that NAPRT silencing can be targeted in oncometabolite-producing cancers and elucidates how oncometabolite-associated hypermethylation can impact diverse cellular processes and lead to therapeutically relevant vulnerabilities in cancer cells. Implications: NAPRT is a novel biomarker for targeting NAD+ metabolism in FH-deficient HLRCCs with NAMPTis alone and targeting DNA repair processes with the combination of NAMPTis and PARP inhibitors.
Collapse
Affiliation(s)
- Katelyn J. Noronha
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut.
| | - Karlie N. Lucas
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
| | - Sateja Paradkar
- Department of Experimental Pathology, Yale University, New Haven, Connecticut.
| | - Joseph Edmonds
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
| | - Sam Friedman
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
| | - Matthew A. Murray
- Department of Experimental Pathology, Yale University, New Haven, Connecticut.
| | - Samantha Liu
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
| | - Dipti P. Sajed
- Department of Pathology, University of California Los Angeles, Los Angeles, California.
| | - Chana Sachs
- Department of Pathology, University of California Los Angeles, Los Angeles, California.
| | | | | | - Jiayu Liang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, P.R. China.
| | - Hao Zeng
- Department of Urology, West China Hospital, Sichuan University, Chengdu, P.R. China.
| | - Ranjini K. Sundaram
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
| | - Brian Shuch
- Institute of Urologic Oncology, Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California.
| | - Juan C. Vasquez
- Department of Pediatric Hematology and Oncology, Yale University, New Haven, Connecticut.
| | - Ranjit S. Bindra
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
| |
Collapse
|
4
|
Huang F, He Y. Epigenetic control of gene expression by cellular metabolisms in plants. CURRENT OPINION IN PLANT BIOLOGY 2024; 81:102572. [PMID: 38875845 DOI: 10.1016/j.pbi.2024.102572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/09/2024] [Accepted: 05/22/2024] [Indexed: 06/16/2024]
Abstract
Covalent modifications on DNA and histones can regulate eukaryotic gene expression and are often referred to as epigenetic modifications. These chemical reactions require various metabolites as donors or co-substrates, such as acetyl coenzyme A, S-adenosyl-l-methionine, and α-ketoglutarate. Metabolic processes that take place in the cytoplasm, nucleus, or other cellular compartments may impact epigenetic modifications in the nucleus. Here, we review recent advances on metabolic control of chromatin modifications and thus gene expression in plants, with a focus on the functions of nuclear compartmentalization of metabolic processes and enzymes in DNA and histone modifications. Furthermore, we discuss the functions of cellular metabolisms in fine-tuning gene expression to facilitate the responses or adaptation to environmental changes in plants.
Collapse
Affiliation(s)
- Fei Huang
- Peking-Tsinghua Center for Life Sciences & National Key Laboratory of Wheat Improvement, School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China
| | - Yuehui He
- Peking-Tsinghua Center for Life Sciences & National Key Laboratory of Wheat Improvement, School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China; Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Shandong 261325, China.
| |
Collapse
|
5
|
Hao J, Huang Z, Zhang S, Song K, Wang J, Gao C, Fang Z, Zhang N. Deciphering the multifaceted roles and clinical implications of 2-hydroxyglutarate in cancer. Pharmacol Res 2024; 209:107437. [PMID: 39349213 DOI: 10.1016/j.phrs.2024.107437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/13/2024] [Accepted: 09/24/2024] [Indexed: 10/02/2024]
Abstract
Increasing evidence indicates that 2-hydroxyglutarate (2HG) is an oncometabolite that drives tumour formation and progression. Due to mutations in isocitrate dehydrogenase (IDH) and the dysregulation of other enzymes, 2HG accumulates significantly in tumour cells. Due to its structural similarity to α-ketoglutarate (αKG), accumulated 2HG leads to the competitive inhibition of αKG-dependent dioxygenases (αKGDs), such as KDMs, TETs, and EGLNs. This inhibition results in epigenetic alterations in both tumour cells and the tumour microenvironment. This review comprehensively discusses the metabolic pathways of 2HG and the subsequent pathways influenced by elevated 2HG levels. We will delve into the molecular mechanisms by which 2HG exerts its oncogenic effects, particularly focusing on epigenetic modifications. This review will also explore the various methods available for the detection of 2HG, emphasising both current techniques and emerging technologies. Furthermore, 2HG shows promise as a biomarker for clinical diagnosis and treatment. By integrating these perspectives, this review aims to provide a comprehensive overview of the current understanding of 2HG in cancer biology, highlight the importance of ongoing research, and discuss future directions for translating these findings into clinical applications.
Collapse
Affiliation(s)
- Jie Hao
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Ziyi Huang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Siyue Zhang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Kefan Song
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Juncheng Wang
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Chao Gao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Zhiqing Fang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Ning Zhang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
6
|
Fuentes Bayne HE, Kasi PM, Ma L, Hart LL, Wong J, Spigel DR, Schnabel CA, Reeves JA, Halfdanarson TR, Treuner K, Greco FA. Personalized Therapy Selection by Integration of Molecular Cancer Classification by the 92-Gene Assay and Tumor Profiling in Patients With Cancer of Unknown Primary. JCO Precis Oncol 2024; 8:e2400191. [PMID: 39231374 PMCID: PMC11382827 DOI: 10.1200/po.24.00191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/18/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
PURPOSE Cancer of unknown primary (CUP) is a syndrome comprising metastatic cancers without a clinically identified primary site. Although patients with CUP have an unfavorable prognosis, treatment with site-specific therapies guided by clinical features, standard pathology, and molecular assays can improve overall survival. The 92-gene assay (CancerTYPE ID) is a gene expression-based classifier that helps identify the tissue of origin for metastatic cancers with unknown or uncertain diagnoses. This study reports the frequency of selected molecular aberrations of oncogenes, including KRAS, IDH1/2, BRCA1/2, and BRAF, in patients with CUP in the MOSAIC database to highlight potential treatment options. METHODS MOSAIC is a database of patients with CUP submitted for CancerTYPE ID testing and NeoTYPE biomarker testing. Tumor biopsy samples were analyzed by CancerTYPE ID for tumor type identification and further tested for molecular aberrations of oncogenes, including KRAS, IDH1/2, BRCA1/2, and BRAF. RESULTS CancerTYPE ID identified a specific tumor type in 92.5% (2,929 of 3,168) of CUP cases in the MOSAIC database. The most commonly identified histological type was adenocarcinoma (75.4%), with pancreaticobiliary being the most common molecularly diagnosed cancer (24.9%). Aberrations in KRAS, IDH1/2, BRCA, and BRAF genes were identified in 18.8% (n = 597) of biopsies. A cancer-specific US Food and Drug Administration (FDA)-approved or investigational targeted therapy was potentially available for 24.6% (n = 147) of these patients. CONCLUSION This retrospective analysis supports incorporating CancerTYPE ID into the evaluation for patients with CUP to help determine the tissue of origin and identify actionable genetic alterations. This approach may allow more patients with CUP to benefit from site-specific FDA-approved targeted therapies or enrollment into clinical trials.
Collapse
Affiliation(s)
| | | | - Li Ma
- Biotheranostics, a Hologic Company, San Diego, CA
| | | | - Jenna Wong
- Biotheranostics, a Hologic Company, San Diego, CA
| | - David R Spigel
- Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN
| | | | | | | | - Kai Treuner
- Biotheranostics, a Hologic Company, San Diego, CA
| | - F Anthony Greco
- Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN
| |
Collapse
|
7
|
Cologni R, Holschbach M, Schneider D, Bier D, Schulze A, Stegmayr C, Endepols H, Ermert J, Neumaier F, Neumaier B. Preparation and Preclinical Evaluation of 18F-Labeled Olutasidenib Derivatives for Non-Invasive Detection of Mutated Isocitrate Dehydrogenase 1 (mIDH1). Molecules 2024; 29:3939. [PMID: 39203017 PMCID: PMC11356819 DOI: 10.3390/molecules29163939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Mutations of isocitrate dehydrogenase 1 (IDH1) are key biomarkers for glioma classification, but current methods for detection of mutated IDH1 (mIDH1) require invasive tissue sampling and cannot be used for longitudinal studies. Positron emission tomography (PET) imaging with mIDH1-selective radioligands is a promising alternative approach that could enable non-invasive assessment of the IDH status. In the present work, we developed efficient protocols for the preparation of four 18F-labeled derivatives of the mIDH1-selective inhibitor olutasidenib. All four probes were characterized by cellular uptake studies with U87 glioma cells harboring a heterozygous IDH1 mutation (U87-mIDH) and the corresponding wildtype cells (U87-WT). In addition, the most promising probe was evaluated by PET imaging in healthy mice and mice bearing subcutaneous U87-mIDH and U87-WT tumors. Although all four probes inhibited mIDH1 with variable potencies, only one of them ([18F]mIDH-138) showed significantly higher in vitro uptake into U87-mIDH compared to U87-WT cells. In addition, PET imaging with [18F]mIDH-138 in mice demonstrated good in vivo stability and low non-specific uptake of the probe, but also revealed significantly higher uptake into U87-WT compared to U87-mIDH tumors. Finally, application of a two-tissue compartment model (2TCM) to the PET data indicated that preferential tracer uptake into U87-WT tumors results from higher specific binding rather than from differences in tracer perfusion. In conclusion, these results corroborate recent findings that mIDH1-selective inhibition may not directly correlate with mIDH1-selective target engagement and indicate that in vivo engagement of wildtype and mutated IDH1 may be governed by factors that are not faithfully reproduced by in vitro assays, both of which could complicate development of PET probes.
Collapse
Affiliation(s)
- Roberta Cologni
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany; (R.C.); (M.H.); (D.S.); (D.B.); (A.S.); (H.E.); (J.E.); (F.N.)
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Marcus Holschbach
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany; (R.C.); (M.H.); (D.S.); (D.B.); (A.S.); (H.E.); (J.E.); (F.N.)
| | - Daniela Schneider
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany; (R.C.); (M.H.); (D.S.); (D.B.); (A.S.); (H.E.); (J.E.); (F.N.)
| | - Dirk Bier
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany; (R.C.); (M.H.); (D.S.); (D.B.); (A.S.); (H.E.); (J.E.); (F.N.)
| | - Annette Schulze
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany; (R.C.); (M.H.); (D.S.); (D.B.); (A.S.); (H.E.); (J.E.); (F.N.)
| | - Carina Stegmayr
- Institute of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
| | - Heike Endepols
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany; (R.C.); (M.H.); (D.S.); (D.B.); (A.S.); (H.E.); (J.E.); (F.N.)
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Johannes Ermert
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany; (R.C.); (M.H.); (D.S.); (D.B.); (A.S.); (H.E.); (J.E.); (F.N.)
| | - Felix Neumaier
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany; (R.C.); (M.H.); (D.S.); (D.B.); (A.S.); (H.E.); (J.E.); (F.N.)
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany; (R.C.); (M.H.); (D.S.); (D.B.); (A.S.); (H.E.); (J.E.); (F.N.)
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
8
|
Al-khatib SM, Al-Bzour AN, Almajali MN, Jarrad TA, AL-Eitan LN, Abdo N. Analysis of IDH and EGFR as biomarkers in glioblastoma multiforme: A case-control study. Heliyon 2024; 10:e35323. [PMID: 39165999 PMCID: PMC11333891 DOI: 10.1016/j.heliyon.2024.e35323] [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: 01/30/2024] [Revised: 07/20/2024] [Accepted: 07/26/2024] [Indexed: 08/22/2024] Open
Abstract
Background Glioblastoma multiforme (GBM) is a very aggressive primary central nervous system (CNS) tumor with limited therapeutic options and poor prognosis. This study aimed to analyze the association between single nucleotide polymorphisms (SNPs), including IDH1 rs121913500C > T, IDH2 rs11540478G > A, and EGFR rs1468727C > T, and their association on the risk and overall survival of GBM patients in Jordan. Methods Using a case-control study design involving 63 GBM patients and 226 healthy controls was conducted at King Abdullah University Hospital in Jordan. DNA extraction was performed using formalin-fixed and paraffin-embedded tissue for GBM samples and blood samples for controls. SNPs analysis was performed using the Sequenom iPLEX assay sequencing technique. Survival outcomes were assessed using Cox models and hazard ratios (HR), and single-cell RNA (scRNA) analysis was performed from GSE70630. Results The study showed a significant association between genotype frequency in GBM cases and controls for specific SNPs, including IDH1 rs121913500C > T, and EGFR rs1468727C > T. The G/G genotype of rs11540478 (IDH2) was associated with better prognostic outcomes in GBM patients. The scRNA analysis demonstrated the differential expression of IDH1, IDH2, and EGFR in GBM, with enrichment in central carbon metabolism in cancer. Conclusion Our findings suggest that SNPs, particularly in IDH1 and IDH2 genes and EGFR, may serve as diagnostic and prognostic biomarkers for GBM. While the study underscores the clinical relevance of these genetic variants, further investigations with larger and more diverse populations are essential to validate and extend these associations.
Collapse
Affiliation(s)
- Sohaib M. Al-khatib
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ayah N. Al-Bzour
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Mohammad N. Almajali
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Tariq A. Jarrad
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Laith N. AL-Eitan
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Nour Abdo
- Department of Public Health, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
9
|
Carosi F, Broseghini E, Fabbri L, Corradi G, Gili R, Forte V, Roncarati R, Filippini DM, Ferracin M. Targeting Isocitrate Dehydrogenase (IDH) in Solid Tumors: Current Evidence and Future Perspectives. Cancers (Basel) 2024; 16:2752. [PMID: 39123479 PMCID: PMC11311780 DOI: 10.3390/cancers16152752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
The isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) enzymes are involved in key metabolic processes in human cells, regulating differentiation, proliferation, and oxidative damage response. IDH mutations have been associated with tumor development and progression in various solid tumors such as glioma, cholangiocarcinoma, chondrosarcoma, and other tumor types and have become crucial markers in molecular classification and prognostic assessment. The intratumoral and serum levels of D-2-hydroxyglutarate (D-2-HG) could serve as diagnostic biomarkers for identifying IDH mutant (IDHmut) tumors. As a result, an increasing number of clinical trials are evaluating targeted treatments for IDH1/IDH2 mutations. Recent studies have shown that the focus of these new therapeutic strategies is not only the neomorphic activity of the IDHmut enzymes but also the epigenetic shift induced by IDH mutations and the potential role of combination treatments. Here, we provide an overview of the current knowledge about IDH mutations in solid tumors, with a particular focus on available IDH-targeted treatments and emerging results from clinical trials aiming to explore IDHmut tumor-specific features and to identify the clinical benefit of IDH-targeted therapies and their combination strategies. An insight into future perspectives and the emerging roles of circulating biomarkers and radiomic features is also included.
Collapse
Affiliation(s)
- Francesca Carosi
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (L.F.); (G.C.)
| | | | - Laura Fabbri
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (L.F.); (G.C.)
| | - Giacomo Corradi
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (L.F.); (G.C.)
| | - Riccardo Gili
- Medical Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Valentina Forte
- Diagnostic Imaging Unit, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Roberta Roncarati
- Istituto di Genetica Molecolare “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche (CNR), 40136 Bologna, Italy;
| | - Daria Maria Filippini
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.C.); (L.F.); (G.C.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Manuela Ferracin
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
10
|
Alati C, Pitea M, Mico MC, Marafioti V, Greve B, Pratico G, Loteta B, Cogliandro F, Porto G, Policastro G, Utano G, Sgarlata A, Imbalzano L, Delfino IM, Montechiarello E, Germano J, Filippelli G, Martino M. Optimizing maintenance therapy in acute myeloid leukemia: where do we stand in the year 2024? Expert Rev Hematol 2024; 17:515-525. [PMID: 39017205 DOI: 10.1080/17474086.2024.2382300] [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: 04/09/2024] [Accepted: 07/15/2024] [Indexed: 07/18/2024]
Abstract
INTRODUCTION Despite the prognosis of patients affected by acute myeloid leukemia (AML) improved in the last decade, most patients relapse. Maintenance therapy after a chemotherapy approach with or without allogeneic stem cell transplantation could be a way to control the undetectable residual burden of leukemic cells. Several studies are being carried out as maintenance therapy in AML. Some critical points need to be defined, how the physician can choose among the various drugs available. AREAS COVERED This review discusses the advances and controversies surrounding maintenance therapy for AML patients. EXPERT OPINION Patients withFLT3-positive AML should receive midostaurin or quizartinib in the first-linesetting. For a patient initially receiving midostaurin, consider switching to sorafenib in the post-transplant setting. Because of the improved safety profile and potency, many experts will lean toward using a second-generation FLT3 inhibitor such as quizartinib or gilteritinib. Finally, no data indicate whether maintenance therapy should be prolonged until progression or for a defined period.
Collapse
Affiliation(s)
- Caterina Alati
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Martina Pitea
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Maria Caterina Mico
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Violetta Marafioti
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Bruna Greve
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Giulia Pratico
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Barbara Loteta
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Francesca Cogliandro
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Gaetana Porto
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Giorgia Policastro
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Giovanna Utano
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Annalisa Sgarlata
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Lucrezia Imbalzano
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Ilaria Maria Delfino
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Elisa Montechiarello
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | - Jessyca Germano
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| | | | - Massimo Martino
- Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO), Reggio Calabria, Italy
- Stem Cell Transplant Program CIC587, Reggio Calabria, Italy
| |
Collapse
|
11
|
Lin C, Yao C, Wang Y, Hsu Y, Yuan C, Chen T, Hsu C, Lee S, Lee J, Shih P, Kao C, Chuang P, Kuo Y, Hou H, Chou W, Tien H. IDH2 mutation accelerates TPO-induced myelofibrosis with enhanced S100a8/a9 and NFκB signaling in vivo. EJHAEM 2024; 5:738-748. [PMID: 39157630 PMCID: PMC11327712 DOI: 10.1002/jha2.983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/23/2024] [Accepted: 07/08/2024] [Indexed: 08/20/2024]
Abstract
Introduction IDH2 mutation is an unfavorable prognostic factor in patients with primary myelofibrosis (PMF) but its effect on myelofibrosis (MF) remains largely unclear. Methods In this study, we aimed to elucidate the roles of IDH2 mutation in the development and progression of MF by transcriptomic and molecular techniques using the Idh2 R172K transgenic mice. Results We found that thrombopoietin (TPO)-overexpressed Idh2 R172K (Idh2 R172K + TPO) mice had accelerated progression to MF, compared with TPO-overexpressed Idh2-wild (WT + TPO) mice, showing activation of multiple inflammatory pathways, among which nuclear factor κB (NFκB) was the most significantly enhanced. Single-cell transcriptomes of the marrow cells in early MF showed that S100a8/a9 expression was mainly confined to neutrophil progenitors in the WT + TPO mice, but highly expressed in several types of myeloid precursor cells, including the megakaryocyte progenitors in the Idh2 R172K + TPO group. Furthermore, Idh2 R172K mice at age of 18 months had larger spleens, increased S100a8/a9-Tlr4 expression, and elevated serum S100a8/a9 levels compared with WT mice. PMF patients with IDH2 mutations had higher bone marrow plasma S100A8/A9 levels than those without IDH2 mutations. Conclusion Overall, our findings showed that IDH2 mutation induced proinflammatory effects, which further exacerbated MF, as evidenced by the increase in S100a8/a9 levels and NFκB hyperactivation in Idh2 R172K + TPO mice.
Collapse
Affiliation(s)
- Chien‐Chin Lin
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Chi‐Yuan Yao
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Yu‐Hung Wang
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Yueh‐Chwen Hsu
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Chang‐Tsu Yuan
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of PathologyGraduate Institute of OncologyCollege of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of PathologyNational Taiwan University Cancer CenterTaipeiTaiwan
| | - Tsung‐Chih Chen
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Division of Hematology and Medical OncologyDepartment of Internal MedicineTaichung Veterans General HospitalTaipeiTaiwan
| | - Chia‐Lang Hsu
- Department of Medical ResearchNational Taiwan University Cancer CenterTaipeiTaiwan
| | - Sze‐Hwei Lee
- Division of Cellular TherapyDepartment of Integrated Diagnostics and TherapeuticsNational Taiwan University HospitalTaipeiTaiwan
| | - Jhih‐Yi Lee
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Pin‐Tsen Shih
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Chein‐Jun Kao
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Po‐Han Chuang
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Yuan‐Yeh Kuo
- Tai‐Cheng Stem Cell Therapy CenterNational Taiwan UniversityTaipeiTaiwan
| | - Hsin‐An Hou
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Wen‐Chien Chou
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Hwei‐Fang Tien
- Division of HematologyDepartment of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
- Division of Hematology and Medical OncologyDepartment of Internal MedicineFar Eastern Memorial HospitalNew TaipeiTaiwan
| |
Collapse
|
12
|
Park J, Peña-Tauber A, Talozzi L, Greicius MD, Guen YL. Genetic associations with human longevity are enriched for oncogenic genes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.30.24311226. [PMID: 39132489 PMCID: PMC11312667 DOI: 10.1101/2024.07.30.24311226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Human lifespan is shaped by both genetic and environmental exposures and their interaction. To enable precision health, it is essential to understand how genetic variants contribute to earlier death or prolonged survival. In this study, we tested the association of common genetic variants and the burden of rare non-synonymous variants in a survival analysis, using age-at-death (N = 35,551, median [min, max] = 72.4 [40.9, 85.2]), and last-known-age (N = 358,282, median [min, max] = 71.9 [52.6, 88.7]), in European ancestry participants of the UK Biobank. The associations we identified seemed predominantly driven by cancer, likely due to the age range of the cohort. Common variant analysis highlighted three longevity-associated loci: APOE, ZSCAN23, and MUC5B. We identified six genes whose burden of loss-of-function variants is significantly associated with reduced lifespan: TET2, ATM, BRCA2, CKMT1B, BRCA1 and ASXL1. Additionally, in eight genes, the burden of pathogenic missense variants was associated with reduced lifespan: DNMT3A, SF3B1, CHL1, TET2, PTEN, SOX21, TP53 and SRSF2. Most of these genes have previously been linked to oncogenic-related pathways and some are linked to and are known to harbor somatic variants that predispose to clonal hematopoiesis. A direction-agnostic (SKAT-O) approach additionally identified significant associations with C1orf52, TERT, IDH2, and RLIM, highlighting a link between telomerase function and longevity as well as identifying additional oncogenic genes. Our results emphasize the importance of understanding genetic factors driving the most prevalent causes of mortality at a population level, highlighting the potential of early genetic testing to identify germline and somatic variants increasing one's susceptibility to cancer and/or early death.
Collapse
Affiliation(s)
- Junyoung Park
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Andrés Peña-Tauber
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Lia Talozzi
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Michael D. Greicius
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Yann Le Guen
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94304, USA
| |
Collapse
|
13
|
Saville KM, Al-Rahahleh RQ, Siddiqui AH, Andrews ME, Roos WP, Koczor CA, Andrews JF, Hayat F, Migaud ME, Sobol RW. Oncometabolite 2-hydroxyglutarate suppresses basal protein levels of DNA polymerase beta that enhances alkylating agent and PARG inhibition induced cytotoxicity. DNA Repair (Amst) 2024; 140:103700. [PMID: 38897003 PMCID: PMC11239280 DOI: 10.1016/j.dnarep.2024.103700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/10/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024]
Abstract
Mutations in isocitrate dehydrogenase isoform 1 (IDH1) are primarily found in secondary glioblastoma (GBM) and low-grade glioma but are rare in primary GBM. The standard treatment for GBM includes radiation combined with temozolomide, an alkylating agent. Fortunately, IDH1 mutant gliomas are sensitive to this treatment, resulting in a more favorable prognosis. However, it's estimated that up to 75 % of IDH1 mutant gliomas will progress to WHO grade IV over time and develop resistance to alkylating agents. Therefore, understanding the mechanism(s) by which IDH1 mutant gliomas confer sensitivity to alkylating agents is crucial for developing targeted chemotherapeutic approaches. The base excision repair (BER) pathway is responsible for repairing most base damage induced by alkylating agents. Defects in this pathway can lead to hypersensitivity to these agents due to unresolved DNA damage. The coordinated assembly and disassembly of BER protein complexes are essential for cell survival and for maintaining genomic integrity following alkylating agent exposure. These complexes rely on poly-ADP-ribose formation, an NAD+-dependent post-translational modification synthesized by PARP1 and PARP2 during the BER process. At the lesion site, poly-ADP-ribose facilitates the recruitment of XRCC1. This scaffold protein helps assemble BER proteins like DNA polymerase beta (Polβ), a bifunctional DNA polymerase containing both DNA synthesis and 5'-deoxyribose-phosphate lyase (5'dRP lyase) activity. Here, we confirm that IDH1 mutant glioma cells have defective NAD+ metabolism, but still produce sufficient nuclear NAD+ for robust PARP1 activation and BER complex formation in response to DNA damage. However, the overproduction of 2-hydroxyglutarate, an oncometabolite produced by the IDH1 R132H mutant protein, suppresses BER capacity by reducing Polβ protein levels. This defines a novel mechanism by which the IDH1 mutation in gliomas confers cellular sensitivity to alkylating agents and to inhibitors of the poly-ADP-ribose glycohydrolase, PARG.
Collapse
Affiliation(s)
- Kate M Saville
- Department of Pharmacology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States
| | - Rasha Q Al-Rahahleh
- Department of Pharmacology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States; Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, United States
| | - Aisha H Siddiqui
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, United States
| | - Morgan E Andrews
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, United States
| | - Wynand P Roos
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, United States
| | - Christopher A Koczor
- Department of Pharmacology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States
| | - Joel F Andrews
- Department Biochemistry and Molecular Biology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States
| | - Faisal Hayat
- Department of Pharmacology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States
| | - Marie E Migaud
- Department of Pharmacology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States
| | - Robert W Sobol
- Department of Pharmacology & Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, United States; Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, United States.
| |
Collapse
|
14
|
Gao H, Wei L, Indulkar S, Nguyen TTL, Liu D, Ho MF, Zhang C, Li H, Weinshilboum RM, Ingle JN, Wang L. Androgen receptor-mediated pharmacogenomic expression quantitative trait loci: implications for breast cancer response to AR-targeting therapy. Breast Cancer Res 2024; 26:111. [PMID: 38965614 PMCID: PMC11225427 DOI: 10.1186/s13058-024-01861-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 06/20/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Endocrine therapy is the most important treatment modality of breast cancer patients whose tumors express the estrogen receptor α (ERα). The androgen receptor (AR) is also expressed in the vast majority (80-90%) of ERα-positive tumors. AR-targeting drugs are not used in clinical practice, but have been evaluated in multiple trials and preclinical studies. METHODS We performed a genome-wide study to identify hormone/drug-induced single nucleotide polymorphism (SNP) genotype - dependent gene-expression, known as PGx-eQTL, mediated by either an AR agonist (dihydrotestosterone) or a partial antagonist (enzalutamide), utilizing a previously well characterized lymphoblastic cell line panel. The association of the identified SNPs-gene pairs with breast cancer phenotypes were then examined using three genome-wide association (GWAS) studies that we have published and other studies from the GWAS catalog. RESULTS We identified 13 DHT-mediated PGx-eQTL loci and 23 Enz-mediated PGx-eQTL loci that were associated with breast cancer outcomes post ER antagonist or aromatase inhibitors (AI) treatment, or with pharmacodynamic (PD) effects of AIs. An additional 30 loci were found to be associated with cancer risk and sex-hormone binding globulin levels. The top loci involved the genes IDH2 and TMEM9, the expression of which were suppressed by DHT in a PGx-eQTL SNP genotype-dependent manner. Both of these genes were overexpressed in breast cancer and were associated with a poorer prognosis. Therefore, suppression of these genes by AR agonists may benefit patients with minor allele genotypes for these SNPs. CONCLUSIONS We identified AR-related PGx-eQTL SNP-gene pairs that were associated with risks, outcomes and PD effects of endocrine therapy that may provide potential biomarkers for individualized treatment of breast cancer.
Collapse
Affiliation(s)
- Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Lixuan Wei
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Shreya Indulkar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Thanh Thanh L Nguyen
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Duan Liu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Ming-Fen Ho
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Richard M Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA.
| |
Collapse
|
15
|
Jen WY, Kantarjian H, Kadia TM, DiNardo CD, Issa GC, Short NJ, Yilmaz M, Borthakur G, Ravandi F, Daver NG. Combination therapy with novel agents for acute myeloid leukaemia: Insights into treatment of a heterogenous disease. Br J Haematol 2024; 205:30-47. [PMID: 38724457 DOI: 10.1111/bjh.19519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/27/2024] [Indexed: 07/13/2024]
Abstract
The treatment landscape of acute myeloid leukaemia (AML) is evolving rapidly. Venetoclax in combination with intensive chemotherapy or doublets or triplets with targeted or immune therapies is the focus of numerous ongoing trials. The development of mutation-targeted therapies has greatly enhanced the treatment armamentarium, with FLT3 inhibitors and isocitrate dehydrogenase inhibitors improving outcomes in frontline and relapsed/refractory (RR) AML, and menin inhibitors showing efficacy in RR NPM1mut and KMT2A-rearranged AML. With so many new drugs approved, the number of potential combinatorial approaches to leverage the maximal benefit of these agents has increased dramatically, while at the same time introducing clinical challenges, such as key preclinical and clinical data supporting the development of combinatorial therapy, how to optimally combine or sequence these novel agents, how to optimise dose and duration to maintain safety while enhancing efficacy, the optimal duration of therapy and the role of measurable residual disease in decision-making in both intensive and low-intensity therapy settings. In this review, we will outline the evidence leading to the approval of key agents in AML, their on-label current approvals and how they may be optimally combined in a safe and deliverable fashion to further improve outcomes in AML.
Collapse
Affiliation(s)
- Wei-Ying Jen
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tapan M Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ghayas C Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Musa Yilmaz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
16
|
Zhu G, Cao L, Wu J, Xu M, Zhang Y, Wu M, Li J. Co-morbid intersections of cancer and cardiovascular disease and targets for natural drug action: Reprogramming of lipid metabolism. Biomed Pharmacother 2024; 176:116875. [PMID: 38850662 DOI: 10.1016/j.biopha.2024.116875] [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: 04/01/2024] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Cancer and cardiovascular diseases are major contributors to global morbidity and mortality, and their seemingly separate pathologies are intricately intertwined. In the context of cancer, the cardiovascular disease encompasses not only the side effects arising from anti-tumor treatments but also the metabolic shifts induced by oncological conditions. A growing body of research indicates that lipid metabolic reprogramming serves as a distinctive hallmark of tumors. Furthermore, anomalies in lipid metabolism play a significant role in the development of cardiovascular disease. This study delves into the cardiac implications of lipid metabolic reprogramming within the cancer context, closely examining abnormalities in lipid metabolism present in tumors, cardiac tissue, and immune cells within the microenvironment. Additionally, we examined risk factors such as obesity and anti-tumor therapy. Despite progress, a gap remains in the availability of drugs targeting lipid metabolism modulation for treating tumors and mitigating cardiac risk, with limited advancement seen in prior studies. Here, we present a review of previous research on natural drugs that exhibit both shared and distinct therapeutic effects on tumors and cardiac health by modulating lipid metabolism. Our aim is to provide insights for potential drug development.
Collapse
Affiliation(s)
- Guanghui Zhu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Luchang Cao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jingyuan Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Manman Xu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ying Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Min Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Jie Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
17
|
Ciccarone F, Ciriolo MR. Reprogrammed mitochondria: a central hub of cancer cell metabolism. Biochem Soc Trans 2024; 52:1305-1315. [PMID: 38716960 DOI: 10.1042/bst20231090] [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] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 06/27/2024]
Abstract
Mitochondria represent the metabolic hub of normal cells and play this role also in cancer but with different functional purposes. While cells in differentiated tissues have the prerogative of maintaining basal metabolism and support the biosynthesis of specialized products, cancer cells have to rewire the metabolic constraints imposed by the differentiation process. They need to balance the bioenergetic supply with the anabolic requirements that entail the intense proliferation rate, including nucleotide and membrane lipid biosynthesis. For this aim, mitochondrial metabolism is reprogrammed following the activation of specific oncogenic pathways or due to specific mutations of mitochondrial proteins. The main process leading to mitochondrial metabolic rewiring is the alteration of the tricarboxylic acid cycle favoring the appropriate orchestration of anaplerotic and cataplerotic reactions. According to the tumor type or the microenvironmental conditions, mitochondria may decouple glucose catabolism from mitochondrial oxidation in favor of glutaminolysis or disable oxidative phosphorylation for avoiding harmful production of free radicals. These and other metabolic settings can be also determined by the neo-production of oncometabolites that are not specific for the tissue of origin or the accumulation of metabolic intermediates able to boost pro-proliferative metabolism also impacting epigenetic/transcriptional programs. The full characterization of tumor-specific mitochondrial signatures may provide the identification of new biomarkers and therapeutic opportunities based on metabolic approaches.
Collapse
Affiliation(s)
- Fabio Ciccarone
- Department of Biology, University of Rome 'Tor Vergata', 00133 Rome, Italy
- IRCCS San Raffaele Roma, 00166 Rome, Italy
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome 'Tor Vergata', 00133 Rome, Italy
- IRCCS San Raffaele Roma, 00166 Rome, Italy
| |
Collapse
|
18
|
Burgermeister S, Stoykova S, Krebs FS, Zoete V, Mbefo M, Egervari K, Reinhard A, Bisig B, Hewer E. Methylation-Based Characterization of a New IDH2 Mutation in Sinonasal Undifferentiated Carcinoma. Int J Mol Sci 2024; 25:6518. [PMID: 38928223 PMCID: PMC11204065 DOI: 10.3390/ijms25126518] [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: 04/22/2024] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Mutations affecting codon 172 of the isocitrate dehydrogenase 2 (IDH2) gene define a subgroup of sinonasal undifferentiated carcinomas (SNUCs) with a relatively favorable prognosis and a globally hypermethylated phenotype. They are also recurrent (along with IDH1 mutations) in gliomas, acute myeloid leukemia, and intrahepatic cholangiocarcinoma. Commonly reported mutations, all associated with aberrant IDH2 enzymatic activity, include R172K, R172S, R172T, R172G, and R172M. We present a case of SNUC with a never-before-described IDH2 mutation, R172A. Our report compares the methylation pattern of our sample to other cases from the Gene Expression Omnibus database. Hierarchical clustering suggests a strong association between our sample and other IDH-mutant SNUCs and a clear distinction between sinonasal normal tissues and tumors. Principal component analysis (PCA), using 100 principal components explaining 94.5% of the variance, showed the position of our sample to be within 1.02 standard deviation of the other IDH-mutant SNUCs. A molecular modeling analysis of the IDH2 R172A versus other R172 variants provides a structural explanation to how they affect the protein active site. Our findings thus suggest that the R172A mutation in IDH2 confers a gain of function similar to other R172 mutations in IDH2, resulting in a similar hypermethylated profile.
Collapse
Affiliation(s)
- Simon Burgermeister
- Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (S.S.); (M.M.); (B.B.)
| | - Simona Stoykova
- Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (S.S.); (M.M.); (B.B.)
| | - Fanny S. Krebs
- Computer-Aided Molecular Engineering, Department of Oncology UNIL-CHUV, University of Lausanne, 1066 Epalinges, Switzerland; (F.S.K.); (V.Z.)
- Ludwig Institute for Cancer Research, 1066 Epalinges, Switzerland
| | - Vincent Zoete
- Computer-Aided Molecular Engineering, Department of Oncology UNIL-CHUV, University of Lausanne, 1066 Epalinges, Switzerland; (F.S.K.); (V.Z.)
- Ludwig Institute for Cancer Research, 1066 Epalinges, Switzerland
- Molecular Modelling Group, SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Martial Mbefo
- Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (S.S.); (M.M.); (B.B.)
| | - Kristof Egervari
- Service of Clinical Pathology, Department of Diagnostics, Geneva University Hospitals, 1206 Geneva, Switzerland;
| | - Antoine Reinhard
- Department of Otorhinolaryngology-Head and Neck Surgery, Lausanne University Hospital, 1011 Lausanne, Switzerland;
| | - Bettina Bisig
- Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (S.S.); (M.M.); (B.B.)
| | - Ekkehard Hewer
- Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (S.S.); (M.M.); (B.B.)
| |
Collapse
|
19
|
Trexler M, Bányai L, Kerekes K, Patthy L. Arginines of the CGN codon family are Achilles' heels of cancer genes. Sci Rep 2024; 14:11715. [PMID: 38778164 PMCID: PMC11111792 DOI: 10.1038/s41598-024-62553-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Recent studies have revealed that arginine is the most favorable target of amino acid alteration in most cancer types and it has been suggested that the high preference for arginine mutations reflects the critical roles of this amino acid in the function of proteins. High rates of mutations of arginine residues in cancer, however, might also be due to increased mutability of arginine codons of the CGN family as the CpG dinucleotides of these codons may be methylated. In the present work we have analyzed spectra of single base substitutions of cancer genes (oncogenes, tumor suppressor genes) and passenger genes in cancer tissues to assess the contributions of CpG hypermutability and selection to arginine mutations. Our studies have shown that arginines encoded by the CGN codon family display higher rates of mutation in both cancer genes and passenger genes than arginine codons AGA and AGG that are devoid of CpG dinucleotide, suggesting that the predominance of arginine mutations in cancer is primarily due to CpG hypermutability, rather than selection for arginine replacement. Nevertheless, our results also suggest that CGN codons for arginines may serve as Achilles' heels of cancer genes. CpG hypermutability of key arginines of proto-oncogenes, leading to high rates of recurrence of driver mutations, contributes significantly to carcinogenesis. Similarly, our results indicate that hypermutability of the CpG dinucleotide of CGA codons (converting them to TGA stop codons) contributes significantly to recurrent truncation and inactivation of tumor suppressor genes.
Collapse
Affiliation(s)
- Mária Trexler
- Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - László Bányai
- Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - Krisztina Kerekes
- Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary
| | - László Patthy
- Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, 1117, Hungary.
| |
Collapse
|
20
|
Norden PR, Wedan RJ, Longenecker JZ, Preston SEJ, Graber N, Ols O, Canfield M, McLaughlin E, Nowinski SM. Mitochondrial Phosphopantetheinylation is Required for Oxidative Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.592977. [PMID: 38766035 PMCID: PMC11100772 DOI: 10.1101/2024.05.09.592977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Sub-cellular compartmentalization of metabolism has important implications for the local production of metabolites and redox co-factors, as well as pathway regulation. 4'-phosphopantetheinyl (4'PP) groups are essential co-factors derived from coenzyme A and added to target proteins in both the cytoplasm and mitochondria by p hospho p antetheinyl transferase (PPTase) enzymes. Mammals express only one PPTase, thought to localize to the cytoplasm: aminoadipate semialdehyde dehydrogenase phosphopantetheinyl transferase (AASDHPPT); raising the question of how mitochondrial proteins are 4'PP-modified. We found that AASDHPPT is required for mitochondrial respiration and oxidative metabolism via the mitochondrial fatty acid synthesis (mtFAS) pathway. Moreover, we discovered that a pool of AASDHPPT localizes to the mitochondrial matrix via an N-terminal mitochondrial targeting sequence contained within the first 13 amino acids of the protein. Our data show that mitochondrial localization of AASDHPPT is required to support mtFAS function, and further identify two variants in Aasdhppt that are likely pathogenic in humans.
Collapse
|
21
|
Aslani S, Armstrong DW. Fast, sensitive LC-MS resolution of α -hydroxy acid biomarkers via SPP-teicoplanin and an alternative UV detection approach. Anal Bioanal Chem 2024; 416:3007-3017. [PMID: 38565719 DOI: 10.1007/s00216-024-05248-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/20/2024] [Accepted: 02/28/2024] [Indexed: 04/04/2024]
Abstract
Enantioseparation of α -hydroxy acids is essential since specific enantiomers of these compounds can be used as disease biomarkers for diagnosis and prognosis of cancer, brain diseases, kidney diseases, diabetes, etc., as well as in the food industry to ensure quality. HPLC methods were developed for the enantioselective separation of 11 α -hydroxy acids using a superficially porous particle-based teicoplanin (TeicoShell) chiral stationary phase. The retention behaviors observed for the hydroxy acids were HILIC, reversed phase, and ion-exclusion. While both mass spectrometry and UV spectroscopy detection methods could be used, specific mobile phases containing ammonium formate and potassium dihydrogen phosphate, respectively, were necessary with each approach. The LC-MS mode was approximately two orders of magnitude more sensitive than UV detection. Mobile phase acidity and ionic strength significantly affected enantioresolution and enantioselectivity. Interestingly, higher ionic strength resulted in increased retention and enantioresolution. It was noticed that for formate-containing mobile phases, using acetonitrile as the organic modifier usually resulted in greater enantioresolution compared to methanol. However, sometimes using acetonitrile with high ammonium formate concentrations led to lengthy retention times which could be avoided by using methanol as the organic modifier. Additionally, the enantiomeric purities of single enantiomer standards were determined and it was shown that almost all standards contained some levels of enantiomeric impurities.
Collapse
Affiliation(s)
- Saba Aslani
- Department of Chemistry and Biochemistry, University of Texas at Arlington, 700 Planetarium Place, Arlington, TX, 76019, USA
| | - Daniel W Armstrong
- Department of Chemistry and Biochemistry, University of Texas at Arlington, 700 Planetarium Place, Arlington, TX, 76019, USA.
| |
Collapse
|
22
|
Zhang J, Wang Y, Yang Y, Han Y, Yu Y, Hu Y, Liang S, Sun Q, Shang D, Bi J, Cui G, Yan L. Noninvasive Isocitrate Dehydrogenase 1 Status Prediction in Grade II/III Glioma Based on Magnetic Resonance Images: A Transfer Learning Strategy. J Comput Assist Tomogr 2024; 48:449-458. [PMID: 38271541 DOI: 10.1097/rct.0000000000001575] [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: 01/27/2024]
Abstract
OBJECTIVE The aim of this study was to evaluate transfer learning combined with various convolutional neural networks (TL-CNNs) in predicting isocitrate dehydrogenase 1 ( IDH1 ) status of grade II/III gliomas. METHODS Grade II/III glioma patients diagnosed at the Tangdu Hospital (August 2009 to May 2017) were retrospectively enrolled, including 54 patients with IDH1 mutant and 56 patients with wild-type IDH1 . Convolutional neural networks, AlexNet, GoogLeNet, ResNet, and VGGNet were fine-tuned with T2-weighted imaging (T2WI), fluid attenuation inversion recovery (FLAIR), and contrast-enhanced T1-weighted imaging (T1CE) images. The single-modal networks were integrated with averaged sigmoid probabilities, logistic regression, and support vector machine. FLAIR-T1CE-fusion (FC-fusion), T2WI-T1CE-fusion (TC-fusion), and FLAIR-T2WI-T1CE-fusion (FTC-fusion) were used for fine-tuning TL-CNNs. RESULTS IDH1 -mutant prediction accuracies using AlexNet, GoogLeNet, ResNet, and VGGNet achieved 70.0% (AUC = 0.660), 65.0% (AUC = 0.600), 70.0% (AUC = 0.700), and 80.0% (AUC = 0.730) for T2WI images, 70.0% (AUC = 0.660), 70.0% (AUC = 0.620), 70.0% (AUC = 0.710), and 80.0% (AUC = 0.720) for FLAIR images, and 73.7% (AUC = 0.744), 73.7% (AUC = 0.656), 73.7% (AUC = 0.633), and 73.7% (AUC = 0.700) for T1CE images, respectively. The highest AUC (0.800) was achieved using VGGNet and FC-fusion images. CONCLUSIONS TL-CNNs (especially VGGNet) had a potential predictive value for IDH1 -mutant status of grade II/III gliomas.
Collapse
Affiliation(s)
- Jin Zhang
- From the Department of Radiology and Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital
| | - Yuyao Wang
- From the Department of Radiology and Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital
| | - Yang Yang
- From the Department of Radiology and Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital
| | - Yu Han
- From the Department of Radiology and Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital
| | - Ying Yu
- From the Department of Radiology and Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital
| | - Yuchuan Hu
- From the Department of Radiology and Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital
| | - Shouheng Liang
- From the Department of Radiology and Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital
| | - Qian Sun
- From the Department of Radiology and Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital
| | - Danting Shang
- From the Department of Radiology and Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital
| | - Jiajun Bi
- College of Basic Medicine, the Fourth Military Medical University (Air Force Medical University), Xi'an, Shaanxi, China
| | - Guangbin Cui
- From the Department of Radiology and Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital
| | - Linfeng Yan
- From the Department of Radiology and Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital
| |
Collapse
|
23
|
Li XY, Xu YY, Wu SY, Zeng XX, Zhou Y, Cheng GB. Identification of ALDOB as a novel prognostic biomarker in kidney clear cell renal cell carcinoma. Heliyon 2024; 10:e29368. [PMID: 38655323 PMCID: PMC11035059 DOI: 10.1016/j.heliyon.2024.e29368] [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: 01/07/2024] [Revised: 04/03/2024] [Accepted: 04/07/2024] [Indexed: 04/26/2024] Open
Abstract
Kidney clear cell renal cell carcinoma (KIRC) is also the most lethal subtype among all kidney cancer subtypes, posing a severe threat to public health. Therefore, it is crucial to identify new, reliable biomarkers in KIRC. Therefore, it is crucial to identify novel, reliable biomarkers associated with KIRC. We analyzed RNA sequence results from TCGA and several GEO datasets. The commonly deregulated gene, ALDOB, was found in multiple data and confirmed its important prognostic value. Subsequently, we explored the specific mechanism by which ALDOB regulates anti-tumor immunity through in vivo and in vitro experiments. We found that ALDOB may play a role in regulating tumor growth by regulating CD8+ T cell infiltration. This is consistent with the results of our immune infiltration-related analysis. In addition, we have also discovered the effect of ALDOB in previous studies on other cancer types. Finally, we concluded that ALDOB may have potential reference value for immunotherapy and can also be used as an independent predictor of prognosis in KIRC.
Collapse
Affiliation(s)
- Xiao-yang Li
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - You-yao Xu
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Sen-yan Wu
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Xi-xi Zeng
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- The Joint Innovation Center for Health and Medicine, Quzhou People's Hospital, The Quzhou Afliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Yan Zhou
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- Department of Nephrology, Quzhou People's Hospital, The Quzhou Afliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Guo-bin Cheng
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| |
Collapse
|
24
|
Li JJ, Yu T, Zeng P, Tian J, Liu P, Qiao S, Wen S, Hu Y, Liu Q, Lu W, Zhang H, Huang P. Wild-type IDH2 is a therapeutic target for triple-negative breast cancer. Nat Commun 2024; 15:3445. [PMID: 38658533 PMCID: PMC11043430 DOI: 10.1038/s41467-024-47536-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Mutations in isocitrate dehydrogenases (IDH) are oncogenic events due to the generation of oncogenic metabolite 2-hydroxyglutarate. However, the role of wild-type IDH in cancer development remains elusive. Here we show that wild-type IDH2 is highly expressed in triple negative breast cancer (TNBC) cells and promotes their proliferation in vitro and tumor growth in vivo. Genetic silencing or pharmacological inhibition of wt-IDH2 causes a significant increase in α-ketoglutarate (α-KG), indicating a suppression of reductive tricarboxylic acid (TCA) cycle. The aberrant accumulation of α-KG due to IDH2 abrogation inhibits mitochondrial ATP synthesis and promotes HIF-1α degradation, leading to suppression of glycolysis. Such metabolic double-hit results in ATP depletion and suppression of tumor growth, and renders TNBC cells more sensitive to doxorubicin treatment. Our study reveals a metabolic property of TNBC cells with active utilization of glutamine via reductive TCA metabolism, and suggests that wild-type IDH2 plays an important role in this metabolic process and could be a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Jiang-Jiang Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Tiantian Yu
- Metabolic Innovation Center, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, 510080, China
| | - Peiting Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Jingyu Tian
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Panpan Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Shuang Qiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Yumin Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Qiao Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Wenhua Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Hui Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Metabolic Innovation Center, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, 510080, China
| | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
- Metabolic Innovation Center, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, 510080, China.
| |
Collapse
|
25
|
Dang Q, Li B, Jin B, Ye Z, Lou X, Wang T, Wang Y, Pan X, Hu Q, Li Z, Ji S, Zhou C, Yu X, Qin Y, Xu X. Cancer immunometabolism: advent, challenges, and perspective. Mol Cancer 2024; 23:72. [PMID: 38581001 PMCID: PMC10996263 DOI: 10.1186/s12943-024-01981-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/06/2024] [Indexed: 04/07/2024] Open
Abstract
For decades, great strides have been made in the field of immunometabolism. A plethora of evidence ranging from basic mechanisms to clinical transformation has gradually embarked on immunometabolism to the center stage of innate and adaptive immunomodulation. Given this, we focus on changes in immunometabolism, a converging series of biochemical events that alters immune cell function, propose the immune roles played by diversified metabolic derivatives and enzymes, emphasize the key metabolism-related checkpoints in distinct immune cell types, and discuss the ongoing and upcoming realities of clinical treatment. It is expected that future research will reduce the current limitations of immunotherapy and provide a positive hand in immune responses to exert a broader therapeutic role.
Collapse
Affiliation(s)
- Qin Dang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Borui Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bing Jin
- School of Clinical Medicine, Zhengzhou University, Zhengzhou, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xin Lou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Ting Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xuan Pan
- Department of Hepatobiliary Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Qiangsheng Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Zheng Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chenjie Zhou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
26
|
Balaji E V, Satarker S, Kumar BH, Pandey S, Birangal SR, Nayak UY, Pai KSR. In-silico lead identification of the pan-mutant IDH1 and IDH2 inhibitors to target glioblastoma. J Biomol Struct Dyn 2024; 42:3764-3789. [PMID: 37227789 DOI: 10.1080/07391102.2023.2215884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023]
Abstract
Glioblastoma (GBM) is an aggressive malignant type of brain tumor. Targeting one single intracellular pathway might not alleviate the disease, rather it activates the other molecular pathways that lead to the worsening of the disease condition. Therefore, in this study, we attempted to target both isocitrate dehydrogenase 1 (IDH1) and IDH2, which are one of the most commonly mutated proteins in GBM and other cancer types. Here, standard precision and extra precision docking, IFD, MM-GBSA, QikProp, and molecular dynamics (MD) simulation were performed to identify the potential dual inhibitor for IDH1 and IDH2 from the enamine database containing 59,161 ligands. Upon docking the ligands with IDH1 (PDB: 6VEI) and IDH2 (PDB: 6VFZ), the top eight ligands were selected, based on the XP Glide score. These ligands produced favourable MMGBSA scores and ADME characteristics. Finally, the top four ligands 12953, 44825, 51295, and 53210 were subjected to MD analysis. Interestingly, 53210 showed maximum interaction with Gln 277 for 99% in IDH1 and Gln 316 for 100% in IDH2, which are the crucial amino acids for the inhibitory function of IDH1 and IDH2 to target GBM. Therefore, the present study attempts to identify the novel molecules which could possess a pan-inhibitory action on both IDH1 and IDH that could be crucial in the management of GBM. Yet further evaluation involving in vitro and in vivo studies is warranted to support the data in our current study.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Vignesh Balaji E
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sairaj Satarker
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - B Harish Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Samyak Pandey
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sumit Raosaheb Birangal
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Usha Y Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - K Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
27
|
Katole VR, Kaple M. Unraveling the Landscape of Pediatric Glioblastoma Biomarkers: A Comprehensive Review of Enhancing Diagnostics and Therapeutic Insights. Cureus 2024; 16:e57272. [PMID: 38686271 PMCID: PMC11057698 DOI: 10.7759/cureus.57272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
Glioblastoma, the most common and aggressive form of primary brain tumor, poses significant challenges to patients, caregivers, and clinicians alike. Pediatric glioblastoma is a rare and aggressive brain tumor that presents unique challenges in treatment. It differs from its adult counterpart in terms of genetic and molecular characteristics. Its incidence is relatively low, but the prognosis remains grim due to its aggressive behavior. Diagnosis relies on imaging techniques and histopathological analysis. The rarity of the disease underscores the need for effective treatment strategies. In recent years, the quest to understand and manage pediatric glioblastoma has seen a significant shift towards unraveling the intricate landscape of biomarkers. Surgery remains a cornerstone of glioblastoma management, aiming to resect as much of the tumor as possible. Glioblastoma's infiltrative nature presents challenges in achieving a complete surgical resection. This comprehensive review delves into the realm of pediatric glioblastoma biomarkers, shedding light on their potential to not only revolutionize diagnostics but also shape therapeutic strategies. From personalized treatment selection to the development of targeted therapies, the potential impact of these biomarkers on clinical outcomes is undeniable. Moreover, this review underscores the substantial implications of biomarker-driven approaches for therapeutic interventions. All advancements in targeted therapies and immunotherapy hold promise for the treatment of pediatric glioblastoma. The genetic profiling of tumors allows for personalized approaches, potentially improving treatment efficacy. The ethical dilemmas surrounding pediatric cancer treatment, particularly balancing potential benefits with risks, are complex. Ongoing clinical trials and preclinical research suggest exciting avenues for future interventions.
Collapse
Affiliation(s)
- Vedant R Katole
- Department of Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Meghali Kaple
- Department of Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
28
|
Agosti E, Zeppieri M, Antonietti S, Ius T, Fontanella MM, Panciani PP. Advancing the Management of Skull Base Chondrosarcomas: A Systematic Review of Targeted Therapies. J Pers Med 2024; 14:261. [PMID: 38541003 PMCID: PMC10971225 DOI: 10.3390/jpm14030261] [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: 02/01/2024] [Revised: 02/24/2024] [Accepted: 02/25/2024] [Indexed: 03/28/2024] Open
Abstract
Background: Chondrosarcomas rank as the second most common primary bone malignancy. Characterized by the production of a cartilaginous matrix, these tumors typically exhibit resistance to both radiotherapy (RT) and chemotherapy (CT), resulting in overall poor outcomes: a high rate of mortality, especially among children and adolescents. Due to the considerable resistance to current conventional therapies such as surgery, CT, and RT, there is an urgent need to identify factors contributing to resistance and discover new strategies for optimal treatment. Over the past decade, researchers have delved into the dysregulation of genes associated with tumor development and therapy resistance to identify potential therapeutic targets for overcoming resistance. Recent studies have suggested several promising biomarkers and therapeutic targets for chondrosarcoma, including isocitrate dehydrogenase (IDH1/2) and COL2A1. Molecule-targeting agents and immunotherapies have demonstrated favorable antitumor activity in clinical studies involving patients with advanced chondrosarcomas. In this systematic review, we delineate the clinical features of chondrosarcoma and provide a summary of gene dysregulation and mutation associated with tumor development, as well as targeted therapies as a promising molecular approach. Finally, we analyze the probable role of the tumor microenvironment in chondrosarcoma drug resistance. Methods: A systematic search was conducted across major medical databases (PubMed, Embase, and Cochrane Library) up to 10 November 2023. The search strategy utilized relevant Medical Subject Heading (MeSH) terms and keywords related to “chondrosarcomas”, “target therapies”, “immunotherapies”, and “outcomes”. The studies included in this review consist of randomized controlled trials, non-randomized controlled trials, and cohort studies reporting on the use of target therapies for the treatment of chondrosarcoma in human subjects. Results: Of the initial 279 articles identified, 40 articles were included in the article. The exclusion of 140 articles was due to reasons such as irrelevance, non-reporting of selected results, systematic literature review or meta-analysis, and lack of details on the method/results. Three tables highlighted clinical studies, preclinical studies, and ongoing clinical trials, encompassing 13, 7, and 20 studies, respectively. For the clinical study, a range of molecular targets, such as death receptors 4/5 (DR4 and DR5) (15%), platelet-derived growth factor receptor-alpha or -beta (PDGFR-α, PDGFR-β) (31%), were investigated. Adverse events were mainly constitutional symptoms emphasizing that to improve therapy tolerance, careful observation and tailored management are essential. Preclinical studies analyzed various molecular targets such as DR4/5 (28.6%) and COX-2 (28.6%). The prevalent indicator of antitumoral activity was the apoptotic rate of both a single agent (tumor necrosis factor-related apoptosis-inducing ligand: TRAIL) and double agents (TRAIL-DOX, TRAIL-MG132). Ongoing clinical trials, the majority in Phase II (53.9%), highlighted possible therapeutic strategies such as IDH1 inhibitors and PD-1/PD-L1 inhibitors (30.8%). Conclusions: The present review offers a comprehensive analysis of targeted therapeutics for skull base chondrosarcomas, highlighting a complex landscape characterized by a range of treatment approaches and new opportunities for tailored interventions. The combination of results from molecular research and clinical trials emphasizes the necessity for specialized treatment strategies and the complexity of chondrosarcoma biology.
Collapse
Affiliation(s)
- Edoardo Agosti
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazza Spedali Civili 1, 25123 Brescia, Italy; (E.A.)
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy
| | - Sara Antonietti
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazza Spedali Civili 1, 25123 Brescia, Italy; (E.A.)
| | - Tamara Ius
- Neurosurgery Unit, Head-Neck and NeuroScience Department, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy
| | - Marco Maria Fontanella
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazza Spedali Civili 1, 25123 Brescia, Italy; (E.A.)
| | - Pier Paolo Panciani
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Piazza Spedali Civili 1, 25123 Brescia, Italy; (E.A.)
| |
Collapse
|
29
|
Pawaskar R, Huang KZ, Pham H, Nagrial A, Wong M, O’Neill S, Pleass H, Yuen L, Lam VWT, Richardson A, Pang T, Nahm CB. Systematic Review of Preoperative Prognostic Biomarkers in Perihilar Cholangiocarcinoma. Cancers (Basel) 2024; 16:698. [PMID: 38398089 PMCID: PMC10886549 DOI: 10.3390/cancers16040698] [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: 01/18/2024] [Revised: 02/03/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Perihilar cholangiocarcinoma (pCCA) is an uncommon malignancy with generally poor prognosis. Surgery is the primary curative treatment; however, the perioperative mortality and morbidity rates are high, with a low 5-year survival rate. Use of preoperative prognostic biomarkers to predict survival outcomes after surgery for pCCA are not well-established currently. This systematic review aimed to identify and summarise preoperative biomarkers associated with survival in pCCA, thereby potentially improving treatment decision-making. The Embase, Medline, and Cochrane databases were searched, and a systematic review was performed using the PRISMA guidelines. English-language studies examining the association between serum and/or tissue-derived biomarkers in pCCA and overall and/or disease-free survival were included. Our systematic review identified 64 biomarkers across 48 relevant studies. Raised serum CA19-9, bilirubin, CEA, neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR) and tumour MMP9, and low serum albumin were most associated with poorer survival; however, the cutoff values used widely varied. Several promising molecular markers with prognostic significance were also identified, including tumour HMGA2, MUC5AC/6, IDH1, PIWIL2, and DNA index. In conclusion, several biomarkers have been identified in serum and tumour specimens that prognosticate overall and disease-free survival after pCCA resection. These, however, require external validation in large cohort studies and/or in preoperatively obtained specimens, especially tissue biopsy, to recommend their use.
Collapse
Affiliation(s)
- Rishaan Pawaskar
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
| | | | - Helen Pham
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
| | - Adnan Nagrial
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW 2145, Australia;
| | - Mark Wong
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW 2145, Australia;
| | - Siobhan O’Neill
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW 2145, Australia;
| | - Henry Pleass
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
| | - Lawrence Yuen
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
| | - Vincent W. T. Lam
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
- Macquarie University Medical School, Macquarie University, Sydney, NSW 2145, Australia
| | - Arthur Richardson
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
| | - Tony Pang
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
| | - Christopher B. Nahm
- Department of Upper GI Surgery, Westmead Hospital, Sydney, NSW 2145, Australia; (R.P.); (H.P.); (H.P.); (L.Y.); (V.W.T.L.); (A.R.); (T.P.)
- Westmead Hospital, Sydney, NSW 2145, Australia;
- Surgical Innovations Unit, Westmead Hospital, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia; (A.N.); (M.W.)
| |
Collapse
|
30
|
Trovarelli G, Sbaraglia M, Angelini A, Bellan E, Pala E, Belluzzi E, Pozzuoli A, Borga C, Dei Tos AP, Ruggieri P. Are IDH1 R132 Mutations Associated With Poor Prognosis in Patients With Chondrosarcoma of the Bone? Clin Orthop Relat Res 2024; 482:00003086-990000000-01457. [PMID: 38170705 PMCID: PMC11124741 DOI: 10.1097/corr.0000000000002960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Because chondrosarcomas vary widely in their behavior, and because anticipating their behavior based on histology alone can be challenging, genetic markers represent an appealing area of inquiry that may help us refine our prognostic approaches. Isocitrate dehydrogenase (IDH) mutations are involved in the pathogenesis of a variety of neoplasms, and recently, IDH1/2 mutations have been found in the tissue of benign cartilage tumors as well as in conventional chondrosarcomas and highly aggressive dedifferentiated chondrosarcomas. However, their association with patient survival is still controversial. QUESTIONS/PURPOSES (1) What proportion of patients with chondrosarcomas carry IDH mutations, and which IDH mutations can be found? (2) Are any specific IDH mutations associated with poorer overall survival, metastasis-free survival, or local recurrence-free survival? METHODS Between April 2017 and December 2022, we treated 74 patients for atypical cartilaginous tumors or chondrosarcomas in a musculoskeletal tumor referral center. Patients were considered potentially eligible for the present study if the histologic diagnosis was confirmed by two expert soft tissue and bone pathologists following the current WHO classification, complete preoperative imaging and follow-up data were available, surgical excision was performed by sarcoma orthopaedic surgeons directed by a team leader, and the minimum follow-up was 2 years after surgical treatment unless the patient died. Data including sex, age, diagnosis, grade, type of operation, local recurrence, metastasis, and oncologic follow-up were recorded. Forty-one patients (55%) were eligible for the study. For each patient, DNA was extracted and quantified from paraffin-embedded sections of tumor tissue, and the mutational status of IDH1 (codons 105 and 132) and IDH2 (codons 140 and 172) genes was assessed. Of those, 56% (23 of 41) of patients had adequate DNA for analysis of IDH mutations: 10 male and 13 female patients, with a median age of 59 years (range 15 to 98 years). There were 22 conventional chondrosarcomas (8 atypical cartilaginous tumors, 11 Grade 2, and 3 Grade 3) and 1 dedifferentiated chondrosarcoma. Stage was IA in 3 patients, IB in 5, IIA in 1, IIB in 13, and III in 1, according to the Musculoskeletal Tumor Society classification. At a median follow-up of 3.5 years (range 4 months to 5.6 years), 14 patients were disease-free, 2 were alive with disease, and 7 died (3 within 2 years from surgery). Eight patients had metastases, and 7 developed local recurrence. We determined the proportion of patients who carried IDH mutations, and compared patients with and without those mutations in terms of overall survival, metastasis-free survival, and local recurrence-free survival using Kaplan-Meier curves. RESULTS Six patients showed wild-type IDH genes, and 17 had IDH mutations (12 had IDH1 R132, 3 had IDH1 G105, and 2 had IDH2 R172). Overall survival at 2 years using the Kaplan-Meier estimator was lower in patients with an IDH mutation than in those with the wild-type gene (75% [95% confidence interval 50% to 99%] versus 100% [95% CI 100% to 100%]; p = 0.002). Two-year metastasis-free survival was also lower in patients with an IDH mutation than in those with the wild-type gene (33% [95% CI 7% to 60%] versus 100% [95% CI 100% to 100%]; p = 0.001), as was 2-year local recurrence-free survival (70% [95% CI 42% to 98%] versus 100% [95% CI 100% to 100%]; p = 0.02). CONCLUSION We found that IDH1 R132 mutations were negatively associated with the prognosis of patients with bone chondrosarcomas. Nevertheless, more extensive studies (such as multicenter international studies) are needed and advisable to confirm our observations in this preliminary small series. Moreover, evaluating mutational status in fresh samples instead of in paraffin-embedded sections could help to increase the number of patients with adequate DNA for analysis. If our findings will be confirmed, the evaluation of IDH mutational status in biopsy samples or resection specimens could be considered when stratifying patients, highlighting those who may benefit from more aggressive treatment (such as adjuvant chemotherapy) or closer follow-up. LEVEL OF EVIDENCE Level III, prognostic study.
Collapse
Affiliation(s)
- Giulia Trovarelli
- Department of Orthopedics and Orthopedic Oncology, University of Padua, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology of University of Padova, Padua, Italy
| | - Marta Sbaraglia
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - Andrea Angelini
- Department of Orthopedics and Orthopedic Oncology, University of Padua, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology of University of Padova, Padua, Italy
| | - Elena Bellan
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - Elisa Pala
- Department of Orthopedics and Orthopedic Oncology, University of Padua, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology of University of Padova, Padua, Italy
| | - Elisa Belluzzi
- Department of Orthopedics and Orthopedic Oncology, University of Padua, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology of University of Padova, Padua, Italy
| | - Assunta Pozzuoli
- Department of Orthopedics and Orthopedic Oncology, University of Padua, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology of University of Padova, Padua, Italy
| | - Chiara Borga
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - Angelo Paolo Dei Tos
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - Pietro Ruggieri
- Department of Orthopedics and Orthopedic Oncology, University of Padua, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology of University of Padova, Padua, Italy
| |
Collapse
|
31
|
Zhang R, Tian Z, Xu Y, Lv L. D-mannose promotes the degradation of IDH2 through upregulation of RNF185 and suppresses breast cancer. Nutr Metab (Lond) 2024; 21:5. [PMID: 38167476 PMCID: PMC10763187 DOI: 10.1186/s12986-023-00774-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND D-mannose, an epimer of glucose, which is abundant in some fruits, such as cranberry, has been previously reported to inhibit urinary tract infection. In recent years, the potential function of D-mannose has been broadened into the regulation of other inflammation diseases and cancer. It was reported that D-mannose can increase reactive oxygen species (ROS) production, while IDH2 is important for the generation of NADPH, the crucial reducing factor. These findings prompted us to determine whether D-mannose can regulate IDH2 and IDH2-mediated NADPH production in tumor. METHODS The breast cancer cell line MDA-MB-231 was cultured and treated with 100mM D-mannose. IDH2 expression was detected by Western Blot and qRT-PCR. RNA-seq was conducted to identify the differentially expressed genes. BioGRID database was used to find the IDH2 interactors. Tumor cells were collected to measure the NADPH production using the NADP+/NADPH detection Kit. Colony formation assay and CCK-8 assay were conducted to evaluate the proliferation of cells. RESULTS D-mannose can promote IDH2 protein degradation through ubiquitination-proteasome pathway. Mechanistically, D-mannose treatment upregulated the expression of an E3 ligase - RNF185, which can interact with IDH2 and promotes its proteasomal degradation. Consequently, IDH2-mediated NADPH production was inhibited by D-mannose, the proliferation of breast cancer cells was retarded, and the sensitivity to pro-oxidant of breast cancer cells was elevated. CONCLUSIONS Our study demonstrated that D-mannose can degrade IDH2 and inhibit the production of NADPH to suppress the proliferation of breast cancer cells and render the breast cancer cells more sensitive to pro-oxidant treatment. Furthermore, we illustrated the E3 ligase RNF185 plays an important role in D-mannose-mediated proteasomal degradation of IDH2.
Collapse
Affiliation(s)
- Ruonan Zhang
- Nourse Centre for Pet Nutrition, 241200, Wuhu, China
| | - Ziyin Tian
- Nourse Centre for Pet Nutrition, 241200, Wuhu, China
| | - Yanping Xu
- Nourse Centre for Pet Nutrition, 241200, Wuhu, China.
| | - Lei Lv
- Nourse Centre for Pet Nutrition, 241200, Wuhu, China.
| |
Collapse
|
32
|
Zhang Y, Yan HJ, Wu J. The Tumor Immune Microenvironment plays a Key Role in Driving the Progression of Cholangiocarcinoma. Curr Cancer Drug Targets 2024; 24:681-700. [PMID: 38213139 DOI: 10.2174/0115680096267791231115101107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 01/13/2024]
Abstract
Cholangiocarcinoma (CCA) is an epithelial cancer distinguished by bile duct cell differentiation and is also a fibroproliferative tumor. It is characterized by a dense mesenchyme and a complex tumor immune microenvironment (TME). The TME comprises both cellular and non-cellular components. The celluar component includes CCA cells, immune cells and mesenchymal cells represented by the cancer-associated fibroblasts (CAFs), while the non-cellular component is represented by mesenchymal elements such as the extracellular matrix (ECM). Recent studies have demonstrated the important role of the TME in the development, progression, and treatment resistance of CCA. These cell-associated prognostic markers as well as intercellular connections, may serve as potential therapeutic targets and could inspire new treatment approaches for CCA in the future. This paper aims to summarize the current understanding of CCA's immune microenvironment, focusing on immune cells, mesenchymal cells, ECM, intercellular interactions, and metabolism within the microenvironment.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian St, Changzhou, 213003, China
| | - Hai-Jiao Yan
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian St, Changzhou, 213003, China
| | - Jun Wu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian St, Changzhou, 213003, China
| |
Collapse
|
33
|
Papadopoulou V, Schoumans J, Basset V, Solly F, Pasquier J, Blum S, Spertini O. Single-center, observational study of AML/MDS-EB with IDH1/2 mutations: genetic profile, immunophenotypes, mutational kinetics and outcomes. Hematology 2023; 28:2180704. [PMID: 36815747 DOI: 10.1080/16078454.2023.2180704] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
OBJECTIVE IDH1/2 mutations, intervening in epigenetic procedures, are frequently encountered in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). Knowledge of the genetics, immunophenotypes, and mutational kinetics of IDH1/2-mutated AML can contribute to the understanding of AML clonal architecture and inform therapeutics and monitoring. METHODS We retrospectively analyzed 50 IDH1/2-mutated AML/MDS-EB cases of our institution, to identify recurrent co-mutations, immunophenotypes, patterns of co-variance of IDH1/2 allele burdens with those of recurrent co-mutations, frequency of persistent IDH1/2 mutation as clonal hematopoiesis of indeterminate potential (CHIP) in remission and response to hypomethylating agents. RESULTS Most frequently co-mutated genes were DNMT3A, SRSF2 and NPM1. Most cases with co-existent IDH1/2 and NPM1 mutations (11/13) showed an 'APL-like' immunophenotype (CD34-HLADR-). Allele burdens of mutated IDH1/2 were identical to mutated SRSF2 allele burdens at diagnosis and remission, but not always to mutated NPM1 allele burden in remission. We show persistence of significant mutIDH1/2 allele burden in approximately one-fourth of patients with deep remissions. IDH1/2 mutations were significantly more frequent among responders to first-line HMA-based regimens than among non-responders, in patients treated for myeloid neoplasms with excess blasts. CONCLUSIONS IDH1/2 mutations are most frequently accompanied by DNMT3A, SRSF2 and NPM1 mutations. NPM1-IDH1/2 mutated AML has a mature phenotype possibly amenable to differentiation therapies. IDH1/2 and SRSF2 mutations probably arise at the same developmental stage of the disease, as their allele burdens covariate. IDH1/2 mutation represents CHIP in a substantial proportion of cases and is therefore no reliable residual disease marker. The preferential presence of IDH1/2 mutations among HMA-responders could inform therapeutic decisions if confirmed in larger series.
Collapse
Affiliation(s)
- Vasiliki Papadopoulou
- Service and Laboratory of Hematology, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Jacqueline Schoumans
- Service and Laboratory of Hematology, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Valentin Basset
- Service and Laboratory of Hematology, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Françoise Solly
- Service and Laboratory of Hematology, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Jérôme Pasquier
- Center for Primary Care and Public Health, University of Lausanne, Lausanne, Switzerland
| | - Sabine Blum
- Service and Laboratory of Hematology, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Olivier Spertini
- Service and Laboratory of Hematology, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
34
|
Malik N, Kundu A, Gupta Y, Irshad K, Arora M, Goswami S, Mahajan S, Sarkar C, Suri V, Suri A, Chattopadhyay P, Sinha S, Chosdol K. Protumorigenic role of the atypical cadherin FAT1 by the suppression of PDCD10 via RelA/miR221-3p/222-3p axis in glioblastoma. Mol Carcinog 2023; 62:1817-1831. [PMID: 37606187 DOI: 10.1002/mc.23617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 07/18/2023] [Accepted: 07/28/2023] [Indexed: 08/23/2023]
Abstract
The atypical cadherin FAT1 function either as a pro or antitumorigenic in tumors of different tissue origins. Our group previously demonstrated the protumorigenic nature of FAT1 signaling in glioblastoma (GBM). In this study, we investigated how FAT1 influences the expression of clustered oncomiRs (miR-221-3p/miR-222-3p) and their downstream effects in GBM. Through several experiments involving the measurement of specific gene/microRNA expression, gene knockdowns, protein and cellular assays, we have demonstrated a novel oncogenic signaling pathway mediated by FAT1 in glioma. These results have been verified using antimiRs and miR-mimic assays. Initially, in glioma-derived cell lines (U87MG and LN229), we observed FAT1 as a novel up-regulator of the transcription factor NFκB-RelA. RelA then promotes the expression of the clustered-oncomiRs, miR-221-3p/miR-222-3p, which in turn suppresses the expression of the tumor suppressor gene (TSG), PDCD10 (Programmed cell death protein10). The suppression of PDCD10, and other known TSG targets (PTEN/PUMA), by miR-221-3p/miR-222-3p, leads to increased clonogenicity, migration, and invasion of glioma cells. Consistent with our in-vitro findings, we observed a positive expression correlation of FAT1 and miR-221-3p, and an inverse correlation of FAT1 and the miR-targets (PDCD10/PTEN/PUMA), in GBM tissue-samples. These findings were also supported by publicly available GBM databases (The Cancer Genome Atlas [TCGA] and The Repository of Molecular Brain Neoplasia Data [Rembrandt]). Patients with tumors displaying high levels of FAT1 and miR-221-3p expression (50% and 65% respectively) experienced shorter overall survival. Similar results were observed in the TCGA-GBM database. Thus, our findings show a novel FAT1/RelA/miR-221/miR-222 oncogenic-effector pathway that downregulates the TSG, PDCD10, in GBM, which could be targeted therapeutically in a specific manner.
Collapse
Affiliation(s)
- Nargis Malik
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Archismita Kundu
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Yakhlesh Gupta
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Khushboo Irshad
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Manvi Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sanjeev Goswami
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Swati Mahajan
- Neuropathology Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Chitra Sarkar
- Neuropathology Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Vaishali Suri
- Neuropathology Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Ashish Suri
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | | | - Subrata Sinha
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Kunzang Chosdol
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
35
|
Wang X, Yuan L, Lu B, Lin D, Xu X. Glutathione promotes the synergistic effects of venetoclax and azacytidine against myelodysplastic syndrome‑refractory anemia by regulating the cell cycle. Exp Ther Med 2023; 26:574. [PMID: 38023359 PMCID: PMC10652243 DOI: 10.3892/etm.2023.12274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 09/27/2023] [Indexed: 12/01/2023] Open
Abstract
Azacitidine is a DNA methyltransferase inhibitor that has been used as a singular agent for the treatment of myelodysplastic syndrome-refractory anemia with excess blast-1 and -2 (MDS-RAEB I/II). However, recurrence and overall response rates following this treatment remain unsatisfactory. The combination of azacitidine and venetoclax has been used for the clinical treatment of a variety of hematological diseases due to the synergistic killing effect of the two drugs. Venetoclax is a BCL-2 inhibitor that can inhibit mitochondrial metabolism. In addition, azacitidine has been shown to reduce the levels of myeloid cell leukemia 1 (MCL-1) in acute myeloid leukemia cells. MCL-1 is an anti-apoptotic protein and a potential source of resistance to venetoclax. However, the mechanism underlying the effects of combined venetoclax and azacitidine treatment remains to be fully elucidated. In the present study, the molecular mechanism underlying the impact of venetoclax on the efficacy of azacitidine was investigated by examining its effects on cell cycle progression. SKM-1 cell lines were treated in vitro with 0-2 µM venetoclax and 0-4 µM azacytidine. After 24, 48 and 72 h of treatment, the impact of the drugs on the cell cycle was assessed by flow cytometry. Following drug treatment, changes in cellular glutamine metabolism pathways was analyzed using western blotting (ATF4, CHOP, ASCT2, IDH2 and RB), quantitative PCR (ASCT2 and IDH2), liquid chromatography-mass spectrometry (α-KG, succinate and glutathione) and ELISA (glutamine and glutaminase). Venetoclax was found to inhibit mitochondrial activity though the alanine-serine-cysteine transporter 2 (ASCT2) pathway, which decreased glutamine uptake. Furthermore, venetoclax partially antagonized the action of azacitidine through this ASCT2 pathway, which was reversed by glutathione (GSH) treatment. These results suggest that GSH treatment can potentiate the synergistic therapeutic effects of venetoclax and azacitidine combined treatment on a myelodysplastic syndrome-refractory anemia cell line at lower concentrations.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Lihua Yuan
- Department of Pediatric Surgery, University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong 518000, P.R. China
| | - Bo Lu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Dongjun Lin
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Xiaojun Xu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518107, P.R. China
| |
Collapse
|
36
|
Kolbinger FR, Bernard V, Lee JJ, Stephens BM, Branchi V, Raghav KPS, Maitra A, Guerrero PA, Semaan A. Significance of Distinct Liquid Biopsy Compartments in Evaluating Somatic Mutations for Targeted Therapy Selection in Cancer of Unknown Primary. J Gastrointest Cancer 2023; 54:1276-1285. [PMID: 36862364 DOI: 10.1007/s12029-023-00922-7] [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: 02/08/2023] [Indexed: 03/03/2023]
Abstract
PURPOSE Cancer of unknown primary (CUP) accounts for 2-5% of all cancer diagnoses, wherein standard investigations fail to reveal the original tumor site. Basket trials allocate targeted therapeutics based on actionable somatic mutations, independent of tumor entity. These trials, however, mostly rely on variants identified in tissue biopsies. Since liquid biopsies (LB) represent the overall tumor genomic landscape, they may provide an ideal diagnostic source in CUP patients. To identify the most informative liquid biopsy compartment, we compared the utility of genomic variant analysis for therapy stratification in two LB compartments (circulating cell-free (cf) and extracellular vesicle (ev) DNA). METHODS CfDNA and evDNA from 23 CUP patients were analyzed using a targeted gene panel covering 151 genes. Identified genetic variants were interpreted regarding diagnostic and therapeutic relevance using the MetaKB knowledgebase. RESULTS LB revealed a total of 22 somatic mutations in evDNA and/or cfDNA in 11/23 patients. Out of the 22 identified somatic variants, 14 are classified as Tier I druggable somatic variants. Comparison of variants identified in evDNA and cfDNA revealed an overlap of 58% of somatic variants in both LB compartments, whereas over 40% of variants were only found in one or the other compartment. CONCLUSION We observed substantial overlap between somatic variants identified in evDNA and cfDNA of CUP patients. Nonetheless, interrogation of both LB compartments can potentially increase the rate of druggable alterations, stressing the significance of liquid biopsies for possible primary-independent basket and umbrella trial inclusion.
Collapse
Affiliation(s)
- Fiona R Kolbinger
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vincent Bernard
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaewon J Lee
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bret M Stephens
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vittorio Branchi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kanwal P S Raghav
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paola A Guerrero
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Alexander Semaan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
37
|
Chen F, Dowerg B, Cordes T. The yin and yang of itaconate metabolism and its impact on the tumor microenvironment. Curr Opin Biotechnol 2023; 84:102996. [PMID: 37806082 DOI: 10.1016/j.copbio.2023.102996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/24/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023]
Abstract
The tumor microenvironment (TME) consists of a network of metabolically interconnected tumor and immune cell types. Macrophages influence the metabolic composition within the TME, which directly impacts the metabolic state and drug response of tumors. The accumulation of oncometabolites, such as succinate, fumarate, and 2-hydroxyglutarate, represents metabolic vulnerabilities in cancer that can be targeted therapeutically. Immunometabolites are emerging as metabolic regulators of the TME impacting immune cell functions and cancer cell growth. Here, we discuss recent discoveries on the potential impact of itaconate on the TME. We highlight how itaconate influences metabolic pathways relevant to immune responses and cancer cell proliferation. We also consider the therapeutic implications of manipulating itaconate metabolism as an immunotherapeutic strategy to constrain tumor growth.
Collapse
Affiliation(s)
- Fangfang Chen
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany; Research Group Cellular Metabolism in Infection, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Birte Dowerg
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Thekla Cordes
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany; Research Group Cellular Metabolism in Infection, Helmholtz Centre for Infection Research, Braunschweig, Germany.
| |
Collapse
|
38
|
Zhang R, Yang Y, Hu C, Huang M, Cen W, Ling D, Long Y, Yang XH, Xu B, Peng J, Wang S, Zhu W, Wei M, Yang J, Xu Y, Zhang X, Ma J, Wang F, Zhang H, Ma P, Zhu X, Song G, Sun LY, Wang DS, Wang FH, Li YH, Santagata S, Li Q, Feng YF, Du Z. Comprehensive analysis reveals potential therapeutic targets and an integrated risk stratification model for solitary fibrous tumors. Nat Commun 2023; 14:7479. [PMID: 37980418 PMCID: PMC10657378 DOI: 10.1038/s41467-023-43249-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023] Open
Abstract
Solitary fibrous tumors (SFTs) are rare mesenchymal tumors with unpredictable evolution and with a recurrence or metastasis rate of 10-40%. Current medical treatments for relapsed SFTs remain ineffective. Here, we identify potential therapeutic targets and risk factors, including IDH1 p.R132S, high PD-L1 expression, and predominant macrophage infiltration, suggesting the potential benefits of combinational immune therapy and targeted therapy for SFTs. An integrated risk model incorporating mitotic count, density of Ki-67+ cells and CD163+ cells, MTOR mutation is developed, applying a discovery cohort of 101 primary non-CNS patients with negative tumor margins (NTM) and validated in three independent cohorts of 210 SFTs with the same criteria, and in 36 primary CNS SFTs with NTM. Compared with the existing models, our model shows significantly improved efficacy in identifying high-risk primary non-CNS and CNS SFTs with NTM for tumor progression.Our findings hold promise for advancing therapeutic strategies and refining risk prediction in SFTs.
Collapse
Affiliation(s)
- Renjing Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yang Yang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chunfang Hu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Mayan Huang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wenjian Cen
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Dongyi Ling
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yakang Long
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xin-Hua Yang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Boheng Xu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Junling Peng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Sujie Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Weijie Zhu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Mingbiao Wei
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jiaojiao Yang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yuxia Xu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xu Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jiangjun Ma
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Hongtu Zhang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peiqing Ma
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiaojun Zhu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Guohui Song
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Li-Yue Sun
- Second Department of Oncology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - De-Shen Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Feng-Hua Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yu-Hong Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Sandro Santagata
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Qin Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Yan-Fen Feng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Ziming Du
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
39
|
Lin HH, Chang CY, Huang YR, Shen CH, Wu YC, Chang KL, Lee YC, Lin YC, Ting WC, Chien HJ, Zheng YF, Lai CC, Hsiao KY. Exon Junction Complex Mediates the Cap-Independent Translation of Circular RNA. Mol Cancer Res 2023; 21:1220-1233. [PMID: 37527157 DOI: 10.1158/1541-7786.mcr-22-0877] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/22/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023]
Abstract
Evidence that circular RNAs (circRNA) serve as protein template is accumulating. However, how the cap-independent translation is controlled remains largely uncharacterized. Here, we show that the presence of intron and thus splicing promote cap-independent translation. By acquiring the exon junction complex (EJC) after splicing, the interaction between circRNA and ribosomes was promoted, thereby facilitating translation. Prevention of splicing by treatment with spliceosome inhibitor or mutating splicing signal hindered cap-independent translation of circRNA. Moreover, EJC-tethering using Cas13 technology reconstituted EJC-dependent circRNA translation. Finally, the level of a coding circRNA from succinate dehydrogenase assembly factor 2 (circSDHAF2) was found to be elevated in the tumorous tissues from patients with colorectal cancer, and shown to be critical in tumorigenesis of colorectal cancer in both cell and murine models. These findings reveal that EJC-dependent control of circSDHAF2 translation is involved in the regulation of oncogenic pathways. IMPLICATIONS EJC-mediated cap-independent translation of circRNA is implicated in the tumorigenesis of colorectal cancer.
Collapse
Affiliation(s)
- Hui-Hsuan Lin
- Doctoral Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Biochemistry, College of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chiu-Yuan Chang
- Institute of Biochemistry, College of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Ren Huang
- Institute of Biochemistry, College of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Che-Hung Shen
- Doctoral Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Yu-Chen Wu
- Institute of Biochemistry, College of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Kai-Li Chang
- Department of Physiology, National Cheng Kung University, Tainan, Taiwan
| | - Yueh-Chun Lee
- Department of Radiation Oncology, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ya-Chi Lin
- Department of Plant Pathology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
| | - Wen-Chien Ting
- Division of Colorectal Surgery, Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Han-Ju Chien
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan
| | - Yi-Feng Zheng
- Institute of Molecular Biology, College of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chien-Chen Lai
- Institute of Molecular Biology, College of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Kuei-Yang Hsiao
- Doctoral Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Biochemistry, College of Life Sciences, National Chung Hsing University, Taichung, Taiwan
- Doctoral Program in Translational Medicine, College of Life Sciences, National Chung Hsing University, Taichung
- Rong Hsing Research Center for Translational Medicine, College of Life Sciences, National Chung Hsing University, Taichung
| |
Collapse
|
40
|
Murnan KM, Horbinski C, Stegh AH. Redox Homeostasis and Beyond: The Role of Wild-Type Isocitrate Dehydrogenases for the Pathogenesis of Glioblastoma. Antioxid Redox Signal 2023; 39:923-941. [PMID: 37132598 PMCID: PMC10654994 DOI: 10.1089/ars.2023.0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/06/2023] [Indexed: 05/04/2023]
Abstract
Significance: Glioblastoma is an aggressive and devastating brain tumor characterized by a dismal prognosis and resistance to therapeutic intervention. To support catabolic processes critical for unabated cellular growth and defend against harmful reactive oxygen species, glioblastoma tumors upregulate the expression of wild-type isocitrate dehydrogenases (IDHs). IDH enzymes catalyze the oxidative decarboxylation of isocitrate to α-ketoglutarate (α-KG), NAD(P)H, and CO2. On molecular levels, IDHs epigenetically control gene expression through effects on α-KG-dependent dioxygenases, maintain redox balance, and promote anaplerosis by providing cells with NADPH and precursor substrates for macromolecular synthesis. Recent Advances: While gain-of-function mutations in IDH1 and IDH2 represent one of the most comprehensively studied mechanisms of IDH pathogenic effects, recent studies identified wild-type IDHs as critical regulators of normal organ physiology and, when transcriptionally induced or down regulated, as contributing to glioblastoma progression. Critical Issues: Here, we will discuss molecular mechanisms of how wild-type IDHs control glioma pathogenesis, including the regulation of oxidative stress and de novo lipid biosynthesis, and provide an overview of current and future research directives that aim to fully characterize wild-type IDH-driven metabolic reprogramming and its contribution to the pathogenesis of glioblastoma. Future Directions: Future studies are required to further dissect mechanisms of metabolic and epigenomic reprogramming in tumors and the tumor microenvironment, and to develop pharmacological approaches to inhibit wild-type IDH function. Antioxid. Redox Signal. 39, 923-941.
Collapse
Affiliation(s)
- Kevin M. Murnan
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Malnati Brain Tumor Institute, Northwestern University, Chicago, Illinois, USA
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Alexander H. Stegh
- Department of Neurological Surgery, The Brain Tumor Center, Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
41
|
Dai Z, Zhu W, Hou Y, Zhang X, Ren X, Lei K, Liao J, Liu H, Chen Z, Peng S, Li S, Lin S, Kuang M. METTL5-mediated 18S rRNA m 6A modification promotes oncogenic mRNA translation and intrahepatic cholangiocarcinoma progression. Mol Ther 2023; 31:3225-3242. [PMID: 37735874 PMCID: PMC10638452 DOI: 10.1016/j.ymthe.2023.09.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/14/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a deadly cancer with rapid tumor progression. While hyperactive mRNA translation caused by mis-regulated mRNA or tRNA modifications promotes ICC development, the role of rRNA modifications remains elusive. Here, we found that 18S rRNA m6A modification and its methyltransferase METTL5 were aberrantly upregulated in ICC and associated with poorer survival (log rank test, p < 0.05). We further revealed the critical role of METTL5-mediated 18S rRNA m6A modification in regulation of ICC cell growth and metastasis using loss- and gain-of function assays in vitro and in vivo. The oncogenic function of METTL5 is corroborated using liver-specific knockout and overexpression ICC mouse models. Mechanistically, METTL5 depletion impairs 18S rRNA m6A modification that hampers ribosome synthesis and inhibits translation of G-quadruplex-containing mRNAs that are enriched in the transforming growth factor (TGF)-β pathway. Our study uncovers the important role of METTL5-mediated 18S rRNA m6A modification in ICC and unravels the mechanism of rRNA m6A modification-mediated oncogenic mRNA translation control.
Collapse
Affiliation(s)
- Zihao Dai
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Wanjie Zhu
- Department of Gastroenterology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong Province, China
| | - Yingdong Hou
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Xinyue Zhang
- Cancer Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Xuxin Ren
- Cancer Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Kai Lei
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Junbin Liao
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Haining Liu
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Zhihang Chen
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Sui Peng
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China; Clinical Trials Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Shaoqiang Li
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China.
| | - Shuibin Lin
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China.
| | - Ming Kuang
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
42
|
Alberghina L. The Warburg Effect Explained: Integration of Enhanced Glycolysis with Heterogeneous Mitochondria to Promote Cancer Cell Proliferation. Int J Mol Sci 2023; 24:15787. [PMID: 37958775 PMCID: PMC10648413 DOI: 10.3390/ijms242115787] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
The Warburg effect is the long-standing riddle of cancer biology. How does aerobic glycolysis, inefficient in producing ATP, confer a growth advantage to cancer cells? A new evaluation of a large set of literature findings covering the Warburg effect and its yeast counterpart, the Crabtree effect, led to an innovative working hypothesis presented here. It holds that enhanced glycolysis partially inactivates oxidative phosphorylation to induce functional rewiring of a set of TCA cycle enzymes to generate new non-canonical metabolic pathways that sustain faster growth rates. The hypothesis has been structured by constructing two metabolic maps, one for cancer metabolism and the other for the yeast Crabtree effect. New lines of investigation, suggested by these maps, are discussed as instrumental in leading toward a better understanding of cancer biology in order to allow the development of more efficient metabolism-targeted anticancer drugs.
Collapse
Affiliation(s)
- Lilia Alberghina
- Centre of Systems Biology, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| |
Collapse
|
43
|
Mohammad AH, Jatana S, Ruiz-Barerra MA, Khalaf R, Al-Saadi T, Diaz RJ. Metformin use is associated with longer survival in glioblastoma patients with MGMT gene silencing. J Neurooncol 2023; 165:209-218. [PMID: 37889443 DOI: 10.1007/s11060-023-04485-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
PURPOSE New treatments are needed to improve the overall survival of patients with glioblastoma Metformin is known for anti-tumorigenic effects in cancers, including breast and pancreas cancers. In this study, we assessed the association between metformin use and overall survival in glioblastoma patients. METHODS We retrospectively studied 241 patients who underwent surgery at diagnosis of glioblastoma between 2014 and 2018. Metformin was used for pre-existing type 2 diabetes mellitus or in the prevention or management of glucocorticoid induced hyperglycemia. Kaplan-Meier curves and log-rank p test were used for univariate analysis. Cox-proportional hazards model was used to generate adjusted hazard ratios for multivariate analysis. RESULTS Metformin use was associated with longer survival in patients with tumors that had a methylated O6-methylguanine DNA methyltransferase gene (MGMT) promoter (484 days 95% CI: 56-911 vs. 394 days 95% CI: 249-538, Log-Rank test: 6.5, p = 0.01). Cox regression analysis shows that metformin associates with lower risk of death at 2 years in patients with glioblastoma containing a methylated MGMT promoter (aHR = 0.497, 95% CI 0.26-0.93, p = 0.028). CONCLUSION Our findings suggest a survival benefit with metformin use in patients with glioblastomas having methylation of the MGMT promoter.
Collapse
Affiliation(s)
| | | | - Miguel Angel Ruiz-Barerra
- Neuro-Oncology Research Group, National Institute of Cancer, Bogotá, Colombia
- Department of Neurosurgery, National Institute of Cancer, Bogotá, Colombia
| | - Roy Khalaf
- Faculty of Medicine, McGill University, Montreal, Canada
| | - Tariq Al-Saadi
- Faculty of Medicine, McGill University, Montreal, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute - McGill University Health Centre, Montreal, Canada
| | - Roberto J Diaz
- Faculty of Medicine, McGill University, Montreal, Canada.
- Department of Neurology and Neurosurgery, Montreal Neurological Institute - McGill University Health Centre, Montreal, Canada.
- Neurosurgical Oncology, Department of Neurology and Neurosurgery, Montreal Neurological Hospital - McGill University Health Centre, Montreal, Canada.
- Neurosurgical Oncology, Department of Neurology and Neurosurgery, Montreal Neurological Hospital, Faculty of Medicine, Faculty of Medicine, McGill University, 3801 University Street, Montreal, QC, H3A 2B4, Canada.
| |
Collapse
|
44
|
Rocca C, Soda T, De Francesco EM, Fiorillo M, Moccia F, Viglietto G, Angelone T, Amodio N. Mitochondrial dysfunction at the crossroad of cardiovascular diseases and cancer. J Transl Med 2023; 21:635. [PMID: 37726810 PMCID: PMC10507834 DOI: 10.1186/s12967-023-04498-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 09/01/2023] [Indexed: 09/21/2023] Open
Abstract
A large body of evidence indicates the existence of a complex pathophysiological relationship between cardiovascular diseases and cancer. Mitochondria are crucial organelles whose optimal activity is determined by quality control systems, which regulate critical cellular events, ranging from intermediary metabolism and calcium signaling to mitochondrial dynamics, cell death and mitophagy. Emerging data indicate that impaired mitochondrial quality control drives myocardial dysfunction occurring in several heart diseases, including cardiac hypertrophy, myocardial infarction, ischaemia/reperfusion damage and metabolic cardiomyopathies. On the other hand, diverse human cancers also dysregulate mitochondrial quality control to promote their initiation and progression, suggesting that modulating mitochondrial homeostasis may represent a promising therapeutic strategy both in cardiology and oncology. In this review, first we briefly introduce the physiological mechanisms underlying the mitochondrial quality control system, and then summarize the current understanding about the impact of dysregulated mitochondrial functions in cardiovascular diseases and cancer. We also discuss key mitochondrial mechanisms underlying the increased risk of cardiovascular complications secondary to the main current anticancer strategies, highlighting the potential of strategies aimed at alleviating mitochondrial impairment-related cardiac dysfunction and tumorigenesis. It is hoped that this summary can provide novel insights into precision medicine approaches to reduce cardiovascular and cancer morbidities and mortalities.
Collapse
Affiliation(s)
- Carmine Rocca
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E and E.S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy
| | - Teresa Soda
- Department of Health Science, University Magna Graecia of Catanzaro, 88100, Catanzaro, Italy
| | - Ernestina Marianna De Francesco
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122, Catania, Italy
| | - Marco Fiorillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E and E.S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy.
- National Institute of Cardiovascular Research (I.N.R.C.), 40126, Bologna, Italy.
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy.
| |
Collapse
|
45
|
Bhuvanadas S, Devi A. JARID2 and EZH2, The Eminent Epigenetic Drivers In Human Cancer. Gene 2023:147584. [PMID: 37353042 DOI: 10.1016/j.gene.2023.147584] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 06/09/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Cancer has become a prominent cause of death, accounting for approximately 10 million death worldwide as per the World Health Organization reports 2020. Epigenetics deal with the alterations of heritable phenotypes, except for DNA alterations. Currently, we are trying to comprehend the role of utmost significant epigenetic genes involved in the burgeoning of human cancer. A sundry of studies reported the Enhancer of Zeste Homologue2 (EZH2) as a prime catalytic subunit of Polycomb Repressive Complex2, which is involved in several pivotal activities, including embryogenesis. In addition, EZH2 has detrimental effects leading to the onset and metastasis of several cancers. Jumonji AT Rich Interacting Domain2 (JARID2), an undebated crucial nuclear factor, has strong coordination with the PRC2 family. In this review, we discuss various epigenetic entities, primarily focusing on the possible role and mechanism of EZH2 and the significant contribution of JARID2 in human cancers.
Collapse
Affiliation(s)
- Sreeshma Bhuvanadas
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India - 603203
| | - Arikketh Devi
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India - 603203.
| |
Collapse
|
46
|
Zong Y, Wang X, Cui B, Xiong X, Wu A, Lin C, Zhang Y. Decoding the regulatory roles of non-coding RNAs in cellular metabolism and disease. Mol Ther 2023; 31:1562-1576. [PMID: 37113055 PMCID: PMC10277898 DOI: 10.1016/j.ymthe.2023.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 04/29/2023] Open
Abstract
Non-coding RNAs, including long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), are being studied extensively in a variety of fields. Their roles in metabolism have received increasing attention in recent years but are not yet clear. The regulation of glucose, fatty acid, and amino acid metabolism is an imperative physiological process that occurs in living organisms and takes part in cancer and cardiovascular diseases. Here, we summarize the important roles played by non-coding RNAs in glucose metabolism, fatty acid metabolism, and amino acid metabolism, as well as the mechanisms involved. We also summarize the therapeutic advances for non-coding RNAs in diseases such as obesity, cardiovascular disease, and some metabolic diseases. Overall, non-coding RNAs are indispensable factors in metabolism and have a significant role in the three major metabolisms, which may be exploited as therapeutic targets in the future.
Collapse
Affiliation(s)
- Yuru Zong
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xuliang Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Bing Cui
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xiaowei Xiong
- Department of Cardiology and Macrovascular Disease, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Andrew Wu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Yaohua Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
47
|
Zhang L, Fritah S, Nazarov PV, Kaoma T, Van Dyck E. Impact of IDH Mutations, the 1p/19q Co-Deletion and the G-CIMP Status on Alternative Splicing in Diffuse Gliomas. Int J Mol Sci 2023; 24:9825. [PMID: 37372972 DOI: 10.3390/ijms24129825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
By generating protein diversity, alternative splicing provides an important oncogenic pathway. Isocitrate dehydrogenase (IDH) 1 and 2 mutations and 1p/19q co-deletion have become crucial for the novel molecular classification of diffuse gliomas, which also incorporates DNA methylation profiling. In this study, we have carried out a bioinformatics analysis to examine the impact of the IDH mutation, as well as the 1p/19q co-deletion and the glioma CpG island methylator phenotype (G-CIMP) status on alternative splicing in a cohort of 662 diffuse gliomas from The Cancer Genome Atlas (TCGA). We identify the biological processes and molecular functions affected by alternative splicing in the various glioma subgroups and provide evidence supporting the important contribution of alternative splicing in modulating epigenetic regulation in diffuse gliomas. Targeting the genes and pathways affected by alternative splicing might provide novel therapeutic opportunities against gliomas.
Collapse
Affiliation(s)
- Lu Zhang
- Bioinformatics Platform, Data Integration and Analysis Unit (DIA), Luxembourg Institute of Health (LIH), L-1445 Strassen, Luxembourg
| | - Sabrina Fritah
- NorLux Neuro-Oncology Laboratory, Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), L-1445 Strassen, Luxembourg
| | - Petr V Nazarov
- Bioinformatics Platform, Data Integration and Analysis Unit (DIA), Luxembourg Institute of Health (LIH), L-1445 Strassen, Luxembourg
- Multiomics Data Science Research Group, DoCR, Luxembourg Institute of Health (LIH), L-1445 Strassen, Luxembourg
| | - Tony Kaoma
- Bioinformatics Platform, Data Integration and Analysis Unit (DIA), Luxembourg Institute of Health (LIH), L-1445 Strassen, Luxembourg
| | - Eric Van Dyck
- DNA Repair and Chemoresistance Group, DoCR, Luxembourg Institute of Health (LIH), L-1445 Strassen, Luxembourg
| |
Collapse
|
48
|
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
|
49
|
Ekhator C, Rak R. Surgical Management of Multiple-Level Lumbar Spinal Schwannomas: A Case Report. Cureus 2023; 15:e41113. [PMID: 37397655 PMCID: PMC10308063 DOI: 10.7759/cureus.41113] [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] [Accepted: 06/28/2023] [Indexed: 07/04/2023] Open
Abstract
The increase in benign spinal tumors among adults over the last decade has been a major cause of concern. This worrisome trend has been attributed to many factors, including improved detection techniques, enhanced access to medical care, and the aging population. The research primarily focuses on Schwannoma, which is a rare type of tumor that arises from Schwann cells, which are responsible for producing the myelin sheath that surrounds and protects nerves. Although the majority of schwannomas are benign, there have been instances where they have transformed into malignant tumors, potentially leading to significant morbidity and mortality. We report a case of a 68-year-old woman who presented with progressive back pain and weakness in both lower limbs over the past months. The pain was initially localized to the lower back but gradually intensified and radiated down to the legs. The patient reported difficulty walking and a sensation of tingling and numbness in the feet. She denied any recent trauma or significant medical history. On physical examination, there was reduced muscle strength (3/5) in both lower limbs. The patient exhibited hyporeflexia in the knees and ankle. A magnetic resonance imaging (MRI) of the spine was performed, revealing a well-defined mass lesion located in the lumbar region, compressing the spinal cord from L2 to L5. The patient was counseled and prepared for surgical resection of the tumor. Histopathological findings revealed features of peripheral nerve sheath tumors and cellular schwannomas. The patient recovered well postoperatively. The surgeon operating should be mindful of the potential presence of a mobile schwannoma, even though it is rarely mentioned in the literature. Being aware of this possibility can help prevent unnecessary surgical dissection, which can lead to higher rates of complications and morbidity. Although it is plausible that this case could have involved a mobile schwannoma, there was not enough evidence to support it as we performed a laminectomy on multiple levels due to the tumor's size.
Collapse
Affiliation(s)
- Chukwuyem Ekhator
- Neuro-Oncology, College of Osteopathic Medicine, New York Institute of Technology, Long Island, USA
| | - Ramin Rak
- Neurosurgery, Neurosurgical PC (NSPC) Brain and Spine Surgery, Rockville Center, USA
| |
Collapse
|
50
|
Chen J, Hong JH, Huang Y, Liu S, Yin J, Deng P, Sun Y, Yu Z, Zeng X, Xiao R, Chan JY, Guan P, Wang Y, Wang P, Liu L, Wen S, Yu Q, Ong CK, Teh BT, Xiong Y, Tan J. EZH2 mediated metabolic rewiring promotes tumor growth independently of histone methyltransferase activity in ovarian cancer. Mol Cancer 2023; 22:85. [PMID: 37210576 DOI: 10.1186/s12943-023-01786-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 05/09/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Enhancer of zeste homolog 2 (EZH2), the key catalytic subunit of polycomb repressive complex 2 (PRC2), is overexpressed and plays an oncogenic role in various cancers through catalysis-dependent or catalysis-independent pathways. However, the related mechanisms contributing to ovarian cancer (OC) are not well understood. METHODS The levels of EZH2 and H3K27me3 were evaluated in 105 OC patients by immunohistochemistry (IHC) staining, and these patients were stratified based on these levels. Canonical and noncanonical binding sites of EZH2 were defined by chromatin immunoprecipitation sequencing (ChIP-Seq). The EZH2 solo targets were obtained by integrative analysis of ChIP-Seq and RNA sequencing data. In vitro and in vivo experiments were performed to determine the role of EZH2 in OC growth. RESULTS We showed that a subgroup of OC patients with high EZH2 expression but low H3K27me3 exhibited the worst prognosis, with limited therapeutic options. We demonstrated that induction of EZH2 degradation but not catalytic inhibition profoundly blocked OC cell proliferation and tumorigenicity in vitro and in vivo. Integrative analysis of genome-wide chromatin and transcriptome profiles revealed extensive EZH2 occupancy not only at genomic loci marked by H3K27me3 but also at promoters independent of PRC2, indicating a noncanonical role of EZH2 in OC. Mechanistically, EZH2 transcriptionally upregulated IDH2 to potentiate metabolic rewiring by enhancing tricarboxylic acid cycle (TCA cycle) activity, which contributed to the growth of OC. CONCLUSIONS These data reveal a novel oncogenic role of EZH2 in OC and identify potential therapeutic strategies for OC by targeting the noncatalytic activity of EZH2.
Collapse
Affiliation(s)
- Jianfeng Chen
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou, Guangdong, 510060, P. R. China.
| | - Jing Han Hong
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Yulin Huang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Shini Liu
- Department of Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510060, P. R. China
| | - Jiaxin Yin
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Peng Deng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yichen Sun
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangdong, 510060, P. R. China
| | - Zhaoliang Yu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, P. R. China
| | - Xian Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Rong Xiao
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Jason Yongsheng Chan
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Peiyong Guan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
- Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Yali Wang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Peili Wang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Lizhen Liu
- Center of Medical Research, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510060, P. R. China
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Qiang Yu
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
- Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Choon Kiat Ong
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
- Genome Institute of Singapore, A*STAR, Singapore, Singapore
- Lymphoma Genomic Translational Research Laboratory, Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
| | - Bin-Tean Teh
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
- Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Ying Xiong
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Jing Tan
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou, Guangdong, 510060, P. R. China.
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangdong, 510060, P. R. China.
- Center of Medical Research, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510060, P. R. China.
| |
Collapse
|