151
|
Game of clones: Diverse implications for clonal hematopoiesis in lymphoma and multiple myeloma. Blood Rev 2022; 56:100986. [PMID: 35753868 DOI: 10.1016/j.blre.2022.100986] [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: 04/13/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 11/23/2022]
Abstract
Clonal hematopoiesis (CH) refers to the disproportionate expansion of hematopoietic stem cell clones and their corresponding progeny following the acquisition of somatic mutations. CH is common at the time of diagnosis in patients with blood cancers, including multiple myeloma (MM) and lymphoma. The presence of CH mutations correlates with IL-6 mediated inflammation and may result in lymphoma or MM modulation through microenvironment effects or by manifestations of the mutations themselves within the founding tumor clone. As might be expected with a variety of mutations and multiple potential mechanisms, CH exerts context-dependent effects, being protective in some settings and harmful in others. Though CH is very common in patients with hematologic malignancies, how it intersects with therapy and the natural disease course of these cancers are active areas of investigation. In lymphomas and MM specifically, patients have high rates of CH at diagnosis and are subsequently exposed to therapies, such as cytotoxic chemotherapy, that can cause CH progression to overt hematologic malignancy. The expanding diversity of treatment modalities for these cancers also increases the opportunities for CH to impact clinical outcome and modulate clinical responses. Here we review the basic biology and known health effects of CH, and we focus on the clinical relevance of CH in lymphoma and MM.
Collapse
|
152
|
Messina M, Piciocchi A, Ottone T, Paolini S, Papayannidis C, Lessi F, Fracchiolla NS, Forghieri F, Candoni A, Mengarelli A, Martelli MP, Venditti A, Carella AM, Albano F, Mancini V, Massimo B, Arena V, Sargentini V, Sciumè M, Pastore D, Todisco E, Roti G, Siragusa S, Ladetto M, Pravato S, De Bellis E, Simonetti G, Marconi G, Cerchione C, Fazi P, Vignetti M, Amadori S, Martinelli G, Voso MT. Prevalence and Prognostic Role of IDH Mutations in Acute Myeloid Leukemia: Results of the GIMEMA AML1516 Protocol. Cancers (Basel) 2022; 14:cancers14123012. [PMID: 35740677 PMCID: PMC9221405 DOI: 10.3390/cancers14123012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/08/2022] [Accepted: 06/15/2022] [Indexed: 02/04/2023] Open
Abstract
IDH1/2 mutations are common in acute myeloid leukemia (AML) and represent a therapeutic target. The GIMEMA AML1516 observational protocol was designed to study the prevalence of IDH1/2 mutations and associations with clinico-biological parameters in a cohort of Italian AML patients. We analyzed a cohort of 284 AML consecutive patients at diagnosis, 139 females and 145 males, of a median age of 65 years (range: 19−86). Of these, 38 (14%) harbored IDH1 and 51 (18%) IDH2 mutations. IDH1/2 mutations were significantly associated with WHO PS >2 (p < 0.001) and non-complex karyotype (p = 0.021) when compared to IDH1/2-WT. Furthermore, patients with IDH1 mutations were more frequently NPM1-mutated (p = 0.007) and had a higher platelet count (p = 0.036). At relapse, IDH1/2 mutations were detected in 6 (25%) patients. As per the outcome, 60.5% of IDH1/2-mutated patients achieved complete remission; overall survival and event-free survival at 2 years were 44.5% and 36.1%, respectively: these rates were similar to IDH1/2-WT. In IDH1/2-mutated patients, high WBC proved to be an independent prognostic factor for survival. In conclusion, the GIMEMA AML1516 confirms that IDH1/2 mutations are frequently detected at diagnosis and underlines the importance of recognizing IDH1/2-mutated cases up-front to offer the most appropriate therapeutic strategy, given the availability of IDH1/2 inhibitors.
Collapse
Affiliation(s)
- Monica Messina
- GIMEMA Foundation, 00182 Roma, Italy; (M.M.); (A.P.); (V.A.); (V.S.); (P.F.); (M.V.)
| | - Alfonso Piciocchi
- GIMEMA Foundation, 00182 Roma, Italy; (M.M.); (A.P.); (V.A.); (V.S.); (P.F.); (M.V.)
| | - Tiziana Ottone
- Ematologia, Dipartimento di Biomedicina e Prevenzione, Università di Roma Tor Vergata, 00133 Roma, Italy; (T.O.); (A.V.); (S.A.)
- Neuro-Oncohematology Unit, IRCCS Fondazione Santa Lucia, 00179 Roma, Italy
| | - Stefania Paolini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna Istituto di Ematologia “Seràgnoli” Bologna, 40138 Bologna, Italy; (S.P.); (C.P.)
| | - Cristina Papayannidis
- IRCCS Azienda Ospedaliero-Universitaria di Bologna Istituto di Ematologia “Seràgnoli” Bologna, 40138 Bologna, Italy; (S.P.); (C.P.)
| | - Federica Lessi
- Ematologia ed Immunologia Clinica, Università degli Studi di Padova, 1222 Padua, Italy; (F.L.); (S.P.)
| | - Nicola Stefano Fracchiolla
- UOC Ematologia, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (N.S.F.); (M.S.)
| | - Fabio Forghieri
- UO Ematologia-AOU Policlinico di Modena, 41125 Modena, Italy;
| | - Anna Candoni
- Clinica Ematologica, ASUFC, Università degli Studi di Udine, 33100 Udine, Italy;
| | - Andrea Mengarelli
- UO Ematologia-IRCCS Istituto Nazionale Tumori Tumori Regina Elena, 00128 Roma, Italy;
| | - Maria Paola Martelli
- Sezione di Ematologia ed Immunologia Clinica, Università degli Studi di Perugia, 06123 Perugia, Italy;
| | - Adriano Venditti
- Ematologia, Dipartimento di Biomedicina e Prevenzione, Università di Roma Tor Vergata, 00133 Roma, Italy; (T.O.); (A.V.); (S.A.)
| | - Angelo Michele Carella
- Ematologia e Centro Trapianti CSE Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Francesco Albano
- Hematology and Stem Cell Transplantation Unit, Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari Aldo Moro, 70121 Bari, Italy;
| | - Valentina Mancini
- Ospedale Niguarda Ca Granda-SC Ematologia Blocco SUD, 20162 Milano, Italy;
| | | | - Valentina Arena
- GIMEMA Foundation, 00182 Roma, Italy; (M.M.); (A.P.); (V.A.); (V.S.); (P.F.); (M.V.)
| | - Valeria Sargentini
- GIMEMA Foundation, 00182 Roma, Italy; (M.M.); (A.P.); (V.A.); (V.S.); (P.F.); (M.V.)
| | - Mariarita Sciumè
- UOC Ematologia, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (N.S.F.); (M.S.)
| | | | - Elisabetta Todisco
- Onco-Hematology Division, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy;
| | - Giovanni Roti
- Azienda Ospedaliera Universitaria di Parma, Ematologia, Università di Parma, 43126 Parma, Italy;
| | - Sergio Siragusa
- U.O. di Ematologia con Trapianto-A.U. Policlinico Paolo Giaccone, 90127 Palermo, Italy;
| | - Marco Ladetto
- AO SS Antonio e Biagio Arrigo, 15121 Alessandria, Italy;
| | - Stefano Pravato
- Ematologia ed Immunologia Clinica, Università degli Studi di Padova, 1222 Padua, Italy; (F.L.); (S.P.)
| | - Eleonora De Bellis
- Hematology Unit, Azienda Sanitaria Universitaria Giuliano Isontina, 34148 Trieste, Italy;
| | - Giorgia Simonetti
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) Dino Amadori, 47014 Meldola, Italy; (G.S.); (G.M.)
| | - Giovanni Marconi
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori [M1] (IRST) IRCCS, 47014 Meldola, Italy; (G.M.); (C.C.)
| | - Claudio Cerchione
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori [M1] (IRST) IRCCS, 47014 Meldola, Italy; (G.M.); (C.C.)
| | - Paola Fazi
- GIMEMA Foundation, 00182 Roma, Italy; (M.M.); (A.P.); (V.A.); (V.S.); (P.F.); (M.V.)
| | - Marco Vignetti
- GIMEMA Foundation, 00182 Roma, Italy; (M.M.); (A.P.); (V.A.); (V.S.); (P.F.); (M.V.)
| | - Sergio Amadori
- Ematologia, Dipartimento di Biomedicina e Prevenzione, Università di Roma Tor Vergata, 00133 Roma, Italy; (T.O.); (A.V.); (S.A.)
| | - Giovanni Martinelli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) Dino Amadori, 47014 Meldola, Italy; (G.S.); (G.M.)
| | - Maria Teresa Voso
- Ematologia, Dipartimento di Biomedicina e Prevenzione, Università di Roma Tor Vergata, 00133 Roma, Italy; (T.O.); (A.V.); (S.A.)
- Neuro-Oncohematology Unit, IRCCS Fondazione Santa Lucia, 00179 Roma, Italy
- Correspondence:
| |
Collapse
|
153
|
Arterial Spin Labeling Perfusion in Determining the IDH1 Status and Ki-67 Index in Brain Gliomas. Diagnostics (Basel) 2022; 12:diagnostics12061444. [PMID: 35741254 PMCID: PMC9221904 DOI: 10.3390/diagnostics12061444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/30/2022] [Accepted: 06/10/2022] [Indexed: 11/22/2022] Open
Abstract
The aim of the study was to evaluate the relationship between tumor blood flow (TBF) measured by the pseudo-continuous arterial spin labeling (PCASL) method and IDH1 mutation status of gliomas as well as Ki-67 proliferative index. Methods. The study included 116 patients with newly diagnosed gliomas of various grades. They received no chemotherapy or radiotherapy before MRI. IDH1 status assessment was performed after tumor removal in 106 cases—48 patients were diagnosed with wildtype gliomas (Grade 1−2—6 patients, Grade 3−4—42 patients) and 58 patients were diagnosed with mutant forms of gliomas (Grade 1−2—28 patients, Grade 3−4—30 patients). In 64 cases out of 116 Ki-67 index was measured. Absolute and normalized tumor blood flow values were measured on 3D PCASL maps. Results. TBF and normalized TBF (nTBF) in wildtype gliomas were significantly higher than in IDH1-mutant gliomas (p < 0.001). ASL perfusion showed high values of sensitivity and specificity in the differential diagnosis of gliomas with distinct IDH1 status (for TBF: specificity 75%, sensitivity 77.6%, AUC 0.783, cutoff 80.57 mL/100 g/min, for nTBF: specificity 77.1%, sensitivity 79.3%, AUC 0.791, cutoff 4.7). TBF and nTBF in wildtype high-grade gliomas (HGG) were significantly higher than in mutant forms (p < 0.001). ASL perfusion showed the following values of sensitivity and specificity in the diagnosis of mutant HGG and wildtype HGG (for TBF: specificity 83.3%, sensitivity 60%, AUC 0.719, cutoff 84.18 mL/100 g/min, for nTBF: specificity 88.1%, sensitivity 60%, AUC 0.729, cutoff 4.7). There was a significant positive correlation between tumor blood flow and Ki-67 (for TBF Rs = 0.63, for nTBF Rs = 0.61). Conclusion. ASL perfusion may be an informative factor in determining the IDH1 status in brain gliomas preoperative and tumor proliferative activity.
Collapse
|
154
|
Raslan O, Garcia-Horton A. Azacitidine and its role in the upfront treatment of acute myeloid leukemia. Expert Opin Pharmacother 2022; 23:873-884. [PMID: 35695017 DOI: 10.1080/14656566.2022.2082284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) predominantly affects elderly population. This poses challenges in management, as patients are frequently not candidates for intensive therapy given comorbidities or frailty. Currently, azacitidine (AZA), either as monotherapy or in combination regimens, is the backbone treatment in this group of patients. AREAS COVERED We review the mechanism of action, pharmacology, clinical efficacy, and safety of AZA. It reviews current combination therapies of AZA with other targeted therapies for the treatment of newly diagnosed AML. EXPERT OPINION AZA is a cornerstone for the treatment of patients considered ineligible for intensive chemotherapy induction, but better results and therapies are required for these patients. AZA has shown synergistic properties when combined with other medications. Its safety profile and few drug interactions make it a suitable medication to use as backbone. Newer therapies are being combined with AZA, demonstrating safety and in cases, improved responses, and survival. AZA/venetoclax has emerged as the standard of care for patients who are ineligible for intensive chemotherapy. Doublet and triplet combinations are increasingly being studied. With the results observed in elderly patients, the intensive chemotherapy paradigm might be put to test in younger populations, with AZA combinations being at the forefront.
Collapse
Affiliation(s)
- Omar Raslan
- Department of Oncology, Juravinski Cancer Centre - Hamilton Health Sciences, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, Division of Hematology, University of Jeddah, College of Medicine, Jeddah, Saudi Arabia
| | - Alejandro Garcia-Horton
- Department of Oncology, Juravinski Cancer Centre - Hamilton Health Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
155
|
Cho SM, Esmail A, Raza A, Dacha S, Abdelrahim M. Timeline of FDA-Approved Targeted Therapy for Cholangiocarcinoma. Cancers (Basel) 2022; 14:2641. [PMID: 35681621 PMCID: PMC9179455 DOI: 10.3390/cancers14112641] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 12/14/2022] Open
Abstract
Cholangiocarcinoma (CCA) represents approximately 3% of gastrointestinal malignancies worldwide and constitutes around 10-15% of all primary liver cancers, being only second to hepatocellular carcinoma. Mortality from CCA has been on the rise in recent decades, and in the United States alone there has been a 36% increase in CCA from 1999 to 2014, with over 7000 CCA mortalities since 2013. Targeted therapies, which have been gaining interest due to their greater specificity toward cancer cells, have only recently started gaining FDA approval for the treatment of CCA. In this manuscript, we will go through the timeline of current FDA-approved targeted therapies as well as those that have gained FDA breakthrough therapy designation.
Collapse
Affiliation(s)
- Su Min Cho
- Department of Medicine, Houston Methodist Hospital, Houston, TX 77030, USA;
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
- Department of Medicine, Texas A&M College of Medicine, Bryan, TX 77807, USA
| | - Abdullah Esmail
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX 77030, USA;
| | - Ali Raza
- Department of Gastroenterology, Houston Methodist Hospital, Houston, TX 77030, USA; (A.R.); (S.D.)
| | - Sunil Dacha
- Department of Gastroenterology, Houston Methodist Hospital, Houston, TX 77030, USA; (A.R.); (S.D.)
| | - Maen Abdelrahim
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX 77030, USA;
- Cockrell Center of Advanced Therapeutics Phase I Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
156
|
Kubik J, Humeniuk E, Adamczuk G, Madej-Czerwonka B, Korga-Plewko A. Targeting Energy Metabolism in Cancer Treatment. Int J Mol Sci 2022; 23:ijms23105572. [PMID: 35628385 PMCID: PMC9146201 DOI: 10.3390/ijms23105572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer is the second most common cause of death worldwide after cardiovascular diseases. The development of molecular and biochemical techniques has expanded the knowledge of changes occurring in specific metabolic pathways of cancer cells. Increased aerobic glycolysis, the promotion of anaplerotic responses, and especially the dependence of cells on glutamine and fatty acid metabolism have become subjects of study. Despite many cancer treatment strategies, many patients with neoplastic diseases cannot be completely cured due to the development of resistance in cancer cells to currently used therapeutic approaches. It is now becoming a priority to develop new treatment strategies that are highly effective and have few side effects. In this review, we present the current knowledge of the enzymes involved in the different steps of glycolysis, the Krebs cycle, and the pentose phosphate pathway, and possible targeted therapies. The review also focuses on presenting the differences between cancer cells and normal cells in terms of metabolic phenotype. Knowledge of cancer cell metabolism is constantly evolving, and further research is needed to develop new strategies for anti-cancer therapies.
Collapse
Affiliation(s)
- Joanna Kubik
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (J.K.); (G.A.); (A.K.-P.)
| | - Ewelina Humeniuk
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (J.K.); (G.A.); (A.K.-P.)
- Correspondence: ; Tel.: +48-81-448-65-20
| | - Grzegorz Adamczuk
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (J.K.); (G.A.); (A.K.-P.)
| | - Barbara Madej-Czerwonka
- Human Anatomy Department, Faculty of Medicine, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Agnieszka Korga-Plewko
- Independent Medical Biology Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland; (J.K.); (G.A.); (A.K.-P.)
| |
Collapse
|
157
|
Chen S, Zhu H, Jin M, Yuan H, Liu Z, Li J, Zhang X, Meng L, Li T, Diao Y, Gao H, Hong C, Zhu X, Zheng J, Li F, Niu Y, Ma T, Li X. Molecular and clinical characteristics of
IDH
mutations in Chinese
NSCLC
patients and potential treatment strategies. Cancer Med 2022; 11:4122-4133. [PMID: 35526267 DOI: 10.1002/cam4.4764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/30/2022] [Accepted: 04/09/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Shuchen Chen
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| | - Honglin Zhu
- Hangzhou Jichenjunchuang Medical Laboratory, Co., Ltd. Hangzhou China
| | - Meizi Jin
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| | - Hongling Yuan
- Hangzhou Jichenjunchuang Medical Laboratory, Co., Ltd. Hangzhou China
| | - Zhenzhen Liu
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| | - Jielin Li
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| | - Xiang Zhang
- Hangzhou Jichenjunchuang Medical Laboratory, Co., Ltd. Hangzhou China
| | - Lihui Meng
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| | - Ting Li
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| | - Yuzhu Diao
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| | - Hong Gao
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| | - Chengyu Hong
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| | - Xinjiang Zhu
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| | - Jian Zheng
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| | - Fei Li
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| | - Yanling Niu
- Hangzhou Jichenjunchuang Medical Laboratory, Co., Ltd. Hangzhou China
| | - Tonghui Ma
- Hangzhou Jichenjunchuang Medical Laboratory, Co., Ltd. Hangzhou China
| | - Xiaoling Li
- Cancer Hospital of China Medical University Shenyang China
- Liaoning Cancer Hospital & Institute Shenyang China
| |
Collapse
|
158
|
SLC1A1-mediated cellular and mitochondrial influx of R-2-hydroxyglutarate in vascular endothelial cells promotes tumor angiogenesis in IDH1-mutant solid tumors. Cell Res 2022; 32:638-658. [PMID: 35459936 DOI: 10.1038/s41422-022-00650-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/08/2022] [Indexed: 12/24/2022] Open
Abstract
Mutant isocitrate dehydrogenase 1 (mIDH1) drives tumorigenesis via producing oncometabolite R-2-hydroxyglutarate (R-2-HG) across various tumor types. However, mIDH1 inhibitors appear only effective in hematological tumors. The therapeutic benefit in solid tumors remains elusive, likely due to the complex tumor microenvironment. In this study, we discover that R-2-HG produced by IDH1-mutant tumor cells is preferentially imported into vascular endothelial cells and remodels mitochondrial respiration to promote tumor angiogenesis, conferring a therapeutic vulnerability in IDH1-mutant solid tumors. Mechanistically, SLC1A1, a Na+-dependent glutamate transporter that is preferentially expressed in endothelial cells, facilitates the influx of R-2-HG from the tumor microenvironment into the endothelial cells as well as the intracellular trafficking of R-2-HG from cytoplasm to mitochondria. R-2-HG hijacks SLC1A1 to promote mitochondrial Na+/Ca2+ exchange, which activates the mitochondrial respiratory chain and fuels vascular endothelial cell migration in tumor angiogenesis. SLC1A1 deficiency in mice abolishes mIDH1-promoted tumor angiogenesis as well as the therapeutic benefit of mIDH1 inhibitor in solid tumors. Moreover, we report that HH2301, a newly discovered mIDH1 inhibitor, shows promising efficacy in treating IDH1-mutant cholangiocarcinoma in preclinical models. Together, we identify a new role of SLC1A1 as a gatekeeper of R-2-HG-mediated crosstalk between IDH1-mutant tumor cells and vascular endothelial cells, and demonstrate the therapeutic potential of mIDH1 inhibitors in treating IDH1-mutant solid tumors via disrupting R-2-HG-promoted tumor angiogenesis.
Collapse
|
159
|
Montesinos P, Recher C, Vives S, Zarzycka E, Wang J, Bertani G, Heuser M, Calado RT, Schuh AC, Yeh SP, Daigle SR, Hui J, Pandya SS, Gianolio DA, de Botton S, Döhner H. Ivosidenib and Azacitidine in IDH1-Mutated Acute Myeloid Leukemia. N Engl J Med 2022; 386:1519-1531. [PMID: 35443108 DOI: 10.1056/nejmoa2117344] [Citation(s) in RCA: 280] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The combination of ivosidenib - an inhibitor of mutant isocitrate dehydrogenase 1 (IDH1) - and azacitidine showed encouraging clinical activity in a phase 1b trial involving patients with newly diagnosed IDH1-mutated acute myeloid leukemia. METHODS In this phase 3 trial, we randomly assigned patients with newly diagnosed IDH1-mutated acute myeloid leukemia who were ineligible for intensive induction chemotherapy to receive oral ivosidenib (500 mg once daily) and subcutaneous or intravenous azacitidine (75 mg per square meter of body-surface area for 7 days in 28-day cycles) or to receive matched placebo and azacitidine. The primary end point was event-free survival, defined as the time from randomization until treatment failure (i.e., the patient did not have complete remission by week 24), relapse from remission, or death from any cause, whichever occurred first. RESULTS The intention-to-treat population included 146 patients: 72 in the ivosidenib-and-azacitidine group and 74 in the placebo-and-azacitidine group. At a median follow-up of 12.4 months, event-free survival was significantly longer in the ivosidenib-and-azacitidine group than in the placebo-and-azacitidine group (hazard ratio for treatment failure, relapse from remission, or death, 0.33; 95% confidence interval [CI], 0.16 to 0.69; P = 0.002). The estimated probability that a patient would remain event-free at 12 months was 37% in the ivosidenib-and-azacitidine group and 12% in the placebo-and-azacitidine group. The median overall survival was 24.0 months with ivosidenib and azacitidine and 7.9 months with placebo and azacitidine (hazard ratio for death, 0.44; 95% CI, 0.27 to 0.73; P = 0.001). Common adverse events of grade 3 or higher included febrile neutropenia (28% with ivosidenib and azacitidine and 34% with placebo and azacitidine) and neutropenia (27% and 16%, respectively); the incidence of bleeding events of any grade was 41% and 29%, respectively. The incidence of infection of any grade was 28% with ivosidenib and azacitidine and 49% with placebo and azacitidine. Differentiation syndrome of any grade occurred in 14% of the patients receiving ivosidenib and azacitidine and 8% of those receiving placebo and azacitidine. CONCLUSIONS Ivosidenib and azacitidine showed significant clinical benefit as compared with placebo and azacitidine in this difficult-to-treat population. Febrile neutropenia and infections were less frequent in the ivosidenib-and-azacitidine group than in the placebo-and-azacitidine group, whereas neutropenia and bleeding were more frequent in the ivosidenib-and-azacitidine group. (Funded by Agios Pharmaceuticals and Servier Pharmaceuticals; AGILE ClinicalTrials.gov number, NCT03173248.).
Collapse
Affiliation(s)
- Pau Montesinos
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Christian Recher
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Susana Vives
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Ewa Zarzycka
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Jianxiang Wang
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Giambattista Bertani
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Michael Heuser
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Rodrigo T Calado
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Andre C Schuh
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Su-Peng Yeh
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Scott R Daigle
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Jianan Hui
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Shuchi S Pandya
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Diego A Gianolio
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Stephane de Botton
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| | - Hartmut Döhner
- From Hospital Universitari i Politècnic La Fe, Valencia (P.M.), and Hospital Universitario Germans Trias i Pujol-Institut Català d'Oncologia Badalona, Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Badalona (S.V.) - both in Spain; Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire de Toulouse, Toulouse (C.R.), and Institut Gustave Roussy, Villejuif (S.B.) - both in France; Klinika Hematologii i Transplantologii, Uniwersyteckie Centrum Kliniczne, Gdansk, Poland (E.Z.); the Institute of Hematology and Hospital of Blood Disease, Peking Union Medical College, Tianjin, China (J.W.); Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan (G.B.); Hannover Medical School, Hannover (M.H.), and Ulm University Hospital, Ulm (H.D.) - both in Germany; Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil (R.T.C.); Princess Margaret Cancer Centre, Toronto (A.C.S.); China Medical University, Taichung, Taiwan (S.-P.Y.); and Servier Pharmaceuticals, Boston (S.R.D., J.H., S.S.P., D.A.G.)
| |
Collapse
|
160
|
Cardio-onco-metabolism: metabolic remodelling in cardiovascular disease and cancer. Nat Rev Cardiol 2022; 19:414-425. [PMID: 35440740 PMCID: PMC10112835 DOI: 10.1038/s41569-022-00698-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2022] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease and cancer are the two leading causes of morbidity and mortality in the world. The emerging field of cardio-oncology has revealed that these seemingly disparate disease processes are intertwined, owing to the cardiovascular sequelae of anticancer therapies, shared risk factors that predispose individuals to both cardiovascular disease and cancer, as well the possible potentiation of cancer growth by cardiac dysfunction. As a result, interest has increased in understanding the fundamental biological mechanisms that are central to the relationship between cardiovascular disease and cancer. Metabolism, appropriate regulation of energy, energy substrate utilization, and macromolecular synthesis and breakdown are fundamental processes for cellular and organismal survival. In this Review, we explore the emerging data identifying metabolic dysregulation as an important theme in cardio-oncology. We discuss the growing recognition of metabolic reprogramming in cardiovascular disease and cancer and view the novel area of cardio-oncology through the lens of metabolism.
Collapse
|
161
|
Kropp EM, Li Q. Mechanisms of Resistance to Targeted Therapies for Relapsed or Refractory Acute Myeloid Leukemia. Exp Hematol 2022; 111:13-24. [PMID: 35417742 PMCID: PMC10116852 DOI: 10.1016/j.exphem.2022.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/29/2022] [Accepted: 04/02/2022] [Indexed: 11/29/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive disease of clonal hematopoiesis with a high rate of relapse and refractory disease despite intensive therapy. Traditionally, relapsed or refractory AML has increased therapeutic resistance and poor long-term survival. In recent years, advancements in the mechanistic understanding of leukemogenesis have allowed for the development of targeted therapies. These therapies offer novel alternatives to intensive chemotherapy and have prolonged survival in relapsed or refractory AML. Unfortunately, a significant portion of patients do not respond to these therapies and relapse occurs in most patients who initially responded. This review focuses on the mechanisms of resistance to targeted therapies in relapsed or refractory AML.
Collapse
Affiliation(s)
- Erin M Kropp
- Department of Internal Medicine, University of Michigan-Ann Arbor, Ann Arbor, MI
| | - Qing Li
- Department of Internal Medicine, University of Michigan-Ann Arbor, Ann Arbor, MI.
| |
Collapse
|
162
|
Nakagawa M, Yamaguchi M, Endo M, Machida Y, Hattori A, Tanzawa F, Tsutsumi S, Kitabayashi I, Kawai A, Nakatani F. Clinical usefulness of 2-hydroxyglutarate as a biomarker in IDH-mutant chondrosarcoma. J Bone Oncol 2022; 34:100430. [PMID: 35493690 PMCID: PMC9048072 DOI: 10.1016/j.jbo.2022.100430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/18/2022] Open
Abstract
2-HG is expected to be a useful biomarker for diagnosing and treating IDH-mutant tumors. Both intratumoral and serum levels of 2-HG were significantly higher in IDH-mutant tumors. Serum 2-HG levels were correlated with tumor volume and tumor progression. MR spectroscopy (MRS) detected 2-HG peaks in a xenograft model of IDH-mutant chondrosarcoma. In vivo MRS can be a useful tool for determining the therapeutic effect of mutant IDH inhibitors.
Background Chondrosarcoma is a common form of malignant bone tumor with limited treatment options. Approximately half of chondrosarcomas harbor gain-of-function mutations in isocitrate dehydrogenase (IDH), and mutant IDH produces 2-hydroxyglutarate (2-HG), which is an oncometabolite that contributes to malignant transformation. Therefore, inhibiting 2-HG production is a novel and promising treatment for advanced chondrosarcoma. 2-HG is also expected to be a useful biomarker for the diagnosis and treatment of IDH-mutant tumors. However, few studies have confirmed this using chondrosarcoma clinical specimens. Non-invasive monitoring of 2-HG levels is useful to infer that mutant IDH inhibitors reach therapeutic targets and to confirm their therapeutic efficacy in clinical practice. Methods To evaluate the clinical utility of 2-HG as a surrogate biomarker for diagnosis and therapeutic efficacy, we measured intra-tumor and serum levels of 2-HG using frozen tissues and peripheral blood from patients with chondrosarcoma. We also developed a non-invasive method to detect intra-tumor 2-HG signals in vivo using magnetic resonance spectroscopy (MRS) Results Both intratumoral and serum 2-HG levels were significantly elevated in IDH-mutant tumors, and these levels correlated with decreased survival. Furthermore, we detected intratumoral 2-HG peaks using MR spectroscopy in a xenograft model of IDH-mutant chondrosarcoma, and observed that 2-HG peak signals disappeared after administering an inhibitor of mutant IDH1. Conclusions Our findings suggest that both intratumoral and serum 2-HG levels represent potentially useful biomarkers for IDH-mutant tumors and that the 2-HG signal in MR spectra has potential value as a non-invasive biomarker. Taken together, these findings may positively impact the clinical development of mutant IDH inhibitors for the treatment of advanced chondrosarcoma.
Collapse
Affiliation(s)
- Makoto Nakagawa
- Department of Musculoskeletal Oncology and Rehabilitation, National Cancer Center Hospital, Tokyo, Japan
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Hematological Malignancy, National Cancer Center Research Institute, Tokyo, Japan
| | - Masayuki Yamaguchi
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Makoto Endo
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yukino Machida
- Department of Veterinary Pathology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Ayuna Hattori
- Division of Hematological Malignancy, National Cancer Center Research Institute, Tokyo, Japan
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Fumie Tanzawa
- Oncology Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Shinji Tsutsumi
- Oncology Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Issay Kitabayashi
- Division of Hematological Malignancy, National Cancer Center Research Institute, Tokyo, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology and Rehabilitation, National Cancer Center Hospital, Tokyo, Japan
| | - Fumihiko Nakatani
- Department of Musculoskeletal Oncology and Rehabilitation, National Cancer Center Hospital, Tokyo, Japan
- Corresponding author at: Department of Musculoskeletal Oncology and Rehabilitation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| |
Collapse
|
163
|
Tu R, Ma J, Zhang P, Kang Y, Xiong X, Zhu J, Li M, Zhang C. The emerging role of deubiquitylating enzymes as therapeutic targets in cancer metabolism. Cancer Cell Int 2022; 22:130. [PMID: 35307036 PMCID: PMC8935717 DOI: 10.1186/s12935-022-02524-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 02/14/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractCancer cells must rewire cellular metabolism to satisfy the unbridled proliferation, and metabolic reprogramming provides not only the advantage for cancer cell proliferation but also new targets for cancer treatment. However, the plasticity of the metabolic pathways makes them very difficult to target. Deubiquitylating enzymes (DUBs) are proteases that cleave ubiquitin from the substrate proteins and process ubiquitin precursors. While the molecular mechanisms are not fully understood, many DUBs have been shown to be involved in tumorigenesis and progression via controlling the dysregulated cancer metabolism, and consequently recognized as potential drug targets for cancer treatment. In this article, we summarized the significant progress in understanding the key roles of DUBs in cancer cell metabolic rewiring and the opportunities for the application of DUBs inhibitors in cancer treatment, intending to provide potential implications for both research purpose and clinical applications.
Collapse
|
164
|
Zuidhof HR, Calkhoven CF. Oncogenic and tumor-suppressive functions of the RNA demethylase FTO. Cancer Res 2022; 82:2201-2212. [PMID: 35303057 DOI: 10.1158/0008-5472.can-21-3710] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/04/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022]
Abstract
The epitranscriptome represents the more than 140 types of chemically varying and reversable RNA modifications affecting RNA fate. Among these, the most relevant for this review are the mRNA-modifications N6-methyladenosine (m6A) and N6,2'-O-dimethyladenosine (m6Am). Epitranscriptomic mRNA biology involves RNA methyltransferases (so called "writers"), RNA demethylases ("erasers"), and RNA-binding proteins ("readers") that interact with methylation sites to determine the functional outcome of the modification. In this review, we discuss the role of a specific RNA demethylase encoded by the fat mass and obesity associated gene (FTO) in cancer. FTO initially became known as the strongest genetic link for human obesity. Only in 2010, 16 years after its discovery, was its enzymatic function as a demethylase clarified, and only recently has its role in the development of cancer been revealed. FTO functions are challenging to study and interpret because of its genome-wide effects on transcript turnover and translation. We review the discovery of FTO and its enzymatic function, the tumor-promoting and suppressive roles of FTO in selected cancer types, and its potential as a therapeutic target.
Collapse
|
165
|
Role of Biomarkers in FLT3 AML. Cancers (Basel) 2022; 14:cancers14051164. [PMID: 35267471 PMCID: PMC8909069 DOI: 10.3390/cancers14051164] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Genetically heterogeneous disorder acute myeloid leukemia (AML) is marked by recurring mutations in FLT3. Current FLT3 inhibitors and other emerging inhibitors have helped in the improvement of the quality of standard of care therapies; however, the overall survival of the patients remains static. This is due to numerous mutations in FLT3, which causes resistance against these FLT3 inhibitors. For effective treatment of AML patients, alternative approaches are required to overcome this resistance. Here, we will summarize the biomarkers for FLT3 inhibitors in AML, as well as the alternative measures to overcome resistance to the current therapies. Abstract Acute myeloid leukemia is a disease characterized by uncontrolled proliferation of clonal myeloid blast cells that are incapable of maturation to leukocytes. AML is the most common leukemia in adults and remains a highly fatal disease with a five-year survival rate of 24%. More than 50% of AML patients have mutations in the FLT3 gene, rendering FLT3 an attractive target for small-molecule inhibition. Currently, there are several FLT3 inhibitors in the clinic, and others remain in clinical trials. However, these inhibitors face challenges due to lack of efficacy against several FLT3 mutants. Therefore, the identification of biomarkers is vital to stratify AML patients and target AML patient population with a particular FLT3 mutation. Additionally, there is an unmet need to identify alternative approaches to combat the resistance to FLT3 inhibitors. Here, we summarize the current knowledge on the utilization of diagnostic, prognostic, predictive, and pharmacodynamic biomarkers for FLT3-mutated AML. The resistance mechanisms to various FLT3 inhibitors and alternative approaches to combat this resistance are also discussed and presented.
Collapse
|
166
|
Byun JM, Yoo SJ, Kim HJ, Ahn JS, Koh Y, Jang JH, Yoon SS. IDH1/2 mutations in acute myeloid leukemia. Blood Res 2022; 57:13-19. [PMID: 35197370 PMCID: PMC8958365 DOI: 10.5045/br.2021.2021152] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/14/2021] [Accepted: 12/24/2021] [Indexed: 11/30/2022] Open
Abstract
The mutational and epigenetic landscape of acute myeloid leukemia (AML) has become increasingly well understood in recent years, informing on biological targets for precision medicine. Among the most notable findings was the recognition of mutational hot-spots in the isocitrate dehydrogenase (IDH) genes. In this review, we provide an overview on the IDH1/2 mutation landscape in Korean AML patients, and compare it with available public data. We also discuss the role of IDH1/2 mutations as biomarkers and drug targets. Taken together, occurrence of IDH1/2 mutations is becoming increasingly important in AML treatment, thus requiring thorough examination and follow-up throughout the clinical course of the disease.
Collapse
Affiliation(s)
- Ja Min Byun
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seung-Joo Yoo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyeong-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Jae-Sook Ahn
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jun Ho Jang
- Division of Hematology and Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
167
|
Super enhancers as master gene regulators in the pathogenesis of hematologic malignancies. Biochim Biophys Acta Rev Cancer 2022; 1877:188697. [PMID: 35150791 DOI: 10.1016/j.bbcan.2022.188697] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/20/2022] [Accepted: 02/04/2022] [Indexed: 12/17/2022]
Abstract
Transcriptional deregulation of multiple oncogenes, tumor suppressors and survival pathways is a cancer cell hallmark. Super enhancers (SE) are long stretches of active enhancers in close linear proximity that ensure extraordinarily high expression levels of key genes associated with cell lineage, function and survival. SE landscape is intrinsically prone to changes and reorganization during the course of normal cell differentiation. This functional plasticity is typically utilized by cancer cells, which remodel their SE landscapes to ensure oncogenic transcriptional reprogramming. Multiple recent studies highlighted structural genetic mechanisms in non-coding regions that create new SE or hijack already existing ones. In addition, alterations in abundance/activity of certain SE-associated proteins or certain viral infections can elicit new super enhancers and trigger SE-driven transcriptional changes. For these reasons, SE profiling emerged as a powerful tool for discovering the core transcriptional regulatory circuits in tumor cells. This, in turn, provides new insights into cancer cell biology, and identifies main nodes of key cellular pathways to be potentially targeted. Since SEs are susceptible to inhibition, their disruption results in exponentially amassing 'butterfly' effect on gene expression and cell function. Moreover, many of SE elements are druggable, opening new therapeutic opportunities. Indeed, SE targeting drugs have been studied preclinically in various hematologic malignancies with promising effects. Herein, we review the unique features of SEs, present different cis- and trans-acting mechanisms through which hematologic tumor cells acquire SEs, and finally, discuss the potential of SE targeting in the therapy of hematologic malignancies.
Collapse
|
168
|
Lewuillon C, Laguillaumie MO, Quesnel B, Idziorek T, Touil Y, Lemonnier L. Put in a “Ca2+ll” to Acute Myeloid Leukemia. Cells 2022; 11:cells11030543. [PMID: 35159351 PMCID: PMC8834247 DOI: 10.3390/cells11030543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 02/05/2023] Open
Abstract
Acute myeloid leukemia (AML) is a clonal disorder characterized by genetic aberrations in myeloid primitive cells (blasts) which lead to their defective maturation/function and their proliferation in the bone marrow (BM) and blood of affected individuals. Current intensive chemotherapy protocols result in complete remission in 50% to 80% of AML patients depending on their age and the AML type involved. While alterations in calcium signaling have been extensively studied in solid tumors, little is known about the role of calcium in most hematologic malignancies, including AML. Our purpose with this review is to raise awareness about this issue and to present (i) the role of calcium signaling in AML cell proliferation and differentiation and in the quiescence of hematopoietic stem cells; (ii) the interplay between mitochondria, metabolism, and oxidative stress; (iii) the effect of the BM microenvironment on AML cell fate; and finally (iv) the mechanism by which chemotherapeutic treatments modify calcium homeostasis in AML cells.
Collapse
Affiliation(s)
- Clara Lewuillon
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Marie-Océane Laguillaumie
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Bruno Quesnel
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Thierry Idziorek
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Yasmine Touil
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Loïc Lemonnier
- Univ. Lille, Inserm, U1003—PHYCEL—Physiologie Cellulaire, F-59000 Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, F-59655 Villeneuve d’Ascq, France
- Correspondence:
| |
Collapse
|
169
|
Muto T, Guillamot M, Yeung J, Fang J, Bennett J, Nadorp B, Lasry A, Redondo LZ, Choi K, Gong Y, Walker CS, Hueneman K, Bolanos LC, Barreyro L, Lee LH, Greis KD, Vasyliev N, Khodadadi-Jamayran A, Nudler E, Lujambio A, Lowe SW, Aifantis I, Starczynowski DT. TRAF6 functions as a tumor suppressor in myeloid malignancies by directly targeting MYC oncogenic activity. Cell Stem Cell 2022; 29:298-314.e9. [PMID: 35045331 PMCID: PMC8822959 DOI: 10.1016/j.stem.2021.12.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/05/2021] [Accepted: 12/15/2021] [Indexed: 02/05/2023]
Abstract
Clonal hematopoiesis (CH) is an aging-associated condition characterized by the clonal outgrowth of pre-leukemic cells that acquire specific mutations. Although individuals with CH are healthy, they are at an increased risk of developing myeloid malignancies, suggesting that additional alterations are needed for the transition from a pre-leukemia stage to frank leukemia. To identify signaling states that cooperate with pre-leukemic cells, we used an in vivo RNAi screening approach. One of the most prominent genes identified was the ubiquitin ligase TRAF6. Loss of TRAF6 in pre-leukemic cells results in overt myeloid leukemia and is associated with MYC-dependent stem cell signatures. TRAF6 is repressed in a subset of patients with myeloid malignancies, suggesting that subversion of TRAF6 signaling can lead to acute leukemia. Mechanistically, TRAF6 ubiquitinates MYC, an event that does not affect its protein stability but rather represses its functional activity by antagonizing an acetylation modification.
Collapse
Affiliation(s)
- Tomoya Muto
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Maria Guillamot
- Department of Pathology and Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Jennifer Yeung
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Jing Fang
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Joshua Bennett
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Bettina Nadorp
- Department of Pathology and Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Audrey Lasry
- Department of Pathology and Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Luna Zea Redondo
- Department of Pathology and Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yixiao Gong
- Department of Pathology and Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Callum S Walker
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kathleen Hueneman
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lyndsey C Bolanos
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Laura Barreyro
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lynn H Lee
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Kenneth D Greis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Nikita Vasyliev
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY 10016, USA
| | - Alireza Khodadadi-Jamayran
- Applied Bioinformatics Laboratories and Genome Technology Center, NYU School of Medicine, New York, NY 10016, USA
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY 10016, USA
| | - Amaia Lujambio
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Scott W Lowe
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Howard Hughes Medical Institute, Chevy Chase, MD 201815, USA
| | - Iannis Aifantis
- Department of Pathology and Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA.
| | - Daniel T Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
170
|
Peng Y, Liu H, Liu J, Long J. Post-translational modifications on mitochondrial metabolic enzymes in cancer. Free Radic Biol Med 2022; 179:11-23. [PMID: 34929314 DOI: 10.1016/j.freeradbiomed.2021.12.264] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/26/2021] [Accepted: 12/15/2021] [Indexed: 12/22/2022]
Abstract
Mitochondrion is the powerhouse of the cell. The research of nearly a century has expanded our understanding of mitochondrion, far beyond the view that mitochondrion is an important energy generator of cells. During the initiation, growth and survival of tumor cells, significant mitochondrial metabolic changes have taken place in the important enzymes of respiratory chain and tricarboxylic acid cycle, mitochondrial biogenesis and dynamics, oxidative stress regulation and molecular signaling. Therefore, mitochondrial metabolic proteins are the key mediators of tumorigenesis. Post-translational modification is the molecular switch that regulates protein function. Understanding how these mitochondria-related post-translational modification function during tumorigenesis will bring new ideas for the next generation of cancer treatment.
Collapse
Affiliation(s)
- Yunhua Peng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Huadong Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; University of Health and Rehabilitation Sciences, Qingdao, 266071, China
| | - Jiangang Long
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
171
|
Kornauth C, Pemovska T, Vladimer GI, Bayer G, Bergmann M, Eder S, Eichner R, Erl M, Esterbauer H, Exner R, Felsleitner-Hauer V, Forte M, Gaiger A, Geissler K, Greinix HT, Gstöttner W, Hacker M, Hartmann BL, Hauswirth AW, Heinemann T, Heintel D, Hoda MA, Hopfinger G, Jaeger U, Kazianka L, Kenner L, Kiesewetter B, Krall N, Krajnik G, Kubicek S, Le T, Lubowitzki S, Mayerhoefer ME, Menschel E, Merkel O, Miura K, Müllauer L, Neumeister P, Noesslinger T, Ocko K, Öhler L, Panny M, Pichler A, Porpaczy E, Prager GW, Raderer M, Ristl R, Ruckser R, Salamon J, Schiefer AI, Schmolke AS, Schwarzinger I, Selzer E, Sillaber C, Skrabs C, Sperr WR, Srndic I, Thalhammer R, Valent P, van der Kouwe E, Vanura K, Vogt S, Waldstein C, Wolf D, Zielinski CC, Zojer N, Simonitsch-Klupp I, Superti-Furga G, Snijder B, Staber PB. Functional Precision Medicine Provides Clinical Benefit in Advanced Aggressive Hematologic Cancers and Identifies Exceptional Responders. Cancer Discov 2022; 12:372-387. [PMID: 34635570 PMCID: PMC9762339 DOI: 10.1158/2159-8290.cd-21-0538] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/22/2021] [Accepted: 09/24/2021] [Indexed: 01/07/2023]
Abstract
Personalized medicine aims to match the right drug with the right patient by using specific features of the individual patient's tumor. However, current strategies of personalized therapy matching provide treatment opportunities for less than 10% of patients with cancer. A promising method may be drug profiling of patient biopsy specimens with single-cell resolution to directly quantify drug effects. We prospectively tested an image-based single-cell functional precision medicine (scFPM) approach to guide treatments in 143 patients with advanced aggressive hematologic cancers. Fifty-six patients (39%) were treated according to scFPM results. At a median follow-up of 23.9 months, 30 patients (54%) demonstrated a clinical benefit of more than 1.3-fold enhanced progression-free survival compared with their previous therapy. Twelve patients (40% of responders) experienced exceptional responses lasting three times longer than expected for their respective disease. We conclude that therapy matching by scFPM is clinically feasible and effective in advanced aggressive hematologic cancers. SIGNIFICANCE: This is the first precision medicine trial using a functional assay to instruct n-of-one therapies in oncology. It illustrates that for patients lacking standard therapies, high-content assay-based scFPM can have a significant value in clinical therapy guidance based on functional dependencies of each patient's cancer.See related commentary by Letai, p. 290.This article is highlighted in the In This Issue feature, p. 275.
Collapse
Affiliation(s)
- Christoph Kornauth
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center Vienna, Medical University of Vienna and Vienna General Hospital, Vienna, Austria
| | - Tea Pemovska
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Gregory I Vladimer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Exscientia GmbH, Vienna, Austria
| | - Günther Bayer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Michael Bergmann
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Sandra Eder
- Department of Internal Medicine and Hematology/Oncology, Klinikum Klagenfurt, Klagenfurt, Austria
| | - Ruth Eichner
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Martin Erl
- Abteilung für Innere Medizin, Krankenhaus der Barmherzigen Brüder Salzburg, Salzburg, Austria
| | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ruth Exner
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Maurizio Forte
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Alexander Gaiger
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center Vienna, Medical University of Vienna and Vienna General Hospital, Vienna, Austria
| | - Klaus Geissler
- Medical School, Sigmund Freud University, Vienna, Austria
| | - Hildegard T Greinix
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Wolfgang Gstöttner
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | | | - Alexander W Hauswirth
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Tim Heinemann
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Daniel Heintel
- Division of Medicine I, Klinik Ottakring, Vienna, Austria
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Georg Hopfinger
- Third Medical Department, Centre for Oncology and Haematology, Klinik Favoriten, Vienna, Austria
| | - Ulrich Jaeger
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center Vienna, Medical University of Vienna and Vienna General Hospital, Vienna, Austria
| | - Lukas Kazianka
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Lukas Kenner
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Barbara Kiesewetter
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Nikolaus Krall
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Exscientia GmbH, Vienna, Austria
| | - Gerhard Krajnik
- Department of Medicine I, Universitätsklinikum St. Pölten, St. Pölten, Austria
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Trang Le
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Simone Lubowitzki
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Marius E Mayerhoefer
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisabeth Menschel
- Third Medical Department, Hematology & Oncology, Hanusch Hospital, Vienna, Austria
| | - Olaf Merkel
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Katsuhiro Miura
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Peter Neumeister
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Thomas Noesslinger
- Third Medical Department, Hematology & Oncology, Hanusch Hospital, Vienna, Austria
| | - Katharina Ocko
- Pharmacy Department, Vienna General Hospital, Vienna, Austria
| | - Leopold Öhler
- Internal Medicine I, Department of Oncology, St. Josef Hospital, Vienna, Austria
| | - Michael Panny
- Third Medical Department, Hematology & Oncology, Hanusch Hospital, Vienna, Austria
| | - Alexander Pichler
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Edit Porpaczy
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Gerald W Prager
- Comprehensive Cancer Center Vienna, Medical University of Vienna and Vienna General Hospital, Vienna, Austria
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Markus Raderer
- Comprehensive Cancer Center Vienna, Medical University of Vienna and Vienna General Hospital, Vienna, Austria
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Robin Ristl
- Section for Medical Statistics, Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | | | - Julius Salamon
- Department of Medicine, Landesklinikum Waidhofen a.d. Ybbs, Waidhofen-Ybbs, Austria
| | - Ana-Iris Schiefer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Ann-Sofie Schmolke
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Ilse Schwarzinger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Edgar Selzer
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
| | - Christian Sillaber
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Cathrin Skrabs
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang R Sperr
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Ismet Srndic
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Renate Thalhammer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Peter Valent
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Emiel van der Kouwe
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Katrina Vanura
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Stefan Vogt
- Department of Medicine and Oncology, LKH Wiener Neustadt, Wiener Neustadt, Austria
| | - Cora Waldstein
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
| | - Dominik Wolf
- Department of Internal Medicine V, Department of Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Niklas Zojer
- Division of Medicine I, Klinik Ottakring, Vienna, Austria
| | | | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Berend Snijder
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Philipp B Staber
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria.
- Comprehensive Cancer Center Vienna, Medical University of Vienna and Vienna General Hospital, Vienna, Austria
| |
Collapse
|
172
|
Leibetseder A, Preusser M, Berghoff AS. New Approaches with Precision Medicine in Adult Brain Tumors. Cancers (Basel) 2022; 14:712. [PMID: 35158978 PMCID: PMC8833635 DOI: 10.3390/cancers14030712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/13/2022] [Accepted: 01/24/2022] [Indexed: 11/30/2022] Open
Abstract
Primary central nervous system (CNS) tumors represent a heterogenous group of tumors. The 2021 fifth edition of the WHO Classification of Tumors of the CNS emphasizes the advanced role of molecular diagnostics with routine implementation of molecular biomarkers in addition to histologic features in the classification of CNS tumors. Thus, novel diagnostic methods such as DNA methylome profiling are increasingly used to provide a more precise diagnostic work-up of CNS tumors. In addition to these diagnostic precision medicine advantages, molecular alterations are also addressed therapeutically with targeted therapies. Like in other tumor entities, precision medicine has therefore also arrived in the treatment of CNS malignancies as the application of targeted therapies has shown promising response rates. Nevertheless, large prospective studies are currently missing as most targeted therapies were evaluated in single arm, basket, or platform trials. In this review, we focus on the current evidence of precision medicine in the treatment of primary CNS tumors in adults. We outline the pathogenic background and prevalence of the most frequent targetable genetic alterations and summarize the existing evidence of precision medicine approaches for the treatment of primary CNS tumors.
Collapse
Affiliation(s)
- Annette Leibetseder
- Department of Neurology 1, Kepler University Hospital, Johannes Kepler University Linz, 4020 Linz, Austria;
- Department of Internal Medicine and Neurooncology, Neuromed Campus, Kepler University Hospital, 4020 Linz, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Anna Sophie Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
173
|
Miwa S, Yamamoto N, Hayashi K, Takeuchi A, Igarashi K, Tsuchiya H. Therapeutic Targets and Emerging Treatments in Advanced Chondrosarcoma. Int J Mol Sci 2022; 23:ijms23031096. [PMID: 35163019 PMCID: PMC8834928 DOI: 10.3390/ijms23031096] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/25/2022] Open
Abstract
Due to resistance to standard anticancer agents, it is difficult to control the disease progression in patients with metastatic or unresectable chondrosarcoma. Novel therapeutic approaches, such as molecule-targeting drugs and immunotherapy, are required to improve clinical outcomes in patients with advanced chondrosarcoma. Recent studies have suggested several promising biomarkers and therapeutic targets for chondrosarcoma, including IDH1/2 and COL2A1. Several molecule-targeting agents and immunotherapies have shown favorable antitumor activity in clinical studies in patients with advanced chondrosarcomas. This review summarizes recent basic studies on biomarkers and molecular targets and recent clinical studies on the treatment of chondrosarcomas.
Collapse
|
174
|
Challenges and Advances in Chimeric Antigen Receptor Therapy for Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:cancers14030497. [PMID: 35158765 PMCID: PMC8833567 DOI: 10.3390/cancers14030497] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
The advent of chimeric antigen receptor (CAR) T-cell therapy has led to dramatic remission rates in multiple relapsed/refractory hematologic malignancies. While CAR T-cell therapy has been particularly successful as a treatment for B-cell malignancies, effectively treating acute myeloid leukemia (AML) with CARs has posed a larger challenge. AML not only creates an immunosuppressive tumor microenvironment that dampens CAR T-cell responses, but it also lacks many unique tumor-associated antigens, making leukemic-specific targeting difficult. One advantage of CAR T-cell therapy compared to alternative treatment options is the ability to provide prolonged antigen-specific immune effector and surveillance functions. Since many AML CAR targets under investigation including CD33, CD117, and CD123 are also expressed on hematopoietic stem cells, CAR T-cell therapy can lead to severe and potentially lethal myeloablation. Novel strategies to combat these issues include creation of bispecific CARs, CAR T-cell "safety switches", TCR-like CARs, NK CARs, and universal CARs, but all vary in their ability to provide a sustained remission, and consolidation with an allogeneic hematopoietic cell transplantation (allo-HCT) will be necessary in most cases This review highlights the delicate balance between effectively eliminating AML blasts and leukemic stem cells, while preserving the ability for bone marrow to regenerate. The impact of CAR therapy on treatment landscape of AML and changing scope of allo-HCT is discussed. Continued advances in AML CAR therapy would be of great benefit to a disease that still has high morbidity and mortality.
Collapse
|
175
|
Belloucif Y, Lobry C. Super-Enhancers Dysregulations in Hematological Malignancies. Cells 2022; 11:196. [PMID: 35053311 PMCID: PMC8774084 DOI: 10.3390/cells11020196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/17/2021] [Accepted: 12/27/2021] [Indexed: 01/27/2023] Open
Abstract
Hematological malignancies affecting either the lymphoid or the myeloid lineages involve epigenetic mutations or dysregulation in the majority of cases. These epigenetic abnormalities can affect regulatory elements in the genome and, particularly, enhancers. Recently, large regulatory elements known as super-enhancers, initially identified for their critical roles in cell-type specific expression regulation of genes controlling cell identity, have been shown to also be involved in tumorigenesis in many cancer types and hematological malignancies via the regulation of numerous oncogenes, including MYC. In this review, we highlight the existing links between super-enhancers and hematological malignancies, with a particular focus on acute myeloid leukemia, a clonal hematopoietic neoplasm with dismal outcomes, resulting in an uncontrolled proliferation of myeloblasts, abnormally blocked during differentiation and accumulating within the patient's bone marrow. We report recent works, performed during the last few years, treating this subject and consider the possibility of targeting oncogenic regulatory elements, as well as the effectiveness and limitations reported so far for such strategies.
Collapse
Affiliation(s)
| | - Camille Lobry
- INSERM U944, CNRS UMR7212, Institut de Recherche Saint Louis, Université de Paris, 75010 Paris, France;
| |
Collapse
|
176
|
Combining Isocitrate Dehydrogenase Inhibitors With Existing Regimens in Acute Myeloid Leukemia. Cancer J 2022; 28:21-28. [DOI: 10.1097/ppo.0000000000000570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
177
|
Diaz-Flores E, Meyer T, Giorkallos A. Evolution of Artificial Intelligence-Powered Technologies in Biomedical Research and Healthcare. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2022; 182:23-60. [DOI: 10.1007/10_2021_189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
178
|
Structure and Function of TET Enzymes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1389:239-267. [DOI: 10.1007/978-3-031-11454-0_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
179
|
Molenaar RJ, Wilmink JW. IDH1/2 Mutations in Cancer Stem Cells and Their Implications for Differentiation Therapy. J Histochem Cytochem 2021; 70:83-97. [PMID: 34967233 PMCID: PMC8721574 DOI: 10.1369/00221554211062499] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Isocitrate dehydrogenase 1 and 2 (IDH1/2) are enzymes recurrently mutated in various types of cancer, including glioma, cholangiocarcinoma, chondrosarcoma, and acute myeloid leukemia. Mutant IDH1/2 induce a block in differentiation and thereby contribute to the stemness and oncogenesis of their cells of origin. Recently, small-molecule inhibitors of mutant IDH1/2 have been Food and Drug Administration-approved for the treatment of IDH1/2-mutated acute myeloid leukemia. These inhibitors decrease the stemness of the targeted IDH1/2-mutated cancer cells and induce their differentiation to more mature cells. In this review, we elucidate the mechanisms by which mutant IDH1/2 induce a block in differentiation and the biological and clinical effects of the release into differentiation by mutant-IDH1/2 inhibitors. (J Histochem Cytochem 70:83-97, 2022).
Collapse
Affiliation(s)
- Remco J Molenaar
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands.,Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Johanna W Wilmink
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
180
|
Hvinden IC, Cadoux-Hudson T, Schofield CJ, McCullagh JS. Metabolic adaptations in cancers expressing isocitrate dehydrogenase mutations. Cell Rep Med 2021; 2:100469. [PMID: 35028610 PMCID: PMC8714851 DOI: 10.1016/j.xcrm.2021.100469] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The most frequently mutated metabolic genes in human cancer are those encoding the enzymes isocitrate dehydrogenase 1 (IDH1) and IDH2; these mutations have so far been identified in more than 20 tumor types. Since IDH mutations were first reported in glioma over a decade ago, extensive research has revealed their association with altered cellular processes. Mutations in IDH lead to a change in enzyme function, enabling efficient conversion of 2-oxoglutarate to R-2-hydroxyglutarate (R-2-HG). It is proposed that elevated cellular R-2-HG inhibits enzymes that regulate transcription and metabolism, subsequently affecting nuclear, cytoplasmic, and mitochondrial biochemistry. The significance of these biochemical changes for tumorigenesis and potential for therapeutic exploitation remains unclear. Here we comprehensively review reported direct and indirect metabolic changes linked to IDH mutations and discuss their clinical significance. We also review the metabolic effects of first-generation mutant IDH inhibitors and highlight the potential for combination treatment strategies and new metabolic targets.
Collapse
Affiliation(s)
- Ingvild Comfort Hvinden
- Chemistry Research Laboratory, 12 Mansfield Road, Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| | - Tom Cadoux-Hudson
- Chemistry Research Laboratory, 12 Mansfield Road, Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| | - Christopher J. Schofield
- Chemistry Research Laboratory, 12 Mansfield Road, Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
- Ineos Oxford Institute for Antimicrobial Research, 12 Mansfield Road, Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| | - James S.O. McCullagh
- Chemistry Research Laboratory, 12 Mansfield Road, Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK
| |
Collapse
|
181
|
Xiao G, Gao X, Li L, Liu C, Liu Z, Peng H, Xia X, Yi X, Zhou R. An Immune-Related Prognostic Signature for Predicting Clinical Outcomes and Immune Landscape in IDH-Mutant Lower-Grade Gliomas. JOURNAL OF ONCOLOGY 2021; 2021:3766685. [PMID: 34961815 PMCID: PMC8710162 DOI: 10.1155/2021/3766685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/30/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND IDH mutation is the most common in diffuse LGGs, correlated with a favorable prognosis. However, the IDH-mutant LGGs patients with poor prognoses need to be identified, and the potential mechanism leading to a worse outcome and treatment options needs to be investigated. METHODS A six-gene immune-related prognostic signature in IDH-mutant LGGs was constructed based on two public datasets and univariate, multivariate, and LASSO Cox regression analysis. Patients were divided into low- and high-risk groups based on the median risk score in the training and validation sets. We analyzed enriched pathways and immune cell infiltration, applying the GSEA and the immune evaluation algorithms. RESULTS Stratification and multivariate Cox analysis unveiled that the six-gene signature was an independent prognostic factor. The signature (0.806/0.795/0.822) showed a remarkable prognostic performance, with 1-, 3-, and 5-year time-dependent AUC, higher than for grade (0.612/0.638/0.649) and 1p19q codeletion status (0.606/0.658/0.676). High-risk patients had higher infiltrating immune cells. However, the specific immune escape was observed in the high-risk group after immune activation, owing to increasing immunosuppressive cells, inhibitory cytokines, and immune checkpoint molecules. Moreover, a novel nomogram model was developed to evaluate the survival in IDH-mutant LGGs patients. CONCLUSION The six-gene signature could be a promising prognostic biomarker, which is promising to promote individual therapy and improve the clinical outcomes of IDH-mutant gliomas. The study also refined the current classification system of IDH-mutant gliomas, classifying patients into two subtypes with distinct immunophenotypes and overall survival.
Collapse
Affiliation(s)
- Gang Xiao
- Department of Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xuan Gao
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- GenePlus- Shenzhen Clinical Laboratory, Shenzhen 518122, China
| | - Lifeng Li
- Geneplus-Beijing, Beijing 102205, China
| | - Chao Liu
- Department of Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhiyuan Liu
- Department of Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Haiqin Peng
- Department of Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | | | - Xin Yi
- Geneplus-Beijing, Beijing 102205, China
| | - Rongrong Zhou
- Department of Radiation Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
182
|
Cadoux-Hudson T, Schofield CJ, McCullagh JS. Isocitrate dehydrogenase gene variants in cancer and their clinical significance. Biochem Soc Trans 2021; 49:2561-2572. [PMID: 34854890 PMCID: PMC8786286 DOI: 10.1042/bst20210277] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/13/2021] [Accepted: 10/18/2021] [Indexed: 11/30/2022]
Abstract
Human isocitrate dehydrogenase (IDH) genes encode for the IDH1, 2 & 3 isoenzymes which catalyse the formation of 2-oxoglutarate from isocitrate and are essential for normal mammalian metabolism. Although mutations in these genes in cancer were long thought to lead to a 'loss of function', combined genomic and metabolomic studies led to the discovery that a common IDH 1 mutation, present in low-grade glioma and acute myeloid leukaemia (AML), yields a variant (R132H) with a striking change of function leading to the production of (2R)-hydroxyglutarate (2HG) which consequently accumulates in large quantities both within and outside cells. Elevated 2HG is proposed to promote tumorigenesis, although the precise mechanism by which it does this remains uncertain. Inhibitors of R132H IDH1, and other subsequently identified cancer-linked 2HG producing IDH variants, are approved for clinical use in the treatment of chemotherapy-resistant AML, though resistance enabled by additional substitutions has emerged. In this review, we provide a current overview of cancer linked IDH mutations focussing on their distribution in different cancer types, the effects of substitution mutations on enzyme activity, the mode of action of recently developed inhibitors, and their relationship with emerging resistance-mediating double mutations.
Collapse
Affiliation(s)
- Thomas Cadoux-Hudson
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Institute for Antimicrobial Research, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Christopher J. Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Institute for Antimicrobial Research, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - James S.O. McCullagh
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Institute for Antimicrobial Research, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| |
Collapse
|
183
|
Abstract
Dysregulation of DNA damage response and repair (DDR) contributes to oncogenesis, yet also generates the potential for targeted cancer therapies by exploiting synthetic lethal interactions. Oncometabolites, small intermediates of metabolism overproduced in certain cancers, have emerged as a new mechanism of DDR modulation through their effects on multiple DNA repair pathways. Increasing evidence suggests that oncometabolite-induced DDR defects may offer the opportunity for tumor-selective chemo- and radio-sensitization. Here we review the biology of oncometabolites and diverse mechanisms by which they impact DDR, with a focus on emerging therapeutic strategies and ongoing clinical trials targeting oncometabolite-induced DDR defects in cancer.
Collapse
Affiliation(s)
- Susan E Gueble
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
184
|
Racial and ethnic survival disparities in patients with haematological malignancies in the USA: time to stop ignoring the numbers. THE LANCET HAEMATOLOGY 2021; 8:e947-e954. [DOI: 10.1016/s2352-3026(21)00303-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 09/05/2021] [Accepted: 09/21/2021] [Indexed: 12/17/2022]
|
185
|
Dow J, Krysztofiak A, Liu Y, Colon-Rios DA, Rogers FA, Glazer PM. Vulnerability of IDH1-Mutant Cancers to Histone Deacetylase Inhibition via Orthogonal Suppression of DNA Repair. Mol Cancer Res 2021; 19:2057-2067. [PMID: 34535560 PMCID: PMC8642278 DOI: 10.1158/1541-7786.mcr-21-0456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/06/2021] [Accepted: 09/10/2021] [Indexed: 11/16/2022]
Abstract
Exploitation of DNA repair defects has enabled major advances in treating specific cancers. Recent work discovered that the oncometabolite 2-hydroxyglutarate (2-HG), produced by neomorphic isocitrate dehydrogenase 1/2 (IDH1/2) mutations, confers a homology-directed repair (HDR) defect through 2-HG-induced histone hypermethylation masking HDR signaling. Here, we report that IDH1-mutant cancer cells are profoundly sensitive to the histone deacetylase inhibitor (HDACi) vorinostat, by further suppressing the residual HDR in 2-HG-producing cells. Vorinostat downregulates repair factors BRCA1 and RAD51 via disrupted E2F-factor regulation, causing increased DNA double-strand breaks, reduced DNA repair factor foci, and functional HDR deficiency even beyond 2-HG's effects. This results in greater cell death of IDH1-mutant cells and confers synergy with radiation and PARPi, both against cells in culture and patient-derived tumor xenografts. Our work identifies HDACi's utility against IDH1-mutant cancers, and presents IDH1/2 mutations as potential biomarkers to guide trials testing HDACi in gliomas and other malignancies. IMPLICATIONS: IDH1-mutant cells show profound vulnerability to HDACi treatment, alone and with PARPi and radiation, via HDR suppression, presenting IDH1/2 mutations as biomarkers for HDACi use in gliomas and other malignancies.
Collapse
Affiliation(s)
- Jonathan Dow
- Department of Therapeutic Radiology, Yale University School of Medicine. New Haven, Connecticut
- Department of Genetics, Yale University School of Medicine. New Haven, Connecticut
| | - Adam Krysztofiak
- Department of Therapeutic Radiology, Yale University School of Medicine. New Haven, Connecticut
| | - Yanfeng Liu
- Department of Therapeutic Radiology, Yale University School of Medicine. New Haven, Connecticut
- Department of Genetics, Yale University School of Medicine. New Haven, Connecticut
| | - Daniel A Colon-Rios
- Department of Therapeutic Radiology, Yale University School of Medicine. New Haven, Connecticut
| | - Faye A Rogers
- Department of Therapeutic Radiology, Yale University School of Medicine. New Haven, Connecticut
| | - Peter M Glazer
- Department of Therapeutic Radiology, Yale University School of Medicine. New Haven, Connecticut.
- Department of Genetics, Yale University School of Medicine. New Haven, Connecticut
| |
Collapse
|
186
|
Wen J, Rusch M, Brady SW, Shao Y, Edmonson MN, Shaw TI, Powers BB, Tian L, Easton J, Mullighan CG, Gruber T, Ellison D, Zhang J. The landscape of coding RNA editing events in pediatric cancer. BMC Cancer 2021; 21:1233. [PMID: 34789196 PMCID: PMC8597231 DOI: 10.1186/s12885-021-08956-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 11/02/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND RNA editing leads to post-transcriptional variation in protein sequences and has important biological implications. We sought to elucidate the landscape of RNA editing events across pediatric cancers. METHODS Using RNA-Seq data mapped by a pipeline designed to minimize mapping ambiguity, we investigated RNA editing in 711 pediatric cancers from the St. Jude/Washington University Pediatric Cancer Genome Project focusing on coding variants which can potentially increase protein sequence diversity. We combined de novo detection using paired tumor DNA-RNA data with analysis of known RNA editing sites. RESULTS We identified 722 unique RNA editing sites in coding regions across pediatric cancers, 70% of which were nonsynonymous recoding variants. Nearly all editing sites represented the canonical A-to-I (n = 706) or C-to-U sites (n = 14). RNA editing was enriched in brain tumors compared to other cancers, including editing of glutamate receptors and ion channels involved in neurotransmitter signaling. RNA editing profiles of each pediatric cancer subtype resembled those of the corresponding normal tissue profiled by the Genotype-Tissue Expression (GTEx) project. CONCLUSIONS In this first comprehensive analysis of RNA editing events in pediatric cancer, we found that the RNA editing profile of each cancer subtype is similar to its normal tissue of origin. Tumor-specific RNA editing events were not identified indicating that successful immunotherapeutic targeting of RNA-edited peptides in pediatric cancer should rely on increased antigen presentation on tumor cells compared to normal but not on tumor-specific RNA editing per se.
Collapse
Affiliation(s)
- Ji Wen
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Michael Rusch
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Samuel W Brady
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ying Shao
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Michael N Edmonson
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Timothy I Shaw
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Brent B Powers
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Liqing Tian
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Tanja Gruber
- Department of Pediatrics, Stanford University, Palo Alto, California, 94305, USA
| | - David Ellison
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
187
|
Fleischmann M, Schnetzke U, Hochhaus A, Scholl S. Management of Acute Myeloid Leukemia: Current Treatment Options and Future Perspectives. Cancers (Basel) 2021; 13:5722. [PMID: 34830877 PMCID: PMC8616498 DOI: 10.3390/cancers13225722] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 12/19/2022] Open
Abstract
Treatment of acute myeloid leukemia (AML) has improved in recent years and several new therapeutic options have been approved. Most of them include mutation-specific approaches (e.g., gilteritinib for AML patients with activating FLT3 mutations), or are restricted to such defined AML subgroups, such as AML-MRC (AML with myeloid-related changes) or therapy-related AML (CPX-351). With this review, we aim to present a comprehensive overview of current AML therapy according to the evolved spectrum of recently approved treatment strategies. We address several aspects of combined epigenetic therapy with the BCL-2 inhibitor venetoclax and provide insight into mechanisms of resistance towards venetoclax-based regimens, and how primary or secondary resistance might be circumvented. Furthermore, a detailed overview on the current status of AML immunotherapy, describing promising concepts, is provided. This review focuses on clinically important aspects of current and future concepts of AML treatment, but will also present the molecular background of distinct targeted therapies, to understand the development and challenges of clinical trials ongoing in AML patients.
Collapse
Affiliation(s)
| | | | | | - Sebastian Scholl
- Klinik für Innere Medizin II, Abteilung Hämatologie und Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07740 Jena, Germany; (M.F.); (U.S.); (A.H.)
| |
Collapse
|
188
|
USP29 coordinates MYC and HIF1α stabilization to promote tumor metabolism and progression. Oncogene 2021; 40:6417-6429. [PMID: 34601505 DOI: 10.1038/s41388-021-02031-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/07/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023]
Abstract
Tumor cells must rewire cellular metabolism to satisfy the demands of unbridled growth and proliferation. How these metabolic processes are integrated to fuel cancer cell growth remains largely unknown. Deciphering the regulatory mechanisms is vital to develop targeted strategies for tumor-selective therapies. We herein performed an unbiased and functional siRNA screen against 96 deubiquitinases, which play indispensable roles in cancer and are emerging as therapeutic targets, and identified USP29 as a top candidate essential for metabolic reprogramming that support biosynthesis and survival in tumor cells. Integrated metabolic flux analysis and molecular investigation reveal that USP29 directly deubiquitinates and stabilizes MYC and HIF1α, two master regulators of metabolic reprogramming, enabling adaptive response of tumor cells in both normoxia and hypoxia. Systemic knockout of Usp29 depleted MYC and HIF1α in MYC-driven neuroblastoma and B cell lymphoma, inhibited critical metabolic targets and significantly prolonged survival of tumor-bearing mice. Strikingly, mice homozygous null for the Usp29 gene are viable, fertile, and display no gross phenotypic abnormalities. Altogether, these results demonstrate that USP29 selectively coordinates MYC and HIF1α to integrate metabolic processes critical for cancer cell growth, and therapeutic targeting of USP29, a potentially targetable enzyme, could create a unique vulnerability given deregulation of MYC and HIF1α frequently occurs in human cancers.
Collapse
|
189
|
Récher C. The beginning of a new therapeutic era in acute myeloid leukemia. EJHAEM 2021; 2:823-833. [PMID: 35845213 PMCID: PMC9175720 DOI: 10.1002/jha2.252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 12/17/2022]
Abstract
In the field of AML, the early 2000s were shaped by the advent of novel molecular biology technologies including high-throughput sequencing that improved prognostic classification, response evaluation through the quantification of minimal residual disease, and the launch of research on targeted therapies. Our knowledge of leukemogenesis, AML genetic diversity, gene-gene interactions, clonal evolution, and treatment response assessment has also greatly improved. New classifications based on chromosomal abnormalities and gene mutations are now integrated on a routine basis. These considerable efforts contributed to the discovery and development of promising drugs which specifically target gene mutations, apoptotic pathways and cell surface antigens as well as reformulate classical cytotoxic agents. In less than 2 years, nine novels drugs have been approved for the treatment of AML patients, and many others are being intensively investigated, in particular immune therapies. There are now numerous clinical research opportunities offered to clinicians, thanks to these new treatment options. We are only at the start of a new era which should see major disruptions in the way we understand, treat, and monitor patients with AML.
Collapse
Affiliation(s)
- Christian Récher
- Service d'HématologieCentre Hospitalier Universitaire de ToulouseInstitut Universitaire du Cancer de Toulouse OncopoleUniversité Toulouse III Paul SabatierCentre de Recherches en Cancérologie de ToulouseToulouseFrance
| |
Collapse
|
190
|
Hanbazazh M, Harada S, Reddy V, Mackinnon AC, Harbi D, Morlote D. The Interpretation of Sequence Variants in Myeloid Neoplasms. Am J Clin Pathol 2021; 156:728-748. [PMID: 34155503 DOI: 10.1093/ajcp/aqab039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES To provide an overview of the challenges encountered during the interpretation of sequence variants detected by next-generation sequencing (NGS) in myeloid neoplasms, as well as the limitations of the technology with the goal of preventing the over- or undercalling of alterations that may have a significant effect on patient management. METHODS Review of the peer-reviewed literature on the interpretation, reporting, and technical challenges of NGS assays for myeloid neoplasms. RESULTS NGS has been integrated widely and rapidly into the standard evaluating of myeloid neoplasms. Review of the literature reveals that myeloid sequence variants are challenging to detect and interpret. Large insertions and guanine-cytosine-heavy areas prove technically challenging while frameshift and truncating alterations may be classified as variants of uncertain significance by tertiary analysis informatics pipelines due to their absence in the literature and databases. CONCLUSIONS The analysis and interpretation of NGS results in myeloid neoplasia are challenging due to the varied number of detectable gene alterations. Familiarity with the genomic landscape of myeloid malignancies and knowledge of the tools available for the interpretation of sequence variants are essential to facilitate translation into clinical and therapy decisions.
Collapse
Affiliation(s)
- Mehenaz Hanbazazh
- Department of Pathology, Division of Genomic Diagnostics and Bioinformatics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shuko Harada
- Department of Pathology, Division of Genomic Diagnostics and Bioinformatics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vishnu Reddy
- Department of Pathology, Division of Genomic Diagnostics and Bioinformatics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexander Craig Mackinnon
- Department of Pathology, Division of Genomic Diagnostics and Bioinformatics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Djamel Harbi
- Department of Pathology, Division of Genomic Diagnostics and Bioinformatics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Diana Morlote
- Department of Pathology, Division of Genomic Diagnostics and Bioinformatics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
191
|
Kalushkova A, Nylund P, Párraga AA, Lennartsson A, Jernberg-Wiklund H. One Omics Approach Does Not Rule Them All: The Metabolome and the Epigenome Join Forces in Haematological Malignancies. EPIGENOMES 2021; 5:epigenomes5040022. [PMID: 34968247 PMCID: PMC8715477 DOI: 10.3390/epigenomes5040022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/17/2021] [Accepted: 09/26/2021] [Indexed: 02/01/2023] Open
Abstract
Aberrant DNA methylation, dysregulation of chromatin-modifying enzymes, and microRNAs (miRNAs) play a crucial role in haematological malignancies. These epimutations, with an impact on chromatin accessibility and transcriptional output, are often associated with genomic instability and the emergence of drug resistance, disease progression, and poor survival. In order to exert their functions, epigenetic enzymes utilize cellular metabolites as co-factors and are highly dependent on their availability. By affecting the expression of metabolic enzymes, epigenetic modifiers may aid the generation of metabolite signatures that could be utilized as targets and biomarkers in cancer. This interdependency remains often neglected and poorly represented in studies, despite well-established methods to study the cellular metabolome. This review critically summarizes the current knowledge in the field to provide an integral picture of the interplay between epigenomic alterations and the cellular metabolome in haematological malignancies. Our recent findings defining a distinct metabolic signature upon response to enhancer of zeste homolog 2 (EZH2) inhibition in multiple myeloma (MM) highlight how a shift of preferred metabolic pathways may potentiate novel treatments. The suggested link between the epigenome and the metabolome in haematopoietic tumours holds promise for the use of metabolic signatures as possible biomarkers of response to treatment.
Collapse
Affiliation(s)
- Antonia Kalushkova
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
- Correspondence:
| | - Patrick Nylund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
| | - Alba Atienza Párraga
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
| | - Andreas Lennartsson
- Department of Biosciences and Nutrition, NEO, Karolinska Institutet, 14157 Huddinge, Sweden;
| | - Helena Jernberg-Wiklund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
| |
Collapse
|
192
|
Koklu A, Ohayon D, Wustoni S, Druet V, Saleh A, Inal S. Organic Bioelectronic Devices for Metabolite Sensing. Chem Rev 2021; 122:4581-4635. [PMID: 34610244 DOI: 10.1021/acs.chemrev.1c00395] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Electrochemical detection of metabolites is essential for early diagnosis and continuous monitoring of a variety of health conditions. This review focuses on organic electronic material-based metabolite sensors and highlights their potential to tackle critical challenges associated with metabolite detection. We provide an overview of the distinct classes of organic electronic materials and biorecognition units used in metabolite sensors, explain the different detection strategies developed to date, and identify the advantages and drawbacks of each technology. We then benchmark state-of-the-art organic electronic metabolite sensors by categorizing them based on their application area (in vitro, body-interfaced, in vivo, and cell-interfaced). Finally, we share our perspective on using organic bioelectronic materials for metabolite sensing and address the current challenges for the devices and progress to come.
Collapse
Affiliation(s)
- Anil Koklu
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE), Organic Bioelectronics Laboratory, Thuwal 23955-6900, Saudi Arabia
| | - David Ohayon
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE), Organic Bioelectronics Laboratory, Thuwal 23955-6900, Saudi Arabia
| | - Shofarul Wustoni
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE), Organic Bioelectronics Laboratory, Thuwal 23955-6900, Saudi Arabia
| | - Victor Druet
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE), Organic Bioelectronics Laboratory, Thuwal 23955-6900, Saudi Arabia
| | - Abdulelah Saleh
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE), Organic Bioelectronics Laboratory, Thuwal 23955-6900, Saudi Arabia
| | - Sahika Inal
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering (BESE), Organic Bioelectronics Laboratory, Thuwal 23955-6900, Saudi Arabia
| |
Collapse
|
193
|
Venetoclax and Azacitidine Compared to Induction Chemotherapy for Newly Diagnosed Patients with Acute Myeloid Leukemia. Blood Adv 2021; 5:5565-5573. [PMID: 34610123 PMCID: PMC8714726 DOI: 10.1182/bloodadvances.2021005538] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/17/2021] [Indexed: 11/27/2022] Open
Abstract
Patients with AML who received IC were compared with those who received ven to investigate predictors of outcomes. Presence of RUNX1 mutations is associated with better outcomes for ven/aza compared with IC.
Venetoclax (ven) plus azacitidine (aza) is the standard of care for patients with newly diagnosed acute myeloid leukemia (AML) who are not candidates for intensive chemotherapy (IC). Some patients who are IC candidates instead receive ven/aza. We retrospectively analyzed patients with newly diagnosed AML who received ven/aza (n = 143) or IC (n = 149) to compare outcomes, seek variables that could predict response to 1 therapy or the other, and ascertain whether treatment recommendations could be refined. The response rates were 76.9% for ven/aza and 70.5% for IC. The median overall survival (OS) was 884 days for IC compared with 483 days for ven/aza (P = .0020). A propensity-matched cohort was used to compare outcomes in the setting of equivalent baseline variables, and when matched for age, biological risk, and transplantation, the median OS was 705 days for IC compared with not reached for ven/aza (P = .0667). Variables that favored response to ven/aza over IC included older age, secondary AML, and RUNX1 mutations. AML M5 favored response to IC over ven/aza. In the propensity-matched cohort analyzing OS, older age, adverse risk, and RUNX1 mutations favored ven/aza over IC, whereas intermediate risk favored IC over ven/aza. In conclusion, patients receiving IC have improved OS compared with those receiving ven/aza. However, in a propensity-matched cohort of patients with equivalent baseline factors, there was a trend toward favorable OS for ven/aza. Specific variables, such as RUNX1 mutations, reported here for the first time, can be identified that favor ven/aza or IC, helping to guide treatment decisions for patients who may be eligible candidates for either therapy.
Collapse
|
194
|
Goldberg L, Negi V, Chung YJ, Onozawa M, Zhu YJ, Walker RL, Pierce R, Patel DP, Krausz KW, Gonzalez FJ, Goodell MA, Rodriguez BAT, Meltzer PS, Aplan PD. Mutant Idh2 Cooperates with a NUP98-HOXD13 Fusion to Induce Early Immature Thymocyte Precursor ALL. Cancer Res 2021; 81:5033-5046. [PMID: 34321240 PMCID: PMC8487989 DOI: 10.1158/0008-5472.can-21-1027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/09/2021] [Accepted: 07/27/2021] [Indexed: 11/16/2022]
Abstract
Mutations in the isocitrate dehydrogenase 1 (IDH1) and IDH2 genes are frequently observed in a wide variety of hematologic malignancies, including myeloid and T-cell leukemias. In this study, we generated Idh2R140Q transgenic mice to examine the role of the Idh2R140Q mutation in leukemia. No leukemia developed in Idh2R140Q transgenic mice, suggesting a need for additional genetic events for leukemia development. Because myeloid cells from NUP98-HOXD13 fusion (NHD13) transgenic mice frequently acquire somatic Idh mutations when they transform to acute myeloid leukemia, we generated Idh2R140Q/NHD13 double transgenic mice. Idh2R140Q/NHD13 transgenic mice developed an immature T-cell leukemia with an immunophenotype similar to double-negative 1 (DN1) or DN2 thymocytes. Idh2R140Q/NHD13 leukemic cells were enriched for an early thymic precursor transcriptional signature, and the gene expression profile for Idh2R140Q/NHD13 DN1/DN2 T-ALL closely matched that of human early/immature T-cell precursor (EITP) acute lymphoblastic leukemia (ALL). Moreover, recurrent mutations found in patients with EITP ALL, including KRAS, PTPN11, JAK3, SH2B3, and EZH2 were also found in Idh2R140Q/NHD13 DN1/DN2 T-ALL. In vitro treatment of Idh2R140Q/NHD13 thymocytes with enasidenib, a selective inhibitor of mutant IDH2, led to a marked decrease in leukemic cell proliferation. These findings demonstrate that Idh2R140Q/NHD13 mice can serve as a useful in vivo model for the study of early/immature thymocyte precursor acute lymphoblastic leukemia development and therapy. SIGNIFICANCE: T-cell leukemia induced in Idh2R140Q/NUP98-HOXD13 mice is immunophenotypically, transcriptionally, and genetically similar to human EITP ALL, providing a model for studying disease development and treatment.
Collapse
Affiliation(s)
- Liat Goldberg
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Vijay Negi
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Yang Jo Chung
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Masahiro Onozawa
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Yuelin J Zhu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Robert L Walker
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Rachel Pierce
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Daxesh P Patel
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Margaret A Goodell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Benjamin A T Rodriguez
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Valo Health, Boston, Massachusetts
| | - Paul S Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Peter D Aplan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
195
|
Hindley A, Catherwood MA, McMullin MF, Mills KI. Significance of NPM1 Gene Mutations in AML. Int J Mol Sci 2021; 22:ijms221810040. [PMID: 34576201 PMCID: PMC8467861 DOI: 10.3390/ijms221810040] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 12/16/2022] Open
Abstract
The aim of this literature review is to examine the significance of the nucleophosmin 1 (NPM1) gene in acute myeloid leukaemia (AML). This will include analysis of the structure and normal cellular function of NPM1, the type of mutations commonly witnessed in NPM1, and the mechanism by which this influences the development and progression of AML. The importance of NPM1 mutation on prognosis and the treatment options available to patients will also be reviewed along with current guidelines recommending the rapid return of NPM1 mutational screening results and the importance of employing a suitable laboratory assay to achieve this. Finally, future developments in the field including research into new therapies targeting NPM1 mutated AML are considered.
Collapse
Affiliation(s)
- Andrew Hindley
- Clinical Haematology, Belfast City Hospital, Belfast BT9 7AB, UK;
- Correspondence:
| | | | - Mary Frances McMullin
- Centre for Medical Education, Queen’s University Belfast, Belfast BT7 1NN, UK;
- Northern Ireland and Belfast Health and Social Care Trust, Belfast BT9 7AB, UK
| | - Ken I. Mills
- Patrick G Johnston Center for Cancer Research, Queens University Belfast, Belfast BT9 7AE, UK;
| |
Collapse
|
196
|
Sasca D, Guezguez B, Kühn MWM. Next generation epigenetic modulators to target myeloid neoplasms. Curr Opin Hematol 2021; 28:356-363. [PMID: 34267079 DOI: 10.1097/moh.0000000000000673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW Comprehensive sequencing studies aimed at determining the genetic landscape of myeloid neoplasms have identified epigenetic regulators to be among the most commonly mutated genes. Detailed studies have also revealed a number of epigenetic vulnerabilities. The purpose of this review is to outline these vulnerabilities and to discuss the new generation of drugs that exploit them. RECENT FINDINGS In addition to deoxyribonucleic acid-methylation, novel epigenetic dependencies have recently been discovered in various myeloid neoplasms and many of them can be targeted pharmacologically. These include not only chromatin writers, readers, and erasers but also chromatin movers that shift nucleosomes to allow access for transcription. Inhibitors of protein-protein interactions represent a novel promising class of drugs that allow disassembly of oncogenic multiprotein complexes. SUMMARY An improved understanding of disease-specific epigenetic vulnerabilities has led to the development of second-generation mechanism-based epigenetic drugs against myeloid neoplasms. Many of these drugs have been introduced into clinical trials and synergistic drug combination regimens have been shown to enhance efficacy and potentially prevent drug resistance.
Collapse
Affiliation(s)
- Daniel Sasca
- Department of Hematology, Oncology, and Pulmonary Medicine, University Medical Center, Johannes Gutenberg-University Mainz, Mainz
| | - Borhane Guezguez
- Department of Hematology, Oncology, and Pulmonary Medicine, University Medical Center, Johannes Gutenberg-University Mainz, Mainz
- German Cancer Research Center (DKFZ), Heidelberg
- German Cancer Consortium (DKTK), Mainz, Germany
| | - Michael W M Kühn
- Department of Hematology, Oncology, and Pulmonary Medicine, University Medical Center, Johannes Gutenberg-University Mainz, Mainz
| |
Collapse
|
197
|
Comprehensive characterization of the single nucleotide polymorphisms located in the isocitrate dehydrogenase isoform 1 and 2 genes using in silico approach. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
198
|
Dragani M, de Botton S. SOHO State of the Art Updates and Next Questions: IDH Inhibition. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2021; 21:567-572. [PMID: 34193376 DOI: 10.1016/j.clml.2021.05.004] [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: 03/17/2021] [Revised: 04/23/2021] [Accepted: 05/03/2021] [Indexed: 06/13/2023]
Abstract
There has been extraordinary progress in the field of targeted therapy for myeloid malignancies in the last few years, especially due to the approval of various agents that can be used as monotherapy or in combination as first-line treatment or when facing a refractory or relapsed disease. Many successful trials have been conducted recently, and a consistent body of work about the efficacy of novel molecules is now available. In this review, we sought to explain how enasidenib and ivosidenib have changed the face of myeloid neoplasm treatment through isocitrate dehydrogenase inhibition and to summarize the trials results that have led to the current commercial indications for the two molecules.
Collapse
Affiliation(s)
- Matteo Dragani
- Hematology Department, Gustave Roussy Cancer Centre, Villejuif, France
| | | |
Collapse
|
199
|
Aung MMK, Mills ML, Bittencourt‐Silvestre J, Keeshan K. Insights into the molecular profiles of adult and paediatric acute myeloid leukaemia. Mol Oncol 2021; 15:2253-2272. [PMID: 33421304 PMCID: PMC8410545 DOI: 10.1002/1878-0261.12899] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/18/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
Acute myeloid leukaemia (AML) is a clinically and molecularly heterogeneous disease characterised by uncontrolled proliferation, block in differentiation and acquired self-renewal of hematopoietic stem and myeloid progenitor cells. This results in the clonal expansion of myeloid blasts within the bone marrow and peripheral blood. The incidence of AML increases with age, and in childhood, AML accounts for 20% of all leukaemias. Whilst there are many clinical and biological similarities between paediatric and adult AML with continuum across the age range, many characteristics of AML are associated with age of disease onset. These include chromosomal aberrations, gene mutations and differentiation lineage. Following chemotherapy, AML cells that survive and result in disease relapse exist in an altered chemoresistant state. Molecular profiling currently represents a powerful avenue of experimentation to study AML cells from adults and children pre- and postchemotherapy as a means of identifying prognostic biomarkers and targetable molecular vulnerabilities that may be age-specific. This review highlights recent advances in our knowledge of the molecular profiles with a focus on transcriptomes and metabolomes, leukaemia stem cells and chemoresistant cells in adult and paediatric AML and focus on areas that hold promise for future therapies.
Collapse
Affiliation(s)
- Myint Myat Khine Aung
- Paul O’Gorman Leukaemia Research CentreInstitute of Cancer SciencesUniversity of GlasgowUK
| | - Megan L. Mills
- Paul O’Gorman Leukaemia Research CentreInstitute of Cancer SciencesUniversity of GlasgowUK
| | | | - Karen Keeshan
- Paul O’Gorman Leukaemia Research CentreInstitute of Cancer SciencesUniversity of GlasgowUK
| |
Collapse
|
200
|
Abstract
ABSTRACT High-grade gliomas are among the deadliest of all cancers despite standard treatments, and new therapeutic strategies are needed to improve patient outcome. Targeting the altered metabolic state of tumors with traditional chemotherapeutic agents has a history of success, and our increased understanding of cellular metabolism in the past 2 decades has reinvigorated the concept of novel metabolic therapies in brain tumors. Here we highlight metabolic alterations in advanced gliomas and their translation into clinical trials using both novel agents and already established drugs repurposed for cancer treatment in an effort to improve outcome for these deadly diseases.
Collapse
Affiliation(s)
- Andrew J. Scott
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
| | - Costas A. Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109; Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI 48109; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
| | - Daniel R. Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109; University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|